51
|
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
|
52
|
Joy MT, Ben Assayag E, Shabashov-Stone D, Liraz-Zaltsman S, Mazzitelli J, Arenas M, Abduljawad N, Kliper E, Korczyn AD, Thareja NS, Kesner EL, Zhou M, Huang S, Silva TK, Katz N, Bornstein NM, Silva AJ, Shohami E, Carmichael ST. CCR5 Is a Therapeutic Target for Recovery after Stroke and Traumatic Brain Injury. Cell 2020; 176:1143-1157.e13. [PMID: 30794775 DOI: 10.1016/j.cell.2019.01.044] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 10/05/2018] [Accepted: 01/24/2019] [Indexed: 12/19/2022]
Abstract
We tested a newly described molecular memory system, CCR5 signaling, for its role in recovery after stroke and traumatic brain injury (TBI). CCR5 is uniquely expressed in cortical neurons after stroke. Post-stroke neuronal knockdown of CCR5 in pre-motor cortex leads to early recovery of motor control. Recovery is associated with preservation of dendritic spines, new patterns of cortical projections to contralateral pre-motor cortex, and upregulation of CREB and DLK signaling. Administration of a clinically utilized FDA-approved CCR5 antagonist, devised for HIV treatment, produces similar effects on motor recovery post stroke and cognitive decline post TBI. Finally, in a large clinical cohort of stroke patients, carriers for a naturally occurring loss-of-function mutation in CCR5 (CCR5-Δ32) exhibited greater recovery of neurological impairments and cognitive function. In summary, CCR5 is a translational target for neural repair in stroke and TBI and the first reported gene associated with enhanced recovery in human stroke.
Collapse
Affiliation(s)
- Mary T Joy
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Einor Ben Assayag
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dalia Shabashov-Stone
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sigal Liraz-Zaltsman
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel; The Joseph Sagol Neuroscience Center, Sheba Medical Center, Israel; Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono, Israel
| | - Jose Mazzitelli
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Marcela Arenas
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Nora Abduljawad
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Efrat Kliper
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Amos D Korczyn
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nikita S Thareja
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Efrat L Kesner
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miou Zhou
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Shan Huang
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Tawnie K Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Noomi Katz
- Institute for Health and Medical Professions, Ono Academic College, Kiryat Ono, Israel
| | - Natan M Bornstein
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory and Brain Research Institute, UCLA, Los Angeles, CA 90095, USA
| | - Esther Shohami
- Department of Pharmacology, The Institute for Drug Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
53
|
Apolipoprotein E Deficiency Aggravates Neuronal Injury by Enhancing Neuroinflammation via the JNK/c-Jun Pathway in the Early Phase of Experimental Subarachnoid Hemorrhage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3832648. [PMID: 31949876 PMCID: PMC6944964 DOI: 10.1155/2019/3832648] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/06/2019] [Accepted: 09/24/2019] [Indexed: 12/20/2022]
Abstract
Neuronal injury is the primary cause of poor outcome after subarachnoid hemorrhage (SAH). The apolipoprotein E (APOE) gene has been suggested to be involved in the prognosis of SAH patients. However, the role of APOE in neuronal injury after SAH has not been well studied. In this study, SAH was induced in APOE-knockout (APOE−/−) and wild-type (WT) mice to investigate the impact of APOE deficiency on neuronal injury in the early phase of SAH. The experiments of this study were performed in murine SAH models in vivo and primary cultured microglia and neurons in vitro. The SAH model was induced by endovascular perforation in APOE−/− and APOE WT mice. The mortality rate, weight loss, and neurological deficits were recorded within 72 h after SAH. The neuronal injury was assessed by detecting the neuronal apoptosis and axonal injury. The activation of microglia was assessed by immunofluorescent staining of Iba-1, and clodronate liposomes were used for inhibiting microglial activation. The expression of JNK/c-Jun was evaluated by immunofluorescent staining or western blotting. The expression of TNF-α, IL-1β, and IL-6 was evaluated by ELISA. Primary cultured microglia were treated with hemoglobin (Hb) in vitro for simulating the pathological process of SAH. SP600125, a JNK inhibitor, was used for evaluating the role of JNK in neuroinflammation. Nitrite production was detected for microglial activation, and flow cytometry was performed to detect apoptosis in vitro. The results suggested that SAH induced early neuronal injury and neurological deficits in mice. APOE deficiency resulted in more severe neurological deficits after SAH in mice. The neurological deficits were associated with exacerbation of neuronal injury, including neuronal apoptosis and axonal injury. Moreover, APOE deficiency enhanced microglial activation and related inflammatory injury on neurons. Inhibition of microglia attenuated neuronal injury in mice, whereas inhibition of JNK inhibited microglia-mediated inflammatory response in vitro. Taken together, JNK/c-Jun was involved in the enhancement of microglia-mediated inflammatory injury in APOE−/− mice. APOE deficiency aggravates neuronal injury which may account for the poor neurological outcomes of APOE−/− mice. The possible protective role of APOE against EBI via the modulation of inflammatory response indicates its potential treatment for SAH.
Collapse
|
54
|
Systemic inflammation in traumatic spinal cord injury. Exp Neurol 2019; 325:113143. [PMID: 31843491 DOI: 10.1016/j.expneurol.2019.113143] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 01/08/2023]
|
55
|
Koganti L, Liu J, DeMajewski A, Agostini MA, Wong TW, Faber DS, Zottoli SJ. Invasion of microglia/macrophages and granulocytes into the Mauthner axon myelin sheath following spinal cord injury of the adult goldfish, Carassius auratus. J Morphol 2019; 281:135-152. [PMID: 31774588 DOI: 10.1002/jmor.21086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 10/23/2019] [Accepted: 11/10/2019] [Indexed: 12/21/2022]
Abstract
Rapid activation of resident glia occurs after spinal cord injury. Somewhat later, innate and adaptive immune responses occur with the invasion of peripheral immune cells into the wound site. The activation of resident and peripheral immune cells has been postulated to play harmful as well as beneficial roles in the regenerative process. Mauthner cells, large identifiable neurons located in the hindbrain of most fish and amphibians, provided the opportunity to study the morphological relationship between reactive cells and Mauthner axons (M-axons) severed by spinal cord crush or by selective axotomy. After crossing in the hindbrain, the M-axons of adult goldfish, Carassius auratus, extend the length of the spinal cord. Following injury, the M-axon undergoes retrograde degeneration within its myelin sheath creating an axon-free zone (proximal dieback zone). Reactive cells invade the wound site, enter the axon-free dieback zone and are observed in the vicinity of the retracted M-axon tip as early as 3 hr postinjury. Transmission electron microscopy allowed the detection of microglia/macrophages and granulocytes, some of which appear to be neutrophil-like, at each of these locations. We believe that this is the first report of the invasion of such cells within the myelin sheath of an identifiable axon in the vertebrate central nervous system (CNS). We speculate that microglia/macrophages and granulocytes that are attracted within a few hours to the damaged M-axon are part of an inflammatory response that allows phagocytosis of debris and plays a role in the regenerative process. Our results provide the baseline from which to utilize immunohistochemical and genetic approaches to elucidate the role of non-neuronal cells in the regenerative process of a single axon in the vertebrate CNS.
Collapse
Affiliation(s)
- Lahari Koganti
- Department of Biology, Williams College, Williamstown, Massachusetts
| | - Jun Liu
- Department of Biology, Williams College, Williamstown, Massachusetts
| | - Andrea DeMajewski
- Department of Biology, Williams College, Williamstown, Massachusetts
| | - Mark A Agostini
- Department of Biology, Williams College, Williamstown, Massachusetts
| | - Tina W Wong
- Department of Biology, Williams College, Williamstown, Massachusetts
| | - Donald S Faber
- Albert Einstein College of Medicine, Rose F. Kennedy Center, Bronx, New York
| | - Steven J Zottoli
- Department of Biology, Williams College, Williamstown, Massachusetts.,Marine Biological Laboratory, Woods Hole, Massachusetts
| |
Collapse
|
56
|
Enos N, Takenaka H, Scott S, Salfity HVN, Kirk M, Egar MW, Sarria DA, Slayback-Barry D, Belecky-Adams T, Chernoff EAG. Meningeal Foam Cells and Ependymal Cells in Axolotl Spinal Cord Regeneration. Front Immunol 2019; 10:2558. [PMID: 31736973 PMCID: PMC6838144 DOI: 10.3389/fimmu.2019.02558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/15/2019] [Indexed: 12/01/2022] Open
Abstract
A previously unreported population of foam cells (foamy macrophages) accumulates in the invasive fibrotic meninges during gap regeneration of transected adult Axolotl spinal cord (salamander Ambystoma mexicanum) and may act beneficially. Multinucleated giant cells (MNGCs) also occurred in the fibrotic meninges. Actin-label localization and transmission electron microscopy showed characteristic foam cell and MNGC podosome and ruffled border-containing sealing ring structures involved in substratum attachment, with characteristic intermediate filament accumulations surrounding nuclei. These cells co-localized with regenerating cord ependymal cell (ependymoglial) outgrowth. Phase contrast-bright droplets labeled with Oil Red O, DiI, and DyRect polar lipid live cell label showed accumulated foamy macrophages to be heavily lipid-laden, while reactive ependymoglia contained smaller lipid droplets. Both cell types contained both neutral and polar lipids in lipid droplets. Foamy macrophages and ependymoglia expressed the lipid scavenger receptor CD36 (fatty acid translocase) and the co-transporter toll-like receptor-4 (TLR4). Competitive inhibitor treatment using the modified fatty acid Sulfo-N-succinimidyl Oleate verified the role of the lipid scavenger receptor CD36 in lipid uptake studies in vitro. Fluoromyelin staining showed both cell types took up myelin fragments in situ during the regeneration process. Foam cells took up DiI-Ox-LDL and DiI-myelin fragments in vitro while ependymoglia took up only DiI-myelin in vitro. Both cell types expressed the cysteine proteinase cathepsin K, with foam cells sequestering cathepsin K within the sealing ring adjacent to the culture substratum. The two cell types act as sinks for Ox-LDL and myelin fragments within the lesion site, with foamy macrophages showing more Ox-LDL uptake activity. Cathepsin K activity and cellular localization suggested that foamy macrophages digest ECM within reactive meninges, while ependymal cells act from within the spinal cord tissue during outgrowth into the lesion site, acting in complementary fashion. Small MNGCs also expressed lipid transporters and showed cathepsin K activity. Comparison of 3H-glucosamine uptake in ependymal cells and foam cells showed that only ependymal cells produce glycosaminoglycan and proteoglycan-containing ECM, while the cathepsin studies showed both cell types remove ECM. Interaction of foam cells and ependymoglia in vitro supported the dispersion of ependymal outgrowth associated with tissue reconstruction in Axolotl spinal cord regeneration.
Collapse
Affiliation(s)
- Nathaniel Enos
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Hidehito Takenaka
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Sarah Scott
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Hai V N Salfity
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Maia Kirk
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Margaret W Egar
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Deborah A Sarria
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Denise Slayback-Barry
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Teri Belecky-Adams
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Ellen A G Chernoff
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| |
Collapse
|
57
|
Bradbury EJ, Burnside ER. Moving beyond the glial scar for spinal cord repair. Nat Commun 2019; 10:3879. [PMID: 31462640 PMCID: PMC6713740 DOI: 10.1038/s41467-019-11707-7] [Citation(s) in RCA: 396] [Impact Index Per Article: 79.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 07/25/2019] [Indexed: 02/08/2023] Open
Abstract
Traumatic spinal cord injury results in severe and irreversible loss of function. The injury triggers a complex cascade of inflammatory and pathological processes, culminating in formation of a scar. While traditionally referred to as a glial scar, the spinal injury scar in fact comprises multiple cellular and extracellular components. This multidimensional nature should be considered when aiming to understand the role of scarring in limiting tissue repair and recovery. In this Review we discuss recent advances in understanding the composition and phenotypic characteristics of the spinal injury scar, the oversimplification of defining the scar in binary terms as good or bad, and the development of therapeutic approaches to target scar components to enable improved functional outcome after spinal cord injury.
Collapse
Affiliation(s)
- Elizabeth J Bradbury
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK.
| | - Emily R Burnside
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| |
Collapse
|
58
|
Sommer D, Corstjens I, Sanchez S, Dooley D, Lemmens S, Van Broeckhoven J, Bogie J, Vanmierlo T, Vidal PM, Rose-John S, Gou-Fabregas M, Hendrix S. ADAM17-deficiency on microglia but not on macrophages promotes phagocytosis and functional recovery after spinal cord injury. Brain Behav Immun 2019; 80:129-145. [PMID: 30851378 DOI: 10.1016/j.bbi.2019.02.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/12/2019] [Accepted: 02/27/2019] [Indexed: 12/26/2022] Open
Abstract
A disintegrin and metalloproteinase 17 (ADAM17) is the major sheddase involved in the cleavage of a plethora of cytokines, cytokine receptors and growth factors, thereby playing a substantial role in inflammatory and regenerative processes after central nervous system trauma. By making use of a hypomorphic ADAM17 knockin mouse model as well as pharmacological ADAM10/ADAM17 inhibitors, we showed that ADAM17-deficiency or inhibition significantly increases clearance of apoptotic cells, promotes axon growth and improves functional recovery after spinal cord injury (SCI) in mice. Microglia-specific ADAM17-knockout (ADAM17flox+/+-Cx3Cr1 Cre+/-) mice also showed improved functional recovery similar to hypomorphic ADAM17 mice. In contrast, endothelial-specific (ADAM17flox+/+-Cdh5Pacs Cre+/-) and macrophage-specific (ADAM17flox+/+-LysM Cre+/-) ADAM17-knockout mice or bone marrow chimera with transplanted ADAM17-deficient macrophages, displayed no functional improvement compared to wild type mice. These data indicate that ADAM17 expression on microglia cells (and not on macrophages or endothelial cells) plays a detrimental role in inflammation and functional recovery after SCI.
Collapse
Affiliation(s)
- Daniela Sommer
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Inge Corstjens
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Selien Sanchez
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Dearbhaile Dooley
- Health Science Centre, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Stefanie Lemmens
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | | | - Jeroen Bogie
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Tim Vanmierlo
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; Division of Translational Neuroscience, MHeNs, Maastricht University, 6229ER Maastricht, the Netherlands
| | - Pia M Vidal
- Laboratory of Neuroimmunology, Fundación Ciencia & Vida, 7780272 Santiago, Chile
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts University Kiel, 24098 Kiel, Germany
| | | | - Sven Hendrix
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium.
| |
Collapse
|
59
|
Toll-like receptor 9 antagonism modulates astrocyte function and preserves proximal axons following spinal cord injury. Brain Behav Immun 2019; 80:328-343. [PMID: 30953770 DOI: 10.1016/j.bbi.2019.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 03/14/2019] [Accepted: 04/02/2019] [Indexed: 02/07/2023] Open
Abstract
Increasing evidence indicates that innate immune receptors play important, yet controversial, roles in traumatic central nervous system (CNS) injury. Despite many advances, the contributions of toll-like receptors (TLRs) to spinal cord injury (SCI) remain inadequately defined. We previously reported that a toll-like receptor 9 (TLR9) antagonist, oligodeoxynucleotide 2088 (ODN 2088), administered intrathecally, improves the functional and histopathological outcomes of SCI. However, the molecular and cellular changes that occur at the injury epicenter following ODN 2088 treatment are not completely understood. Following traumatic SCI, a glial scar, consisting primarily of proliferating reactive astrocytes, forms at the injury epicenter and assumes both beneficial and detrimental roles. Increased production of chondroitin sulfate proteoglycans (CSPGs) by reactive astrocytes inhibits the regeneration of injured axons. Astrocytes express TLR9, which can be activated by endogenous ligands released by damaged cells. It is not yet known how TLR9 antagonism modifies astrocyte function at the glial scar and how this affects axonal preservation or re-growth following SCI. The present studies were undertaken to address these issues. We report that in female mice sustaining a severe mid-thoracic (T8) contusion injury, the number of proliferating astrocytes in regions rostral and caudal to the lesion border increased significantly by 30- and 24-fold, respectively, compared to uninjured controls. Intrathecal ODN 2088 treatment significantly reduced the number of proliferating astrocytes by 60% in both regions. This effect appeared to be, at least partly, mediated through the direct actions of ODN 2088 on astrocytes, since the antagonist decreased proliferation in pure SC astrocyte cultures by preventing the activation of the Erk/MAPK signaling pathway. In addition, CSPG immunoreactivity at the lesion border was more pronounced in vehicle-treated injured mice compared to uninjured controls and was significantly reduced following administration of ODN 2088 to injured mice. Moreover, ODN 2088 significantly decreased astrocyte migration in an in vitro scratch-wound assay. Anterograde tracing and quantification of corticospinal tract (CST) axons in injured mice, indicated that ODN 2088 preserves proximal axons. Taken together, these findings suggest that ODN 2088 modifies the glial scar and creates a milieu that fosters axonal protection at the injury site.
Collapse
|
60
|
Katoh H, Yokota K, Fehlings MG. Regeneration of Spinal Cord Connectivity Through Stem Cell Transplantation and Biomaterial Scaffolds. Front Cell Neurosci 2019; 13:248. [PMID: 31244609 PMCID: PMC6563678 DOI: 10.3389/fncel.2019.00248] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/17/2019] [Indexed: 12/20/2022] Open
Abstract
Significant progress has been made in the treatment of spinal cord injury (SCI). Advances in post-trauma management and intensive rehabilitation have significantly improved the prognosis of SCI and converted what was once an “ailment not to be treated” into a survivable injury, but the cold hard fact is that we still do not have a validated method to improve the paralysis of SCI. The irreversible functional impairment of the injured spinal cord is caused by the disruption of neuronal transduction across the injury lesion, which is brought about by demyelination, axonal degeneration, and loss of synapses. Furthermore, refractory substrates generated in the injured spinal cord inhibit spontaneous recovery. The discovery of the regenerative capability of central nervous system neurons in the proper environment and the verification of neural stem cells in the spinal cord once incited hope that a cure for SCI was on the horizon. That hope was gradually replaced with mounting frustration when neuroprotective drugs, cell transplantation, and strategies to enhance remyelination, axonal regeneration, and neuronal plasticity demonstrated significant improvement in animal models of SCI but did not translate into a cure in human patients. However, recent advances in SCI research have greatly increased our understanding of the fundamental processes underlying SCI and fostered increasing optimism that these multiple treatment strategies are finally coming together to bring about a new era in which we will be able to propose encouraging therapies that will lead to appreciable improvements in SCI patients. In this review, we outline the pathophysiology of SCI that makes the spinal cord refractory to regeneration and discuss the research that has been done with cell replacement and biomaterial implantation strategies, both by itself and as a combined treatment. We will focus on the capacity of these strategies to facilitate the regeneration of neural connectivity necessary to achieve meaningful functional recovery after SCI.
Collapse
Affiliation(s)
- Hiroyuki Katoh
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Department of Orthopaedic Surgery - Surgical Sciences, School of Medicine, Tokai University, Tokyo, Japan
| | - Kazuya Yokota
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Division of Neurosurgery, University of Toronto, Toronto, ON, Canada.,Spine Program, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
61
|
Courtine G, Sofroniew MV. Spinal cord repair: advances in biology and technology. Nat Med 2019; 25:898-908. [PMID: 31160817 DOI: 10.1038/s41591-019-0475-6] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Individuals with spinal cord injury (SCI) can face decades with permanent disabilities. Advances in clinical management have decreased morbidity and improved outcomes, but no randomized clinical trial has demonstrated the efficacy of a repair strategy for improving recovery from SCI. Here, we summarize recent advances in biological and engineering strategies to augment neuroplasticity and/or functional recovery in animal models of SCI that are pushing toward clinical translation.
Collapse
Affiliation(s)
- Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland. .,Department of Neurosurgery, University Hospital Lausanne (CHUV), Lausanne, Switzerland.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
62
|
Schwab JM, Kleinschnitz C. Overcoming trivialization: The neuroimmune response after acute central nervous system injury. J Neuroimmunol 2019; 330:28-30. [PMID: 30772755 DOI: 10.1016/j.jneuroim.2019.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 10/27/2022]
Affiliation(s)
- Jan M Schwab
- Belford Center for Spinal Cord Injury, The Ohio State University, Wexner Medical Center, Columbus, OH, United States; Spinal Cord Injury Division (Paraplegiology), Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH, United States; Discovery Theme Initiative of the Ohio State University (Chronic Brain Injury), United States.
| | | |
Collapse
|
63
|
Stoica SI, Tănase I, Ciobanu V, Onose G. Initial researches on neuro-functional status and evolution in chronic ethanol consumers with recent traumatic spinal cord injury. J Med Life 2019; 12:97-112. [PMID: 31406510 PMCID: PMC6685305 DOI: 10.25122/jml-2019-0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/21/2019] [Indexed: 12/01/2022] Open
Abstract
We found differences related to the neuro-functional deficiency and clinical progress, among non-consumers and chronic consumers of ethanol, with recent traumatic spinal cord injury (SCI). We present a synthesis of related data on lesion mechanisms in post-traumatic myelogenous disorders, namely some of the alcohols and their actions on the nervous system, with details on the influences exerted, in such afflictions, by the chronic consumption of ethanol. The subject is not frequently approached - according to a literature review with systematic elements, which we have done before - thus constituting a niche that deserves to be further explored. The applicative component of the article highlights statistical data resulted from a retrospective study regarding the specialized casuistry from the Neuromuscular Recovery Clinic of the "Bagdasar Arseni" Emergency Clinical Hospital, following the comparative analysis of two groups of patients with recent SCI: non-consumers - the control group (n=780) - and chronic ethanol consumers - the study group (n=225) - with the addition of a prospective pilot component. Data processing has been achieved with SPSS 24. The American Spinal Injury Association Impairment Scale (AIS) mean motor scores differ significantly (tests: Mann-Whitney and t) between the control and study group in favor of the second, both at admission (p<0.001) and at discharge (p<0.001). AIS mean sensitive scores differ between the two lots, and also in favor of the study, but statistically significant only at discharge (p=0.048); the difference at admission is not significant (p=0.51) - possibly because of alcoholic-nutritional polyneuropathy. These findings, with numerous related details, later presented in the text, are surprising, which requires further studies and attempts of understanding.
Collapse
Affiliation(s)
- Simona Isabelle Stoica
- “Carol Davila” University of Medicine and Pharmacy (UMPCD), Bucharest, Romania
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), Bucharest, Romania
| | - Ioana Tănase
- “Carol Davila” University of Medicine and Pharmacy (UMPCD), Bucharest, Romania
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), Bucharest, Romania
| | - Vlad Ciobanu
- Politehnica University of Bucharest (PUB), Bucharest, Romania
| | - Gelu Onose
- “Carol Davila” University of Medicine and Pharmacy (UMPCD), Bucharest, Romania
- Teaching Emergency Hospital “Bagdasar-Arseni” (TEHBA), Bucharest, Romania
| |
Collapse
|
64
|
Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front Neurol 2019; 10:282. [PMID: 30967837 PMCID: PMC6439316 DOI: 10.3389/fneur.2019.00282] [Citation(s) in RCA: 635] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a life changing neurological condition with substantial socioeconomic implications for patients and their care-givers. Recent advances in medical management of SCI has significantly improved diagnosis, stabilization, survival rate and well-being of SCI patients. However, there has been small progress on treatment options for improving the neurological outcomes of SCI patients. This incremental success mainly reflects the complexity of SCI pathophysiology and the diverse biochemical and physiological changes that occur in the injured spinal cord. Therefore, in the past few decades, considerable efforts have been made by SCI researchers to elucidate the pathophysiology of SCI and unravel the underlying cellular and molecular mechanisms of tissue degeneration and repair in the injured spinal cord. To this end, a number of preclinical animal and injury models have been developed to more closely recapitulate the primary and secondary injury processes of SCI. In this review, we will provide a comprehensive overview of the recent advances in our understanding of the pathophysiology of SCI. We will also discuss the neurological outcomes of human SCI and the available experimental model systems that have been employed to identify SCI mechanisms and develop therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Scott Matthew Dyck
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
65
|
Zupanc GK. Stem‐Cell‐Driven Growth and Regrowth of the Adult Spinal Cord in Teleost Fish. Dev Neurobiol 2019; 79:406-423. [DOI: 10.1002/dneu.22672] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/12/2019] [Accepted: 02/25/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Günther K.H. Zupanc
- Laboratory of Neurobiology, Department of Biology Northeastern University Boston Massachusetts
| |
Collapse
|
66
|
Hassannejad Z, Yousefifard M, Azizi Y, Zadegan SA, Sajadi K, Sharif-Alhoseini M, Shakouri-Motlagh A, Mokhatab M, Rezvan M, Shokraneh F, Hosseini M, Vaccaro AR, Harrop JS, Rahimi-Movaghar V. Axonal degeneration and demyelination following traumatic spinal cord injury: A systematic review and meta-analysis. J Chem Neuroanat 2019; 97:9-22. [PMID: 30726717 DOI: 10.1016/j.jchemneu.2019.01.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/22/2018] [Accepted: 01/22/2019] [Indexed: 12/17/2022]
Abstract
The pathophysiology of spinal cord injury (SCI) related processes of axonal degeneration and demyelination are poorly understood. The present systematic review and meta-analysis were performed such to establish quantitative results of animal studies regarding the role of injury severity, SCI models and level of injury on the pathophysiology of axon and myelin sheath degeneration. 39 related articles were included in the analysis. The compiled data showed that the total number of axons, number of myelinated axons, myelin sheath thickness, axonal conduction velocity, and internode length steadily decreased as time elapsed from the injury (Pfor trend<0.0001). The rate of axonal retrograde degeneration was affected by SCI model and severity of the injury. Axonal degeneration was higher in injuries of the thoracic region. The SCI model and the site of the injury also affected axonal retrograde degeneration. The number of myelinated axons in the caudal region of the injury was significantly higher than the lesion site and the rostral region. The findings of the present meta-analysis show that the pathophysiology of axons and myelin sheath differ in various phases of SCI and are affected by multiple factors related to the injury.
Collapse
Affiliation(s)
- Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Yaser Azizi
- Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Abdollah Zadegan
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiavash Sajadi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Sharif-Alhoseini
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Shakouri-Motlagh
- Department of Chemical and Biomolecular Engineering, University of Melbourne, Victoria 3010, Australia
| | - Mona Mokhatab
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Motahareh Rezvan
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Shokraneh
- Cochrane Schizophrenia Group, Institute of Mental Health, University of Nottingham, Nottingham, UK
| | - Mostafa Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Alexander R Vaccaro
- Department of Orthopedics and Neurosurgery, Rothman Institute, Thomas Jefferson University Philadelphia, USA
| | - James S Harrop
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Brain and Spinal Injuries Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
67
|
Zeng Y, Wang N, Guo T, Zheng Q, Wang S, Wu S, Li X, Wu J, Chen Z, Xu H, Wang X, Lin B. Snx27 Deletion Promotes Recovery From Spinal Cord Injury by Neuroprotection and Reduces Macrophage/Microglia Proliferation. Front Neurol 2018; 9:1059. [PMID: 30619032 PMCID: PMC6300502 DOI: 10.3389/fneur.2018.01059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 11/21/2018] [Indexed: 12/16/2022] Open
Abstract
Sorting nexin 27 (SNX27) is an endosome-associated cargo adaptor that is involved in various pathologies and development of neurological diseases. However, the role of SNX27 in spinal cord injury (SCI) remains unclear. In this study, we found that SNX27 was up-regulated in injured mice spinal cords by western blot and immunofluorescence. A comparative analysis of Basso mouse scale (BMS), footprint test and corticospinal tract (CST) tracing in Snx27 +/+ and Snx27 +/- mice revealed that haploinsufficiency of SNX27 ameliorated the clinical symptoms of SCI. Based on the results of western blot and immunofluorescence, mechanistically, we found that SNX27 deficiency suppresses apoptotic caspase-3 induced neuronal death. In addition, SNX27 haploinsufficiency lowers the infiltration and activation of macrophage/microglia by suppressing their proliferation at the SCI lesion site. Together, these results suggest that down-regulation of SNX27 is a potential therapy targeting both acute neuronal death and chronic neuroinflammation, and promoting nerve repair after SCI.
Collapse
Affiliation(s)
- Yuzhe Zeng
- Department of Orthopaedics, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, China
| | - Nawen Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Collaborative Innovation Center for Brain Science, Xiamen University, Xiamen, China
| | - Tiantian Guo
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Collaborative Innovation Center for Brain Science, Xiamen University, Xiamen, China
| | - Qiuyang Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Collaborative Innovation Center for Brain Science, Xiamen University, Xiamen, China
| | - Shuang Wang
- Department of Orthopaedics, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, China
| | - Songsong Wu
- Department of Orthopaedics, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, China
| | - Xi Li
- Department of Orthopaedics, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, China
| | - Jin Wu
- Department of Orthopaedics, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, China
| | - Zhida Chen
- Department of Orthopaedics, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, China
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Xin Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Collaborative Innovation Center for Brain Science, Xiamen University, Xiamen, China.,State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| | - Bin Lin
- Department of Orthopaedics, The Affiliated Southeast Hospital of Xiamen University, Orthopaedic Center of People's Liberation Army, Zhangzhou, China
| |
Collapse
|
68
|
Zuidema JM, Gilbert RJ, Gottipati MK. Biomaterial Approaches to Modulate Reactive Astroglial Response. Cells Tissues Organs 2018; 205:372-395. [PMID: 30517922 PMCID: PMC6397084 DOI: 10.1159/000494667] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/18/2018] [Indexed: 11/19/2022] Open
Abstract
Over several decades, biomaterial scientists have developed materials to spur axonal regeneration and limit secondary injury and tested these materials within preclinical animal models. Rarely, though, are astrocytes examined comprehensively when biomaterials are placed into the injury site. Astrocytes support neuronal function in the central nervous system. Following an injury, astrocytes undergo reactive gliosis and create a glial scar. The astrocytic glial scar forms a dense barrier which restricts the extension of regenerating axons through the injury site. However, there are several beneficial effects of the glial scar, including helping to reform the blood-brain barrier, limiting the extent of secondary injury, and supporting the health of regenerating axons near the injury site. This review provides a brief introduction to the role of astrocytes in the spinal cord, discusses astrocyte phenotypic changes that occur following injury, and highlights studies that explored astrocyte changes in response to biomaterials tested within in vitro or in vivo environments. Overall, we suggest that in order to improve biomaterial designs for spinal cord injury applications, investigators should more thoroughly consider the astrocyte response to such designs.
Collapse
Affiliation(s)
- Jonathan M Zuidema
- Department of Chemistry and Biochemistry, University of California, San Diego, California, USA
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Manoj K Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA,
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA,
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA,
| |
Collapse
|
69
|
Rudman MD, Choi JS, Lee HE, Tan SK, Ayad NG, Lee JK. Bromodomain and extraterminal domain-containing protein inhibition attenuates acute inflammation after spinal cord injury. Exp Neurol 2018; 309:181-192. [DOI: 10.1016/j.expneurol.2018.08.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/02/2018] [Accepted: 08/15/2018] [Indexed: 01/18/2023]
|
70
|
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
|
71
|
Sanchez S, Lemmens S, Baeten P, Sommer D, Dooley D, Hendrix S, Gou Fabregas M. HDAC3 Inhibition Promotes Alternative Activation of Macrophages but Does Not Affect Functional Recovery after Spinal Cord Injury. Exp Neurobiol 2018; 27:437-452. [PMID: 30429652 PMCID: PMC6221838 DOI: 10.5607/en.2018.27.5.437] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/07/2018] [Accepted: 09/28/2018] [Indexed: 12/31/2022] Open
Abstract
After spinal cord injury (SCI), monocyte derived macrophages play a detrimental role. Histone deacetylases (HDACs) are central epigenetic regulators of macrophage-polarization. We hypothesized that HDAC3 inhibition suppresses the pro-inflammatory macrophage phenotype (M1), promotes the anti-inflammatory phenotype (M2) and improves functional recovery after SCI. Therefore, two inhibitors of HDAC3 were selected, namely scriptaid and RGFP966. The impact on macrophage polarization was studied by investigating the effect on gene and protein expression of selected M1 and M2 markers. We show that scriptaid differentially influences M1 and M2 markers. It increases CD86 and iNOS gene expression and decreases GPR18, CD38, FPR2 and Arg-1 gene expression as well as the production of IL-6 and NO. RGFP966 primarily increased the expression of the M2 markers Arg-1 and Ym1 and reduced the production of IL-6 (M1). RGFP966 and scriptaid reduced the formation of foamy macrophages. Finally, to investigate the impact of HDAC3 inhibition on functional recovery after SCI, we studied the effects of RGFP966 and scriptaid in an in vivo T-cut hemisection SCI model. Histological analyses were performed on spinal cord sections to determine lesion size and astrogliosis, demyelinated area and selected infiltrating immune cells. RGFP966 and scriptaid did not affect functional recovery or histopathological outcome after SCI. In conclusion, these results indicate that specific HDAC3 inhibition with RGFP966 promotes alternative activation of macrophages and reduces the formation of foamy macrophages, but does not lead to a better functional recovery after SCI.
Collapse
Affiliation(s)
- Selien Sanchez
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Stefanie Lemmens
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Paulien Baeten
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Daniela Sommer
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Dearbhaile Dooley
- Health Science Centre, School of Medicine, University College Dublin, Dublin D04 V1W8, Ireland
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Myriam Gou Fabregas
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| |
Collapse
|
72
|
Wang X, Xuan W, Zhu ZY, Li Y, Zhu H, Zhu L, Fu DY, Yang LQ, Li PY, Yu WF. The evolving role of neuro-immune interaction in brain repair after cerebral ischemic stroke. CNS Neurosci Ther 2018; 24:1100-1114. [PMID: 30350341 DOI: 10.1111/cns.13077] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/23/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke is the world's leading cause of disability with limited brain repair treatments which effectively improve long-term neurological deficits. The neuroinflammatory responses persist into the late repair phase of stroke and participate in all brain repair elements, including neurogenesis, angiogenesis, synaptogenesis, remyelination and axonal sprouting, shedding new light on post-stroke brain recovery. Resident brain glial cells, such as astrocytes not only contribute to neuroinflammation after stroke, but also secrete a wide range of trophic factors that can promote post-stroke brain repair. Alternatively, activated microglia, monocytes, and neutrophils in the innate immune system, traditionally considered as major damaging factors after stroke, have been suggested to be extensively involved in brain repair after stroke. The adaptive immune system may also have its bright side during the late regenerative phase, affecting the immune suppressive regulatory T cells and B cells. This review summarizes the recent findings in the evolving role of neuroinflammation in multiple post-stroke brain repair mechanisms and poses unanswered questions that may generate new directions for future research and give rise to novel therapeutic targets to improve stroke recovery.
Collapse
Affiliation(s)
- Xin Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei Xuan
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Yu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hao Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dan-Yun Fu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li-Qun Yang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pei-Ying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
73
|
Norden DM, Faw TD, McKim DB, Deibert RJ, Fisher LC, Sheridan JF, Godbout JP, Basso DM. Bone Marrow-Derived Monocytes Drive the Inflammatory Microenvironment in Local and Remote Regions after Thoracic Spinal Cord Injury. J Neurotrauma 2018; 36:937-949. [PMID: 30014767 DOI: 10.1089/neu.2018.5806] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Spinal cord injury (SCI) produces a toxic inflammatory microenvironment that negatively affects plasticity and recovery. Recently, we showed glial activation and peripheral myeloid cell infiltration extending beyond the epicenter through the remote lumbar cord after thoracic SCI. The presence and role of infiltrating monocytes is important, especially in the lumbar cord where locomotor central pattern generators are housed. Therefore, we compared the inflammatory profile of resident microglia and peripheral myeloid cells after SCI. Bone marrow chimeras received midthoracic contusive SCI, and trafficking was determined 1-7 days later. Fluorescence-activated cell (FAC) sorting showed similar infiltration timing of both neutrophils and macrophages in epicenter and lumbar regions. While neutrophil numbers were attenuated by day 3, macrophages remained unchanged at day 7, suggesting that macrophages have important long-term influence on the microenvironment. Nanostring gene array identified a strong proinflammatory profile of infiltrating macrophages relative to microglia at both epicenter and lumbar sites. Macrophages had elevated expression of inflammatory cytokines (IL-1β, IFNγ), chemokines (CCL2, CXCL2), mediators (COX-1, MMP-9), and receptors (CCR2, Ly6C), and decreased expression of growth promoting genes (GDNF, BDNF). Importantly, lumbar macrophages had elevated expression of active trafficking genes (CCR2, l-selectin, MMP-9) compared with epicenter macrophages. Further, acute rehabilitation exacerbated the inflammatory profile of infiltrated macrophages in the lumbar cord. Such high inflammatory potential and negative response to rehabilitation of infiltrating macrophages within lumbar locomotor central pattern generators likely impedes activity-dependent recovery. Therefore, limiting active trafficking of macrophages into the lumbar cord identifies a novel target for SCI therapies to improve locomotion.
Collapse
Affiliation(s)
- Diana M Norden
- 1 School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio.,2 Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio
| | - Timothy D Faw
- 1 School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio.,2 Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio.,3 Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio
| | - Daniel B McKim
- 3 Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio.,4 Department of Neuroscience, The Ohio State University, Columbus, Ohio.,5 Division of Biosciences, The Ohio State University, Columbus, Ohio
| | - Rochelle J Deibert
- 1 School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio.,2 Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio
| | - Lesley C Fisher
- 1 School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio.,2 Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio
| | - John F Sheridan
- 4 Department of Neuroscience, The Ohio State University, Columbus, Ohio.,5 Division of Biosciences, The Ohio State University, Columbus, Ohio
| | - Jonathan P Godbout
- 2 Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio.,4 Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - D Michele Basso
- 1 School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio.,2 Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio
| |
Collapse
|
74
|
Golabchi A, Wu B, Li X, Carlisle DL, Kozai TDY, Friedlander RM, Cui XT. Melatonin improves quality and longevity of chronic neural recording. Biomaterials 2018; 180:225-239. [PMID: 30053658 PMCID: PMC6179369 DOI: 10.1016/j.biomaterials.2018.07.026] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/06/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022]
Abstract
The chronic performance of implantable neural electrodes is hindered by inflammatory brain tissue responses, including microglia activation, glial scarring, and neuronal loss. Melatonin (MT) has shown remarkable neuroprotective and neurorestorative effects in treating central nervous system (CNS) injuries and degeneration by inhibiting caspase-1, -3, and -9 activation and mitochondrial cytochrome c release, as well as reducing oxidative stress and neuroinflammation. This study examined the effect of MT administration on the quality and longevity of neural recording from an implanted microelectrode in the visual cortex of mice for 16 weeks. MT (30 mg/kg) was administered via daily intraperitoneal injection for acute (3 days before and 14 days post-implantation) and chronic (3 days before and 16 weeks post-implantation) exposures. During the first 4 weeks, both MT groups showed significantly higher single-unit (SU) yield, signal-to-noise ratio (SNR), and amplitude compared to the vehicle control group. However, after 4 weeks of implantation, the SU yield of the acute treatment group dropped to the same level as the control group, while the chronic treatment group maintained significantly higher SU yield compared to both acute (week 5-16) and control (week 0-16) mice. Histological studies revealed a significant increase in neuronal viability and decrease in neuronal apoptosis around the implanted electrode at week 16 in the chronic group in comparison to control and acute subjects, which is correlated with reduced oxidative stress and increased number of pro-regeneration arginase-1 positive microglia cells. These results demonstrate the potent effect of MT treatment in maintaining a high-quality electrode-tissue interface and suggest that MT promotes neuroprotection possibly through its anti-apoptotic, anti-inflammatory, and anti-oxidative properties.
Collapse
Affiliation(s)
- Asiyeh Golabchi
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA
| | - Bingchen Wu
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA
| | - Xia Li
- Department of Bioengineering, University of Pittsburgh, USA
| | - Diane L Carlisle
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, USA; Neurotechnology Division of the University of Pittsburgh Brain Institute, USA
| | - Robert M Friedlander
- Neuroapoptosis Laboratory, Department of Neurological Surgery, University of Pittsburgh School of Medicine, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, USA.
| |
Collapse
|
75
|
Anti-Chondroitin Sulfate Proteoglycan Strategies in Spinal Cord Injury: Temporal and Spatial Considerations Explain the Balance between Neuroplasticity and Neuroprotection. J Neurotrauma 2018. [DOI: 10.1089/neu.2018.5928] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
76
|
Hermann JK, Lin S, Soffer A, Wong C, Srivastava V, Chang J, Sunil S, Sudhakar S, Tomaszewski WH, Protasiewicz G, Selkirk SM, Miller RH, Capadona JR. The Role of Toll-Like Receptor 2 and 4 Innate Immunity Pathways in Intracortical Microelectrode-Induced Neuroinflammation. Front Bioeng Biotechnol 2018; 6:113. [PMID: 30159311 PMCID: PMC6104445 DOI: 10.3389/fbioe.2018.00113] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/19/2018] [Indexed: 12/13/2022] Open
Abstract
We have recently demonstrated that partial inhibition of the cluster of differentiation 14 (CD14) innate immunity co-receptor pathway improves the long-term performance of intracortical microelectrodes better than complete inhibition. We hypothesized that partial activation of the CD14 pathway was critical to a neuroprotective response to the injury associated with initial and sustained device implantation. Therefore, here we investigated the role of two innate immunity receptors that closely interact with CD14 in inflammatory activation. We implanted silicon planar non-recording neural probes into knockout mice lacking Toll-like receptor 2 (Tlr2-/-), knockout mice lacking Toll-like receptor 4 (Tlr4-/-), and wildtype (WT) control mice, and evaluated endpoint histology at 2 and 16 weeks after implantation. Tlr4-/- mice exhibited significantly lower BBB permeability at acute and chronic time points, but also demonstrated significantly lower neuronal survival at the chronic time point. Inhibition of the Toll-like receptor 2 (TLR2) pathway had no significant effect compared to control animals. Additionally, when investigating the maturation of the neuroinflammatory response from 2 to 16 weeks, transgenic knockout mice exhibited similar histological trends to WT controls, except that knockout mice did not exhibit changes in microglia and macrophage activation over time. Together, our results indicate that complete genetic removal of Toll-like receptor 4 (TLR4) was detrimental to the integration of intracortical neural probes, while inhibition of TLR2 had no impact within the tests performed in this study. Therefore, approaches focusing on incomplete or acute inhibition of TLR4 may still improve intracortical microelectrode integration and long term recording performance.
Collapse
Affiliation(s)
- John K. Hermann
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Shushen Lin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Arielle Soffer
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Chun Wong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Vishnupriya Srivastava
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Jeremy Chang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Smrithi Sunil
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Shruti Sudhakar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - William H. Tomaszewski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Grace Protasiewicz
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Stephen M. Selkirk
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Department of Neurology, Case Western Reserve University, Cleveland, OH, United States
- Spinal Cord Injury Division, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Robert H. Miller
- Neurosciences, George Washington University, Washington, DC, United States
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| |
Collapse
|
77
|
Nakayama H, Abe M, Morimoto C, Iida T, Okabe S, Sakimura K, Hashimoto K. Microglia permit climbing fiber elimination by promoting GABAergic inhibition in the developing cerebellum. Nat Commun 2018; 9:2830. [PMID: 30026565 PMCID: PMC6053401 DOI: 10.1038/s41467-018-05100-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/23/2018] [Indexed: 01/01/2023] Open
Abstract
Circuit refinement during postnatal development is finely regulated by neuron–neuron interactions. Recent studies suggest participation of microglia in this process but it is unclear how microglia cooperatively act with neuronal mechanisms. To examine roles of microglia, we ablate microglia by microglia-selective deletion of colony-stimulating factor 1 receptor (Csf1r) by crossing floxed-Csf1r and Iba1-iCre mice (Csf1r-cKO). In Csf1r-cKO mice, refinement of climbing fiber (CF) to Purkinje cell (PC) innervation after postnatal day 10 (P10)–P12 is severely impaired. However, there is no clear morphological evidence suggesting massive engulfment of CFs by microglia. In Csf1r-cKO mice, inhibitory synaptic transmission is impaired and CF elimination is restored by diazepam, which suggests that impairment of CF elimination is caused by a defect of GABAergic inhibition on PCs, a prerequisite for CF elimination. These results indicate that microglia primarily promote GABAergic inhibition and secondarily facilitate the mechanism for CF elimination inherent in PCs. In the mammalian cerebellum, surplus synapses between climbing fibers (CF) and Purkinje cells (PC) are developmentally pruned. Here, Nakayama and colleagues show that ablation of microglia impairs pruning of CF-PC synapses because of dysfunction of GABAergic inhibition prerequisite for pruning.
Collapse
Affiliation(s)
- Hisako Nakayama
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.,Department of Physiology, School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Sinjuku-ku, Tokyo, 162-8666, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8585, Japan.,Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8585, Japan
| | - Chie Morimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.,Department of Psychosocial Rehabilitation Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Tadatsune Iida
- Department of Cellular Neurobiology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8585, Japan.,Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuo-ku, Niigata, 951-8585, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
78
|
Orr MB, Gensel JC. Spinal Cord Injury Scarring and Inflammation: Therapies Targeting Glial and Inflammatory Responses. Neurotherapeutics 2018; 15:541-553. [PMID: 29717413 PMCID: PMC6095779 DOI: 10.1007/s13311-018-0631-6] [Citation(s) in RCA: 350] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Deficits in neuronal function are a hallmark of spinal cord injury (SCI) and therapeutic efforts are often focused on central nervous system (CNS) axon regeneration. However, secondary injury responses by astrocytes, microglia, pericytes, endothelial cells, Schwann cells, fibroblasts, meningeal cells, and other glia not only potentiate SCI damage but also facilitate endogenous repair. Due to their profound impact on the progression of SCI, glial cells and modification of the glial scar are focuses of SCI therapeutic research. Within and around the glial scar, cells deposit extracellular matrix (ECM) proteins that affect axon growth such as chondroitin sulfate proteoglycans (CSPGs), laminin, collagen, and fibronectin. This dense deposition of material, i.e., the fibrotic scar, is another barrier to endogenous repair and is a target of SCI therapies. Infiltrating neutrophils and monocytes are recruited to the injury site through glial chemokine and cytokine release and subsequent upregulation of chemotactic cellular adhesion molecules and selectins on endothelial cells. These peripheral immune cells, along with endogenous microglia, drive a robust inflammatory response to injury with heterogeneous reparative and pathological properties and are targeted for therapeutic modification. Here, we review the role of glial and inflammatory cells after SCI and the therapeutic strategies that aim to replace, dampen, or alter their activity to modulate SCI scarring and inflammation and improve injury outcomes.
Collapse
Affiliation(s)
- Michael B Orr
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky College of Medicine, 741 S. Limestone, B463 BBSRB, Lexington, Kentucky, 40536, USA
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky College of Medicine, 741 S. Limestone, B463 BBSRB, Lexington, Kentucky, 40536, USA.
| |
Collapse
|
79
|
Abstract
Glial cell types were classified less than 100 years ago by del Rio-Hortega. For instance, he correctly surmised that microglia in pathologic central nervous system (CNS) were "voracious monsters" that helped clean the tissue. Although these historical predictions were remarkably accurate, innovative technologies have revealed novel molecular, cellular, and dynamic physiologic aspects of CNS glia. In this review, we integrate recent findings regarding the roles of glia and glial interactions in healthy and injured spinal cord. The three major glial cell types are considered in healthy CNS and after spinal cord injury (SCI). Astrocytes, which in the healthy CNS regulate neurotransmitter and neurovascular dynamics, respond to SCI by becoming reactive and forming a glial scar that limits pathology and plasticity. Microglia, which in the healthy CNS scan for infection/damage, respond to SCI by promoting axon growth and remyelination-but also with hyperactivation and cytotoxic effects. Oligodendrocytes and their precursors, which in healthy tissue speed axon conduction and support axonal function, respond to SCI by differentiating and producing myelin, but are susceptible to death. Thus, post-SCI responses of each glial cell can simultaneously stimulate and stifle repair. Interestingly, potential therapies could also target interactions between these cells. Astrocyte-microglia cross-talk creates a feed-forward loop, so shifting the response of either cell could amplify repair. Astrocytes, microglia, and oligodendrocytes/precursors also influence post-SCI cell survival, differentiation, and remyelination, as well as axon sparing. Therefore, optimizing post-SCI responses of glial cells-and interactions between these CNS cells-could benefit neuroprotection, axon plasticity, and functional recovery.
Collapse
Affiliation(s)
- Andrew D Gaudet
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO, 80309, USA.
- Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO, 80309, USA.
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
80
|
Attwell CL, van Zwieten M, Verhaagen J, Mason MRJ. The Dorsal Column Lesion Model of Spinal Cord Injury and Its Use in Deciphering the Neuron-Intrinsic Injury Response. Dev Neurobiol 2018; 78:926-951. [PMID: 29717546 PMCID: PMC6221129 DOI: 10.1002/dneu.22601] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 12/13/2022]
Abstract
The neuron‐intrinsic response to axonal injury differs markedly between neurons of the peripheral and central nervous system. Following a peripheral lesion, a robust axonal growth program is initiated, whereas neurons of the central nervous system do not mount an effective regenerative response. Increasing the neuron‐intrinsic regenerative response would therefore be one way to promote axonal regeneration in the injured central nervous system. The large‐diameter sensory neurons located in the dorsal root ganglia are pseudo‐unipolar neurons that project one axon branch into the spinal cord, and, via the dorsal column to the brain stem, and a peripheral process to the muscles and skin. Dorsal root ganglion neurons are ideally suited to study the neuron‐intrinsic injury response because they exhibit a successful growth response following peripheral axotomy, while they fail to do so after a lesion of the central branch in the dorsal column. The dorsal column injury model allows the neuron‐intrinsic regeneration response to be studied in the context of a spinal cord injury. Here we will discuss the advantages and disadvantages of this model. We describe the surgical methods used to implement a lesion of the ascending fibers, the anatomy of the sensory afferent pathways and anatomical, electrophysiological, and behavioral techniques to quantify regeneration and functional recovery. Subsequently we review the results of experimental interventions in the dorsal column lesion model, with an emphasis on the molecular mechanisms that govern the neuron‐intrinsic injury response and manipulations of these after central axotomy. Finally, we highlight a number of recent advances that will have an impact on the design of future studies in this spinal cord injury model, including the continued development of adeno‐associated viral vectors likely to improve the genetic manipulation of dorsal root ganglion neurons and the use of tissue clearing techniques enabling 3D reconstruction of regenerating axon tracts. © 2018 The Authors. Developmental Neurobiology Published by Wiley Periodicals, Inc. Develop Neurobiol 00: 000–000, 2018
Collapse
Affiliation(s)
- Callan L Attwell
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, Amsterdam, 1105BA, The Netherlands
| | - Mike van Zwieten
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, Amsterdam, 1105BA, The Netherlands
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, Amsterdam, 1105BA, The Netherlands.,Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam, 1081HV, The Netherlands
| | - Matthew R J Mason
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Science, Meibergdreef 47, Amsterdam, 1105BA, The Netherlands
| |
Collapse
|
81
|
Wrobel MR, Sundararaghavan HG. Biomaterial Cues to Direct a Pro-regenerative Phenotype in Macrophages and Schwann Cells. Neuroscience 2018; 376:172-187. [DOI: 10.1016/j.neuroscience.2018.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/23/2018] [Accepted: 02/09/2018] [Indexed: 12/11/2022]
|
82
|
Ghosh S, Hui SP. Axonal regeneration in zebrafish spinal cord. REGENERATION (OXFORD, ENGLAND) 2018; 5:43-60. [PMID: 29721326 PMCID: PMC5911453 DOI: 10.1002/reg2.99] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
In the present review we discuss two interrelated events-axonal damage and repair-known to occur after spinal cord injury (SCI) in the zebrafish. Adult zebrafish are capable of regenerating axonal tracts and can restore full functionality after SCI. Unlike fish, axon regeneration in the adult mammalian central nervous system is extremely limited. As a consequence of an injury there is very little repair of disengaged axons and therefore functional deficit persists after SCI in adult mammals. In contrast, peripheral nervous system axons readily regenerate following injury and hence allow functional recovery both in mammals and fish. A better mechanistic understanding of these three scenarios could provide a more comprehensive insight into the success or failure of axonal regeneration after SCI. This review summarizes the present understanding of the cellular and molecular basis of axonal regeneration, in both the peripheral nervous system and the central nervous system, and large scale gene expression analysis is used to focus on different events during regeneration. The discovery and identification of genes involved in zebrafish spinal cord regeneration and subsequent functional experimentation will provide more insight into the endogenous mechanism of myelination and remyelination. Furthermore, precise knowledge of the mechanism underlying the extraordinary axonal regeneration process in zebrafish will also allow us to unravel the potential therapeutic strategies to be implemented for enhancing regrowth and remyelination of axons in mammals.
Collapse
Affiliation(s)
- Sukla Ghosh
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
| | - Subhra Prakash Hui
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
- Victor Chang Cardiac Research InstituteLowy Packer Building, 405 Liverpool StDarlinghurstNSW 2010Australia.
| |
Collapse
|
83
|
Truflandier K, Beaumont E, Charbonney E, Maghni K, de Marchie M, Spahija J. Mechanical ventilation modulates pro-inflammatory cytokine expression in spinal cord tissue after injury in rats. Neurosci Lett 2018; 671:13-18. [PMID: 29355694 DOI: 10.1016/j.neulet.2018.01.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 01/15/2018] [Accepted: 01/15/2018] [Indexed: 11/24/2022]
Abstract
RATIONALE Spinal cord injury (SCI) may induce significant respiratory muscle weakness and paralysis, which in turn may cause a patient to require ventilator support. Central nervous system alterations can also exacerbate local inflammatory responses with immune cell infiltration leading to additional risk of inflammation at the injury site. Although mechanical ventilation is the traditional treatment for respiratory insufficiency, evidence has shown that it may directly affect distant organs through systemic inflammation. OBJECTIVES This study aimed to better understand the impact of invasive mechanical ventilation on local spinal cord inflammatory responses following cervical or thoracic SCI. METHODS Five groups of female Sprague-Dawley rats were anesthetised for 24 h. Three groups received mechanical ventilation: seven rats without SCI, seven rats with cervical injury (C4-C5), and seven rats with thoracic injury (T10); whereas, two groups were non-ventilated: six rats without SCI; and six rats with thoracic injury (T10). Changes in inflammatory responses were determined in the spinal cord tissues collected at the local site of injury. Cytokines were measured using ELISA. MAIN RESULTS SCI induced local pro-inflammatory cytokine IL-6 expression for all groups. Mechanical ventilation also had effects on pro-inflammatory cytokines and independently increased TNF-α and decreased IL-1β levels in the spinal cords of anesthetized rats. CONCLUSION These data provide the first evidence that mechanical ventilation contributes to local inflammation after SCI and in the absence of direct tissue injury.
Collapse
Affiliation(s)
- Karine Truflandier
- Research Center, CIUSSS du Nord-de-l'Ile-de-Montréal, Sacré-Coeur Hospital, Department of Medicine, Université de Montréal, 5400 boul. Gouin Ouest, Montréal, Quebec, H4J 1C5, Canada
| | - Eric Beaumont
- Research Center, CIUSSS du Nord-de-l'Ile-de-Montréal, Sacré-Coeur Hospital, Department of Medicine, Université de Montréal, 5400 boul. Gouin Ouest, Montréal, Quebec, H4J 1C5, Canada; Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Emmanuel Charbonney
- Research Center, CIUSSS du Nord-de-l'Ile-de-Montréal, Sacré-Coeur Hospital, Department of Medicine, Université de Montréal, 5400 boul. Gouin Ouest, Montréal, Quebec, H4J 1C5, Canada
| | - Karim Maghni
- Research Center, CIUSSS du Nord-de-l'Ile-de-Montréal, Sacré-Coeur Hospital, Department of Medicine, Université de Montréal, 5400 boul. Gouin Ouest, Montréal, Quebec, H4J 1C5, Canada
| | - Michel de Marchie
- Department of Adult Critical Care, Jewish General Hospital, McGill University, Montreal, Quebec, H3T 1E2, Canada
| | - Jadranka Spahija
- Research Center, CIUSSS du Nord-de-l'Ile-de-Montréal, Sacré-Coeur Hospital, Department of Medicine, Université de Montréal, 5400 boul. Gouin Ouest, Montréal, Quebec, H4J 1C5, Canada; School of Physical and Occupational Therapy, McGill University, 3654 Promenade Sir William Osler, Montreal, Quebec, H3G 1Y5, Canada; Center for Interdisciplinary Research in Rehabilitation in Montreal, CISSS de Laval, Jewish Rehabilitation Hospital, 3205, Place Alton-Goldbloom, Laval, Quebec, H7V 1J1, Canada.
| |
Collapse
|
84
|
Usage of Multiparameter Flow Cytometry to Study Microglia and Macrophage Heterogeneity in the Central Nervous System During Neuroinflammation and Neurodegeneration. Methods Mol Biol 2018; 1745:167-177. [PMID: 29476469 DOI: 10.1007/978-1-4939-7680-5_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The resident macrophages of the central nervous system (CNS), also known as microglia, and blood-derived macrophages play an important role in the functional activity of the normal CNS, as well as in the development of neuroinflammation during various neurodegenerative disorders. Microglia and macrophages represent heterogeneous populations, which can modulate CNS environment and have different effects on neuronal regeneration. In this chapter, the main features of microglial and macrophage subsets and current methods for investigation of their heterogeneity will be discussed.
Collapse
|
85
|
van der Merwe Y, Faust AE, Conner I, Gu X, Feturi F, Zhao W, Leonard B, Roy S, Gorantla VS, Venkataramanan R, Washington KM, Wagner WR, Steketee MB. An Elastomeric Polymer Matrix, PEUU-Tac, Delivers Bioactive Tacrolimus Transdurally to the CNS in Rat. EBioMedicine 2017; 26:47-59. [PMID: 29208469 PMCID: PMC5832622 DOI: 10.1016/j.ebiom.2017.11.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/10/2017] [Accepted: 11/20/2017] [Indexed: 12/13/2022] Open
Abstract
Central nervous system (CNS) neurons fail to regrow injured axons, often resulting in permanently lost neurologic function. Tacrolimus is an FDA-approved immunosuppressive drug with known neuroprotective and neuroregenerative properties in the CNS. However, tacrolimus is typically administered systemically and blood levels required to effectively treat CNS injuries can lead to lethal, off-target organ toxicity. Thus, delivering tacrolimus locally to CNS tissues may provide therapeutic control over tacrolimus levels in CNS tissues while minimizing off-target toxicity. Herein we show an electrospun poly(ester urethane) urea and tacrolimus elastomeric matrix (PEUU-Tac) can deliver tacrolimus trans-durally to CNS tissues. In an acute CNS ischemia model in rat, the optic nerve (ON) was clamped for 10s and then PEUU-Tac was used as an ON wrap and sutured around the injury site. Tacrolimus was detected in PEUU-Tac wrapped ONs at 24 h and 14 days, without significant increases in tacrolimus blood levels. Similar to systemically administered tacrolimus, PEUU-Tac locally decreased glial fibrillary acidic protein (GFAP) at the injury site and increased growth associated protein-43 (GAP-43) expression in ischemic ONs from the globe to the chiasm, consistent with decreased astrogliosis and increased retinal ganglion cell (RGC) axon growth signaling pathways. These initial results suggest PEUU-Tac is a biocompatible elastic matrix that delivers bioactive tacrolimus trans-durally to CNS tissues without significantly increasing tacrolimus blood levels and off-target toxicity. PEUU-Tac locally delivers tacrolimus to CNS tissues PEUU-Tac positively modulates CNS tissue remodeling PEUU-Tac minimizes off-target tacrolimus toxicity
Central nervous system (CNS) injury typically results in permanently lost neurological function. Tacrolimus is an FDA-approved drug used during organ transplantation that also has CNS neuroprotective and neuroregenerative properties. However, tacrolimus is typically delivered systemically in the blood and delivering effective concentrations to CNS tissues requires tacrolimus blood levels that can lead to adverse side effects in multiple organs. Herein we show that PEUU-Tac, a tacrolimus-eluting matrix, can locally deliver tacrolimus to injured CNS tissues without increasing blood levels, suggesting PEUU-Tac can be used to treat CNS injuries locally while minimizing adverse side effects.
Collapse
Affiliation(s)
- Yolandi van der Merwe
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anne E Faust
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ian Conner
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xinzhu Gu
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Firuz Feturi
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | - Wenchen Zhao
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bianca Leonard
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Souvik Roy
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - Vijay S Gorantla
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Departments of Surgery, Ophthalmology and Bioengineering, Wake Forest School of Medicine, Wake Forest Institute of Regenerative Medicine, Winston Salem, NC, United States
| | - Raman Venkataramanan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, United States; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kia M Washington
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, United States; VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - William R Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael B Steketee
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
86
|
Cell-Based Delivery of Interleukin-13 Directs Alternative Activation of Macrophages Resulting in Improved Functional Outcome after Spinal Cord Injury. Stem Cell Reports 2017; 7:1099-1115. [PMID: 27974221 PMCID: PMC5161742 DOI: 10.1016/j.stemcr.2016.11.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 11/10/2016] [Accepted: 11/10/2016] [Indexed: 01/19/2023] Open
Abstract
The therapeutic effects of mesenchymal stem cell (MSC) transplantation following spinal cord injury (SCI) to date have been limited. Therefore, we aimed to enhance the immunomodulatory properties of MSCs via continuous secretion of the anti-inflammatory cytokine interleukin-13 (IL-13). By using MSCs as carriers of IL-13 (MSC/IL-13), we investigated their therapeutic potential, compared with non-engineered MSCs, in a mouse model of SCI. We show that transplanted MSC/IL-13 significantly improve functional recovery following SCI, and also decrease lesion size and demyelinated area by more than 40%. Further histological analyses in CX3CR1EGFP/+ CCR2RFP/+ transgenic mice indicated that MSC/IL-13 significantly decrease the number of resident microglia and increase the number of alternatively activated macrophages. In addition, the number of macrophage-axon contacts in MSC/IL-13-treated mice was decreased by 50%, suggesting a reduction in axonal dieback. Our data provide evidence that transplantation of MSC/IL-13 leads to improved functional and histopathological recovery in a mouse model of SCI.
Collapse
|
87
|
Filous AR, Schwab JM. Determinants of Axon Growth, Plasticity, and Regeneration in the Context of Spinal Cord Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:53-62. [PMID: 29030051 DOI: 10.1016/j.ajpath.2017.09.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 09/21/2017] [Indexed: 12/30/2022]
Abstract
The mechanisms that underlie recovery after injury of the central nervous system have rarely been definitively established. Axon regrowth remains the major prerequisite for plasticity, regeneration, circuit formation, and eventually functional recovery. The attributed functional relevance of axon regrowth, however, will depend on several subsequent conditional neurobiological modifications, including myelination and synapse formation, but also pruning of aberrant connectivity. Despite the ability to revamp axon outgrowth by altering an increasing number of extracellular and intracellular targets, disentangling which axons are responsible for the recovery of function from those that are functionally silent, or even contributing to aberrant functions, represents a pertinent void in our understanding, challenging the intuitive translational link between anatomical and functional regeneration. Anatomic hallmarks of regeneration are not static and are largely activity dependent. Herein, we survey mechanisms leading to the formation of dystrophic growth cone at the injured axonal tip, the subsequent axonal dieback, and the molecular determinants of axon growth, plasticity, and regeneration in the context of spinal cord injury.
Collapse
Affiliation(s)
- Angela R Filous
- Spinal Cord Injury Division, Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, Ohio.
| | - Jan M Schwab
- Spinal Cord Injury Division, Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, Ohio; Department of Neuroscience, The Ohio State University, Wexner Medical Center, Columbus, Ohio; Department of Physical Medicine and Rehabilitation, The Ohio State University, Wexner Medical Center, Columbus, Ohio; Center for Brain and Spinal Cord Repair, Spinal Cord Injury Medicine, The Ohio State University, Wexner Medical Center, Columbus, Ohio.
| |
Collapse
|
88
|
Ludwig PE, Patil AA, Chamczuk AJ, Agrawal DK. Hormonal therapy in traumatic spinal cord injury. Am J Transl Res 2017; 9:3881-3895. [PMID: 28979667 PMCID: PMC5622236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/25/2017] [Indexed: 06/07/2023]
Abstract
Traumatic spinal cord injuries are major health problems and the underlying pathophysiological events and treatment strategies are currently under investigation. In this article, we critically reviewed the literature investigating the effects of estrogen, progesterone, and human chorionic gonadotropin on spinal cord damage or preservation following traumatic spinal cord injury. The National Library of Medicine database was searched through December 2016 using PubMed for articles addressing the clinical relevance of the hormones to improve neural structural integrity following traumatic spinal cord injury. It was found that each of these hormones, through varied mechanisms, could serve to reduce the harmful effects associated with spinal cord injury, and could aid in restoring some function to the injured spinal cord in the animal models. The most striking effects were seen in the reduction of inflammation commonly linked to injury of the central nervous system. The effects of human chorionic gonadotropin administration following spinal cord injury have received far less attention than those of either estrogen or progesterone, and additional inquiry could be of general benefit. In this article, we discussed the outstanding questions and suggested future directions for further investigation.
Collapse
Affiliation(s)
- Parker E Ludwig
- Department of Clinical and Translational Science, Creighton University School of MedicineOmaha, NE, USA
| | - Arun A Patil
- Department of Clinical and Translational Science, Creighton University School of MedicineOmaha, NE, USA
- Department of Neurosurgery, Creighton University School of MedicineOmaha, NE, USA
| | - Andrea J Chamczuk
- Department of Neurosurgery, Creighton University School of MedicineOmaha, NE, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of MedicineOmaha, NE, USA
| |
Collapse
|
89
|
Chedly J, Soares S, Montembault A, von Boxberg Y, Veron-Ravaille M, Mouffle C, Benassy MN, Taxi J, David L, Nothias F. Physical chitosan microhydrogels as scaffolds for spinal cord injury restoration and axon regeneration. Biomaterials 2017; 138:91-107. [DOI: 10.1016/j.biomaterials.2017.05.024] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/04/2023]
|
90
|
Orlandin JR, Ambrósio CE, Lara VM. Glial scar-modulation as therapeutic tool in spinal cord injury in animal models. Acta Cir Bras 2017; 32:168-174. [PMID: 28300871 DOI: 10.1590/s0102-865020170209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/20/2017] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Spinal Cord injury represents, in veterinary medicine, most of the neurological attendances and may result in permanent disability, death or euthanasia. Due to inflammation resulting from trauma, it originates the glial scar, which is a cell interaction complex system. Its function is to preserve the healthy circuits, however, it creates a physical and molecular barrier that prevents cell migration and restricts the neuroregeneration ability. METHODS This review aims to present innovations in the scene of treatment of spinal cord injury, approaching cell therapy, administration of enzyme, anti-inflammatory, and other active principles capable of modulating the inflammatory response, resulting in glial scar reduction and subsequent functional improvement of animals. RESULTS Some innovative therapies as cell therapy, administration of enzymes, immunosuppressant or other drugs cause the modulation of inflammatory response proved to be a promising tool for the reduction of gliosis. CONCLUSION Those tools promise to reduce gliosis and promote locomotor recovery in animals with spinal cord injury.
Collapse
Affiliation(s)
- Jéssica Rodrigues Orlandin
- Veterinary Medicine Department, Faculty of Animal Science and Food Engineering, Universidade de São Paulo, Pirassununga, SP, Brazil
| | | | | |
Collapse
|
91
|
Jeong SJ, Cooper JG, Ifergan I, McGuire TL, Xu D, Hunter Z, Sharma S, McCarthy D, Miller SD, Kessler JA. Intravenous immune-modifying nanoparticles as a therapy for spinal cord injury in mice. Neurobiol Dis 2017; 108:73-82. [PMID: 28823935 DOI: 10.1016/j.nbd.2017.08.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/17/2017] [Accepted: 08/16/2017] [Indexed: 01/16/2023] Open
Abstract
Intravenously infused synthetic 500nm nanoparticles composed of poly(lactide-co-glycolide) are taken up by blood-borne inflammatory monocytes via a macrophage scavenger receptor (macrophage receptor with collagenous structure), and the monocytes no longer traffic to sites of inflammation. Intravenous administration of the nanoparticles after experimental spinal cord injury in mice safely and selectively limited infiltration of hematogenous monocytes into the injury site. The nanoparticles did not bind to resident microglia, and did not change the number of microglia in the injured spinal cord. Nanoparticle administration reduced M1 macrophage polarization and microglia activation, reduced levels of inflammatory cytokines, and markedly reduced fibrotic scar formation without altering glial scarring. These findings thus implicate early-infiltrating hematogenous monocytes as highly selective contributors to fibrosis that do not play an indispensable role in gliosis after SCI. Further, the nanoparticle treatment reduced accumulation of chondroitin sulfate proteoglycans, increased axon density inside and caudal to the lesion site, and significantly improved functional recovery after both moderate and severe injuries to the spinal cord. These data provide further evidence that hematogenous monocytes contribute to inflammatory damage and fibrotic scar formation after spinal cord injury in mice. Further, since the nanoparticles are simple to administer intravenously, immunologically inert, stable at room temperature, composed of an FDA-approved material, and have no known toxicity, these findings suggest that the nanoparticles potentially offer a practical treatment for human spinal cord injury.
Collapse
Affiliation(s)
- Su Ji Jeong
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John G Cooper
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Igal Ifergan
- Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, IL 60611, USA
| | - Tammy L McGuire
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dan Xu
- Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, IL 60611, USA
| | - Zoe Hunter
- Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, IL 60611, USA
| | - Sripadh Sharma
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Derrick McCarthy
- Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, IL 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology and the Interdepartmental Immunobiology Center, Northwestern University Medical School, Chicago, IL 60611, USA.
| | - John A Kessler
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
92
|
The toll-like receptor 2 agonist Pam3CSK4 is neuroprotective after spinal cord injury. Exp Neurol 2017; 294:1-11. [DOI: 10.1016/j.expneurol.2017.04.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/20/2017] [Accepted: 04/22/2017] [Indexed: 12/21/2022]
|
93
|
Abstract
Spinal cord injury (SCI) lesions present diverse challenges for repair strategies. Anatomically complete injuries require restoration of neural connectivity across lesions. Anatomically incomplete injuries may benefit from augmentation of spontaneous circuit reorganization. Here, we review SCI cell biology, which varies considerably across three different lesion-related tissue compartments: (a) non-neural lesion core, (b) astrocyte scar border, and (c) surrounding spared but reactive neural tissue. After SCI, axon growth and circuit reorganization are determined by neuron-cell-autonomous mechanisms and by interactions among neurons, glia, and immune and other cells. These interactions are shaped by both the presence and the absence of growth-modulating molecules, which vary markedly in different lesion compartments. The emerging understanding of how SCI cell biology differs across lesion compartments is fundamental to developing rationally targeted repair strategies.
Collapse
|
94
|
miR-155 Deletion in Mice Overcomes Neuron-Intrinsic and Neuron-Extrinsic Barriers to Spinal Cord Repair. J Neurosci 2017; 36:8516-32. [PMID: 27511021 DOI: 10.1523/jneurosci.0735-16.2016] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/08/2016] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Axon regeneration after spinal cord injury (SCI) fails due to neuron-intrinsic mechanisms and extracellular barriers including inflammation. microRNA (miR)-155-5p is a small, noncoding RNA that negatively regulates mRNA translation. In macrophages, miR-155-5p is induced by inflammatory stimuli and elicits a response that could be toxic after SCI. miR-155 may also independently alter expression of genes that regulate axon growth in neurons. Here, we hypothesized that miR-155 deletion would simultaneously improve axon growth and reduce neuroinflammation after SCI by acting on both neurons and macrophages. New data show that miR-155 deletion attenuates inflammatory signaling in macrophages, reduces macrophage-mediated neuron toxicity, and increases macrophage-elicited axon growth by ∼40% relative to control conditions. In addition, miR-155 deletion increases spontaneous axon growth from neurons; adult miR-155 KO dorsal root ganglion (DRG) neurons extend 44% longer neurites than WT neurons. In vivo, miR-155 deletion augments conditioning lesion-induced intraneuronal expression of SPRR1A, a regeneration-associated gene; ∼50% more injured KO DRG neurons expressed SPRR1A versus WT neurons. After dorsal column SCI, miR-155 KO mouse spinal cord has reduced neuroinflammation and increased peripheral conditioning-lesion-enhanced axon regeneration beyond the epicenter. Finally, in a model of spinal contusion injury, miR-155 deletion improves locomotor function at postinjury times corresponding with the arrival and maximal appearance of activated intraspinal macrophages. In miR-155 KO mice, improved locomotor function is associated with smaller contusion lesions and decreased accumulation of inflammatory macrophages. Collectively, these data indicate that miR-155 is a novel therapeutic target capable of simultaneously overcoming neuron-intrinsic and neuron-extrinsic barriers to repair after SCI. SIGNIFICANCE STATEMENT Axon regeneration after spinal cord injury (SCI) fails due to neuron-intrinsic mechanisms and extracellular barriers, including inflammation. Here, new data show that deleting microRNA-155 (miR-155) affects both mechanisms and improves repair and functional recovery after SCI. Macrophages lacking miR-155 have altered inflammatory capacity, which enhances neuron survival and axon growth of cocultured neurons. In addition, independent of macrophages, adult miR-155 KO neurons show enhanced spontaneous axon growth. Using either spinal cord dorsal column crush or contusion injury models, miR-155 deletion improves indices of repair and recovery. Therefore, miR-155 has a dual role in regulating spinal cord repair and may be a novel therapeutic target for SCI and other CNS pathologies.
Collapse
|
95
|
Hill CE. A view from the ending: Axonal dieback and regeneration following SCI. Neurosci Lett 2017; 652:11-24. [DOI: 10.1016/j.neulet.2016.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/20/2016] [Accepted: 11/01/2016] [Indexed: 12/22/2022]
|
96
|
Hilton BJ, Moulson AJ, Tetzlaff W. Neuroprotection and secondary damage following spinal cord injury: concepts and methods. Neurosci Lett 2017; 652:3-10. [DOI: 10.1016/j.neulet.2016.12.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/28/2016] [Accepted: 12/02/2016] [Indexed: 01/29/2023]
|
97
|
Microglia Are Irrelevant for Neuronal Degeneration and Axon Regeneration after Acute Injury. J Neurosci 2017; 37:6113-6124. [PMID: 28539419 DOI: 10.1523/jneurosci.0584-17.2017] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 03/29/2017] [Accepted: 04/26/2017] [Indexed: 12/15/2022] Open
Abstract
The role of microglia in degenerative and regenerative processes after damage of the nervous system remains ambiguous, partially due to the paucity of appropriate investigative methods. Here, we show that treatment with the pharmacological colony stimulating factor 1 receptor inhibitor PLX5622 specifically eliminated microglia in murine retinae and optic nerves with high efficiency. Interestingly, time course and extent of retinal ganglion cell (RGC) degeneration after optic nerve crush remained unaffected upon microglia depletion, although remnants of prelabeled apoptotic RGCs were not cleared from the retina in these animals. In addition, microglia depletion neither affected the induction of regeneration associated genes upon optic nerve injury nor the increased regenerative potential of RGCs upon lens injury (LI). However, although the repopulation of the optic nerve lesion site by astrocytes was significantly delayed upon microglia depletion, spontaneous and LI-induced axon regeneration were unaffected by PLX5622 treatment or peripheral macrophage depletion by clodronate liposome treatment. Only concurrent double depletion of microglia and infiltrated macrophages slightly, but significantly, compromised optic nerve regeneration. Therefore, microglia are not essentially involved in RGC degeneration or axonal regeneration after acute CNS injury.SIGNIFICANCE STATEMENT The roles of microglia, the phagocytosing cells of the CNS, and invading macrophages in degenerative and regenerative processes after injury are still controversial and insufficiently characterized. Here, we show that application of a CSF1R inhibitor eliminated virtually all microglia from the visual system, whereas macrophages were spared. Specific microglia depletion impaired the removal of dead labeled retinal ganglion cells after optic nerve crush, but remarkable had no influence on their degeneration. Similarly, optic nerve regeneration was completely unaffected, although repopulation of the lesion site by astrocytes was delayed significantly. Therefore, contrary to previous reports, this experimental approach revealed that microglia seemingly neither promote nor inhibit neuronal degeneration or axonal regrowth within the injured visual system.
Collapse
|
98
|
Low-level laser facilitates alternatively activated macrophage/microglia polarization and promotes functional recovery after crush spinal cord injury in rats. Sci Rep 2017; 7:620. [PMID: 28377600 PMCID: PMC5428709 DOI: 10.1038/s41598-017-00553-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/03/2017] [Indexed: 12/14/2022] Open
Abstract
Macrophages and resident microglia play an import role in the secondary neuroinflammation response following spinal cord injury. Reprogramming of macrophage/microglia polarization is an import strategy for spinal cord injury restoration. Low-level laser therapy (LLLT) is a noninvasive treatment that has been widely used in neurotrauma and neurodegenerative diseases. However, the influence of low-level laser on polarization of macrophage/microglia following spinal cord injury remains unknown. The present study applied low-level laser therapy on a crush spinal cord injury rat model. Using immunofluorescence, flow cytometry, RT-qPCR, and western blot assays, we found that low-level laser therapy altered the polarization state to a M2 tendency. A greater number of neurons survived in the pare injury site, which was accompanied by higher BBB scores in the LLLT group. Furthermore, low-level laser therapy elevated expression of interleukin 4 (IL-4) and interleukin 13 (IL-13). Results from this study show that low-level laser therapy has the potential for reducing inflammation, regulating macrophage/microglia polarization, and promoting neuronal survival. These beneficial effects demonstrate that low-level laser therapy may be an effective candidate for clinical treatment of spinal cord injury.
Collapse
|
99
|
Faust A, Kandakatla A, van der Merwe Y, Ren T, Huleihel L, Hussey G, Naranjo JD, Johnson S, Badylak S, Steketee M. Urinary bladder extracellular matrix hydrogels and matrix-bound vesicles differentially regulate central nervous system neuron viability and axon growth and branching. J Biomater Appl 2017; 31:1277-1295. [PMID: 28447547 DOI: 10.1177/0885328217698062] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Central nervous system neurons often degenerate after trauma due to the inflammatory innate immune response to injury, which can lead to neuronal cell death, scarring, and permanently lost neurologic function. Extracellular matrix bioscaffolds, derived by decellularizing healthy tissues, have been widely used in both preclinical and clinical studies to promote positive tissue remodeling, including neurogenesis, in numerous tissues, with extracellular matrix from homologous tissues often inducing more positive responses. Extracellular matrix hydrogels are liquid at room temperature and enable minimally invasive extracellular matrix injections into central nervous system tissues, before gelation at 37℃. However, few studies have analyzed how extracellular matrix hydrogels influence primary central nervous system neuron survival and growth, and whether central nervous system and non-central nervous system extracellular matrix specificity is critical to neuronal responses. Urinary bladder extracellular matrix hydrogels increase both primary hippocampal neuron survival and neurite growth to similar or even greater extents, suggesting extracellular matrix from non-homologous tissue sources, such as urinary bladder matrix-extracellular matrix, may be a more economical and safer alternative to developing central nervous system extracellular matrices for central nervous system applications. Additionally, we show matrix-bound vesicles derived from urinary bladder extracellular matrix are endocytosed by hippocampal neurons and positively regulate primary hippocampal neuron neurite growth. Matrix-bound vesicles carry protein and RNA cargos, including noncoding RNAs and miRNAs that map to the human genome and are known to regulate cellular processes. Thus, urinary bladder matrix-bound vesicles provide natural and transfectable cargoes which offer new experimental tools and therapeutic applications to study and treat central nervous system neuron injury.
Collapse
Affiliation(s)
- Anne Faust
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | - Apoorva Kandakatla
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | - Yolandi van der Merwe
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,3 Swanson School of Engineering, Department of Bioengineering University of Pittsburgh, Pittsburgh, PA, USA
| | - Tanchen Ren
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| | - Luai Huleihel
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - George Hussey
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juan Diego Naranjo
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott Johnson
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen Badylak
- 2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,4 Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Steketee
- 1 Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,2 McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA.,5 Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
100
|
Deletion of the Fractalkine Receptor, CX3CR1, Improves Endogenous Repair, Axon Sprouting, and Synaptogenesis after Spinal Cord Injury in Mice. J Neurosci 2017; 37:3568-3587. [PMID: 28264978 DOI: 10.1523/jneurosci.2841-16.2017] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 01/12/2023] Open
Abstract
Impaired signaling via CX3CR1, the fractalkine receptor, promotes recovery after traumatic spinal contusion injury in mice, a benefit achieved in part by reducing macrophage-mediated injury at the lesion epicenter. Here, we tested the hypothesis that CX3CR1-dependent changes in microglia and macrophage functions also will enhance neuroplasticity, at and several segments below the injury epicenter. New data show that in the presence of inflammatory stimuli, CX3CR1-deficient (CX3CR1-/-) microglia and macrophages adopt a reparative phenotype and increase expression of genes that encode neurotrophic and gliogenic proteins. At the lesion epicenter (mid-thoracic spinal cord), the microenvironment created by CX3CR1-/- microglia/macrophages enhances NG2 cell responses, axon sparing, and sprouting of serotonergic axons. In lumbar spinal cord, inflammatory signaling is reduced in CX3CR1-/- microglia. This is associated with reduced dendritic pathology and improved axonal and synaptic plasticity on ventral horn motor neurons. Together, these data indicate that CX3CR1, a microglia-specific chemokine receptor, is a novel therapeutic target for enhancing neuroplasticity and recovery after SCI. Interventions that specifically target CX3CR1 could reduce the adverse effects of inflammation and augment activity-dependent plasticity and restoration of function. Indeed, limiting CX3CR1-dependent signaling could improve rehabilitation and spinal learning.SIGNIFICANCE STATEMENT Published data show that genetic deletion of CX3CR1, a microglia-specific chemokine receptor, promotes recovery after traumatic spinal cord injury in mice, a benefit achieved in part by reducing macrophage-mediated injury at the lesion epicenter. Data in the current manuscript indicate that CX3CR1 deletion changes microglia and macrophage function, creating a tissue microenvironment that enhances endogenous repair and indices of neuroplasticity, at and several segments below the injury epicenter. Interventions that specifically target CX3CR1 might be used in the future to reduce the adverse effects of intraspinal inflammation and augment activity-dependent plasticity (e.g., rehabilitation) and restoration of function.
Collapse
|