801
|
Singhal G, Morgan J, Jawahar MC, Corrigan F, Jaehne EJ, Toben C, Manavis J, Hannan AJ, Baune BT. Duration of Environmental Enrichment Determines Astrocyte Number and Cervical Lymph Node T Lymphocyte Proportions but Not the Microglial Number in Middle-Aged C57BL/6 Mice. Front Cell Neurosci 2020; 14:57. [PMID: 32256319 PMCID: PMC7094170 DOI: 10.3389/fncel.2020.00057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/27/2020] [Indexed: 11/13/2022] Open
Abstract
Environmental enrichment (EE) has been shown to modulate behavior and immunity. We recently reported that both short and long-term EE enhance baseline locomotion and alleviate depressive-like behavior, but only long-term EE affects locomotion adversely in a threatening environment and enhances anxiety-like behavior in middle-age mice. We have now investigated whether the observed changes in behavior after short- and long-term EE were associated with underlying immune changes. Hence, at the end of behavioral testing, mice were sacrificed, and brains and cervical lymph nodes were collected to investigate the differential effects of the duration of EE (short- and long-term) on the number of immunopositive glial cells in the dentate gyrus, CA1, CA2, and CA3 regions of the hippocampus and proportions of T cell subsets in the cervical lymph nodes using immunohistochemistry and flow cytometry, respectively. EE, regardless of duration, caused an increase in microglia number within the dentate gyrus, CA1 and CA3 hippocampal regions, but only long-term EE increased astrocytes number within the dentate gyrus and CA3 hippocampal regions. A significantly higher proportion of CD8+ naive T cells was observed after long-term EE vs. short-term EE. No significant differences were observed in the proportion of central memory and effector memory T cells or early activated CD25+ cells between any of the test groups. Our results suggest that EE, irrespective of duration, enhances the numbers of microglia, but long-term EE is required to modify astrocyte number and peripheral T cell proportions in middle-aged mice. Our findings provide new insights into the therapeutic effects of EE on various brain disorders, which may be at least partly mediated by glial and neuroimmune modulation.
Collapse
Affiliation(s)
- Gaurav Singhal
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, The University of Adelaide, Adelaide, SA, Australia
| | - Julie Morgan
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, The University of Adelaide, Adelaide, SA, Australia
| | - Magdalene C Jawahar
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, The University of Adelaide, Adelaide, SA, Australia
| | - Frances Corrigan
- Division of Health Sciences, The University of South Australia, Adelaide, SA, Australia
| | - Emily J Jaehne
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, The University of Adelaide, Adelaide, SA, Australia.,School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Catherine Toben
- Psychiatric Neuroscience Lab, Discipline of Psychiatry, The University of Adelaide, Adelaide, SA, Australia
| | - Jim Manavis
- Faculty of Health, Centre for Neurological Diseases, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Anthony J Hannan
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Bernhard T Baune
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.,Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia.,Department of Psychiatry, University of Münster, Münster, Germany
| |
Collapse
|
802
|
Ben-David Y, Kagan S, Cohen Ben-Ami H, Rostami J, Mizrahi T, Kulkarni AR, Thakur GA, Vaknin-Dembinsky A, Healy LM, Brenner T, Treinin M. RIC3, the cholinergic anti-inflammatory pathway, and neuroinflammation. Int Immunopharmacol 2020; 83:106381. [PMID: 32179243 DOI: 10.1016/j.intimp.2020.106381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/19/2020] [Accepted: 03/04/2020] [Indexed: 01/16/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are ligand-gated ion channels having many functions including inflammation control, as part of the cholinergic anti-inflammatory pathway. Genome wide association studies implicated RIC3, a chaperone of nAChRs, in multiple sclerosis (MS), a neuroinflammatory disease. To understand the involvement of RIC3 in inflammatory diseases we examined its expression, regulation, and function in activated immune cells. Our results show that immune activation leads to dynamic changes in RIC3 expression, in a mouse model of MS and in human lymphocytes and macrophages. We also show similarities in the expression dynamics of RIC3 and CHRNA7, encoding for the α7 nAChR subunit. Homomeric α7 nAChRs were shown to mediate the anti-inflammatory effects of cholinergic agonists. Thus, similarity in expression dynamics between RIC3 and CHRNA7 is suggestive of functional concordance. Indeed, siRNA mediated silencing of RIC3 in a mouse macrophage cell line eliminates the anti-inflammatory effects of cholinergic agonists. Furthermore, we show increased average expression of RIC3 and CHRNA7 in lymphocytes from MS patients, and a strong correlation between expression levels of these two genes in MS patients but not in healthy donors. Together, our results are consistent with a role for RIC3 and for the mechanisms regulating its expression in inflammatory processes and in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Yael Ben-David
- Medical Neurobiology, Hadassah Medical School, Hebrew University, Jerusalem, Israel
| | - Sara Kagan
- Medical Neurobiology, Hadassah Medical School, Hebrew University, Jerusalem, Israel
| | - Hagit Cohen Ben-Ami
- Medical Neurobiology, Hadassah Medical School, Hebrew University, Jerusalem, Israel
| | - Jinar Rostami
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden
| | - Tehila Mizrahi
- Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Abhijit R Kulkarni
- Pharmaceutical Science, Bouve College of Health Science, Northeastern University, Boston, USA
| | - Ganesh A Thakur
- Pharmaceutical Science, Bouve College of Health Science, Northeastern University, Boston, USA
| | - Adi Vaknin-Dembinsky
- Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Luke M Healy
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Talma Brenner
- Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Millet Treinin
- Medical Neurobiology, Hadassah Medical School, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
803
|
Pestana F, Edwards-Faret G, Belgard TG, Martirosyan A, Holt MG. No Longer Underappreciated: The Emerging Concept of Astrocyte Heterogeneity in Neuroscience. Brain Sci 2020; 10:brainsci10030168. [PMID: 32183137 PMCID: PMC7139801 DOI: 10.3390/brainsci10030168] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 12/18/2022] Open
Abstract
Astrocytes are ubiquitous in the central nervous system (CNS). These cells possess thousands of individual processes, which extend out into the neuropil, interacting with neurons, other glia and blood vessels. Paralleling the wide diversity of their interactions, astrocytes have been reported to play key roles in supporting CNS structure, metabolism, blood-brain-barrier formation and control of vascular blood flow, axon guidance, synapse formation and modulation of synaptic transmission. Traditionally, astrocytes have been studied as a homogenous group of cells. However, recent studies have uncovered a surprising degree of heterogeneity in their development and function, in both the healthy and diseased brain. A better understanding of astrocyte heterogeneity is urgently needed to understand normal brain function, as well as the role of astrocytes in response to injury and disease.
Collapse
Affiliation(s)
- Francisco Pestana
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
| | - Gabriela Edwards-Faret
- Laboratory of Neuronal Wiring, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
- Life & Medical Sciences (LIMES) Institute, Faculty of Mathematics and Natural Sciences, University of Bonn, 53115 Bonn, Germany
| | | | - Araks Martirosyan
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
- Correspondence: (A.M.); (M.G.H.); Tel.: +32-16-37-31-27 (M.G.H.)
| | - Matthew G. Holt
- Laboratory of Glia Biology, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium;
- Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
- Correspondence: (A.M.); (M.G.H.); Tel.: +32-16-37-31-27 (M.G.H.)
| |
Collapse
|
804
|
Gong Z, Wang C, Ni L, Ying L, Shu J, Wang J, Yu C, Xia K, Cheng F, Shi K, Xu G, Yu Q, Shen J, Chen Q, Li F, Liang C. An injectable recombinant human milk fat globule-epidermal growth factor 8-loaded copolymer system for spinal cord injury reduces inflammation through NF-κB and neuronal cell death. Cytotherapy 2020; 22:193-203. [PMID: 32173261 DOI: 10.1016/j.jcyt.2020.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/19/2020] [Accepted: 01/30/2020] [Indexed: 01/17/2023]
Abstract
Spinal cord injury (SCI) is a common disease and a major cause of paralysis, carrying much burden around the world. Despite the progress made with growth factors therapy, the response rate of acute SCI treatment still remains unsatisfactory, due largely to complex and severe inflammatory reactions. Herein, we prepare a MFG-E8-loaded copolymer system-based anti-inflammation therapy for SCI treatment. It is shown that the MFG-E8-loaded copolymer system can decrease pro-inflammatory cytokine expression and neuron death. In a rat model of crush-caused SCI, the copolymer system shows significant therapeutic efficacy by ameliorating inflammation, decreasing fibrotic scar, promoting myelin regeneration and suppressing overall SCI severity.
Collapse
Affiliation(s)
- Zhe Gong
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Chenggui Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Licheng Ni
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Liwei Ying
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jiawei Shu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jingkai Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Chao Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Feng Cheng
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Kesi Shi
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Guoping Xu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | - Qunfei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | | | - Qixin Chen
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | - Fangcai Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
805
|
Zhang X, Guo X, Kang X, Yang H, Guo W, Guan L, Wu H, Du L. Surface Functionalization of Pegylated Gold Nanoparticles with Antioxidants Suppresses Nanoparticle-Induced Oxidative Stress and Neurotoxicity. Chem Res Toxicol 2020; 33:1195-1205. [PMID: 32125152 DOI: 10.1021/acs.chemrestox.9b00368] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Because of their biocompatibility and biosafety, pegylated Au NPs (Au@PEG), as a nanodrug-carrier, have been widely applied in different biomedical applications, including imaging and drug delivery systems. Under such conditions, the biosafety of Au@PEG has attracted tremendous attention. However, only a small number of studies focused on the neurotoxicity of Au@PEG used as drug delivery carriers not to mention reducing the neurotoxicity of Au@PEG. To address this issue, the adverse effects of Au@PEG on human neuroblastoma SHSY5Y cells were first investigated. The results showed that 4.5 nm Au@PEG significantly induced cell apoptosis through upregulating reactive oxygen species (ROS) production and disordering the mitochondrial membrane potential. To further evaluate whether the neurotoxicity of Au@PEG could be improved through conjugating antioxidants on the surface of Au@PEG, Trolox (a vitamin E analogue)-functionalized Au@PEG (Au@Trolox) was synthesized. The results showed that the neurotoxicity of Au@PEG on SHSY5Y cells could be significantly improved by Au@Trolox. Next, mice were subjected to administration of 4.5 nm Au@PEG and Au@Trolox for 3 months. An increase of oxidative stress and a decrease in the activity of key antioxidant enzymes including glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), and catalase (CAT) were observed after long-term injection of Au@PEG. More importantly, both the apoptosis of neurons and the activation of astrocytes were observed in the hippocampus of mice injected with Au@PEG. In contrast, the adverse effects of Au@PEG could be improved when injected with Au@Trolox. In short, the present study provided new insights into the toxicity evaluation of nanoparticles and would help to better understand and prevent the neurotoxicity of nanomaterials used in pharmaceutics.
Collapse
Affiliation(s)
- Xiaojie Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Xueling Guo
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Xiaoxuan Kang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.,Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei 050024, P. R. China
| | - Hui Yang
- Immunology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P. R. China
| | - Weiyi Guo
- College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Lingmei Guan
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Hai Wu
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Libo Du
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Center for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| |
Collapse
|
806
|
Serapide MF, L’Episcopo F, Tirolo C, Testa N, Caniglia S, Giachino C, Marchetti B. Boosting Antioxidant Self-defenses by Grafting Astrocytes Rejuvenates the Aged Microenvironment and Mitigates Nigrostriatal Toxicity in Parkinsonian Brain via an Nrf2-Driven Wnt/β-Catenin Prosurvival Axis. Front Aging Neurosci 2020; 12:24. [PMID: 32226376 PMCID: PMC7081734 DOI: 10.3389/fnagi.2020.00024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/22/2020] [Indexed: 12/19/2022] Open
Abstract
Astrocyte (As) bidirectional dialog with neurons plays a fundamental role in major homeostatic brain functions, particularly providing metabolic support and antioxidant self-defense against reactive oxygen (ROS) and nitrogen species (RNS) via the activation of NF-E2-related factor 2 (Nrf2), a master regulator of oxidative stress. Disruption of As-neuron crosstalk is chiefly involved in neuronal degeneration observed in Parkinson's disease (PD), the most common movement disorder characterized by the selective degeneration of dopaminergic (DAergic) cell bodies of the substantia nigra (SN) pars compacta (SNpc). Ventral midbrain (VM)-As are recognized to exert an important role in DAergic neuroprotection via the expression of a variety of factors, including wingless-related MMTV integration site 1 (Wnt1), a principal player in DAergic neurogenesis. However, whether As, by themselves, might fulfill the role of chief players in DAergic neurorestoration of aged PD mice is presently unresolved. Here, we used primary postnatal mouse VM-As as a graft source for unilateral transplantation above the SN of aged 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mice after the onset of motor symptoms. Spatio-temporal analyses documented that the engrafted cells promoted: (i) a time-dependent nigrostriatal rescue along with increased high-affinity synaptosomal DA uptake and counteraction of motor deficit, as compared to mock-grafted counterparts; and (ii) a restoration of the impaired microenvironment via upregulation of As antioxidant self-defense through the activation of Nrf2/Wnt/β-catenin signaling, suggesting that grafting As has the potential to switch the SN neurorescue-unfriendly environment to a beneficial antioxidant/anti-inflammatory prosurvival milieu. These findings highlight As-derived factors/mechanisms as the crucial key for successful therapeutic outcomes in PD.
Collapse
Affiliation(s)
- Maria Francesca Serapide
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, Medical School, University of Catania, Catania, Italy
| | | | - Cataldo Tirolo
- Section of Neuropharmacology, OASI Research Institute-IRCCS, Troina, Italy
| | - Nunzio Testa
- Section of Neuropharmacology, OASI Research Institute-IRCCS, Troina, Italy
| | - Salvatore Caniglia
- Section of Neuropharmacology, OASI Research Institute-IRCCS, Troina, Italy
| | - Carmela Giachino
- Section of Neuropharmacology, OASI Research Institute-IRCCS, Troina, Italy
| | - Bianca Marchetti
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, Medical School, University of Catania, Catania, Italy
- Section of Neuropharmacology, OASI Research Institute-IRCCS, Troina, Italy
| |
Collapse
|
807
|
Huang L, Wang Y, Zhu M, Wan X, Zhang H, Lei T, Blesch A, Liu S. Anisotropic Alginate Hydrogels Promote Axonal Growth across Chronic Spinal Cord Transections after Scar Removal. ACS Biomater Sci Eng 2020; 6:2274-2286. [PMID: 33455324 DOI: 10.1021/acsbiomaterials.9b01802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have previously reported that cell-seeded alginate hydrogels (AHs) with anisotropic capillaries can restore the continuity of the spinal cord and support axonal regeneration in a rat model of acute partial spinal cord transection. Whether similar effects can be found after transplantation into sites of complete chronic spinal cord transections without additional growth-promoting stimuli has not been investigated. We therefore implanted AHs into the cavity of a chronic thoracic transection following scar resection (SR) 4 weeks postinjury and examined electrophysiological and functional recovery as well as regeneration of descending and ascending projections within and beyond the AH scaffold up to 3 months after engraftment. Our results indicate that both electrophysiological conductivity and locomotor function are significantly improved after AH engraftment. SR transiently impairs locomotor function immediately after surgery but does not affect long-term outcomes. Histological analysis shows numerous host cells migrating into the scaffold channels and a reduction of fibroglial scaring around the lesion by AH grafts. In contrast to corticospinal axons, raphaespinal and propriospinal descending axons and ascending sensory axons regenerate throughout the scaffolds and extend into the distal host parenchyma. These results further support the pro-regenerative properties of AHs and their therapeutic potential for chronic SCI in combination with other strategies to improve functional outcomes after spinal cord injury.
Collapse
Affiliation(s)
- Lulu Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei 430030, P.R. China
| | - Yu Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei 430030, P.R. China
| | - Mingxin Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei 430030, P.R. China
| | - Xueyan Wan
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei 430030, P.R. China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei 430030, P.R. China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei 430030, P.R. China
| | - Armin Blesch
- Department of Neurosciences, Center for Neural Repair, University of California, San Diego, Biomedical Research Facility 2, Room 2131, 9500 Gilman Drive, La Jolla, California 92093-0626, United States
| | - Shengwen Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
808
|
Pericyte-Mediated Tissue Repair through PDGFRβ Promotes Peri-Infarct Astrogliosis, Oligodendrogenesis, and Functional Recovery after Acute Ischemic Stroke. eNeuro 2020; 7:ENEURO.0474-19.2020. [PMID: 32046974 PMCID: PMC7070447 DOI: 10.1523/eneuro.0474-19.2020] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Post-stroke functional recovery can occur spontaneously during the subacute phase; however, how post-stroke fibrotic repair affects functional recovery is highly debated. Platelet-derived growth factor receptor β (PDGFRβ)-expressing pericytes are responsible for post-stroke fibrotic repair within infarct areas; therefore, we examined peri-infarct neural reorganization and functional recovery after permanent middle cerebral artery occlusion (pMCAO) using pericyte-deficient Pdgfrb+/- mice. Time-dependent reduction of infarct area sizes, i.e., repair, was significantly impaired in Pdgfrb+/- mice with recovery of cerebral blood flow (CBF) in ischemic areas attenuated by defective leptomeningeal arteriogenesis and intrainfarct angiogenesis. Peri-infarct astrogliosis, accompanied by increased STAT3 phosphorylation, was attenuated in Pdgfrb+/- mice. Pericyte-conditioned medium (PCM), particularly when treated with platelet-derived growth factor subunit B (PDGFB) homodimer (PDGF-BB; PCM/PDGF-BB), activated STAT3 and enhanced the proliferation and activity of cultured astrocytes. Although peri-infarct proliferation of oligodendrocyte (OL) precursor cells (OPCs) was induced promptly after pMCAO regardless of intrainfarct repair, OPC differentiation and remyelination were significantly attenuated in Pdgfrb+/- mice. Consistently, astrocyte-CM (ACM) promoted OPC differentiation and myelination, which were enhanced remarkably by adding PCM/PDGF-BB to the medium. Post-stroke functional recovery correlated well with the extent and process of intrainfarct repair and peri-infarct oligodendrogenesis. Overall, pericyte-mediated intrainfarct fibrotic repair through PDGFRβ may promote functional recovery through enhancement of peri-infarct oligodendrogenesis as well as astrogliosis after acute ischemic stroke.
Collapse
|
809
|
Buzoianu-Anguiano V, Rivera-Osorio J, Orozco-Suárez S, Vega-García A, García-Vences E, Sánchez-Torres S, Jiménez-Estrada I, Guizar-Sahagún G, Mondragon-Caso J, Fernández-Valverde F, Madrazo I, Grijalva I. Single vs. Combined Therapeutic Approaches in Rats With Chronic Spinal Cord Injury. Front Neurol 2020; 11:136. [PMID: 32210903 PMCID: PMC7076126 DOI: 10.3389/fneur.2020.00136] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/06/2020] [Indexed: 12/30/2022] Open
Abstract
The regenerative capability of the central nervous system is limited after traumatic spinal cord injury (SCI) due to intrinsic and extrinsic factors that inhibit spinal cord regeneration, resulting in deficient functional recovery. It has been shown that strategies, such as pre-degenerated peripheral nerve (PPN) grafts or the use of bone marrow stromal cells (BMSCs) or exogenous molecules, such as chondroitinase ABC (ChABC) promote axonal growth and remyelination, resulting in an improvement in locomotor function. These treatments have been primarily assessed in acute injury models. The aim of the present study is to evaluate the ability of several single and combined treatments in order to modify the course of chronic complete SCI in rats. A complete cord transection was performed at the T9 level. One month later, animals were divided into five groups: original injury only (control group), and original injury plus spinal cord re-transection to create a gap to accommodate BMSCs, PPN, PPN + BMSCs, and PPN + BMSCs + ChABC. In comparison with control and single-treatment groups (PPN and BMSCs), combined treatment groups (PPN + BMSCs and PPN + BMSCs + ChABC) showed significative axonal regrowth, as revealed by an increase in GAP-43 and MAP-1B expression in axonal fibers, which correlated with an improvement in locomotor function. In conclusion, the combined therapies tested here improve locomotor function by enhancing axonal regeneration in rats with chronic SCI. Further studies are warranted to refine this promising line of research for clinical purposes.
Collapse
Affiliation(s)
- Vinnitsa Buzoianu-Anguiano
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico
| | - Jared Rivera-Osorio
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico
| | - Sandra Orozco-Suárez
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico
| | - Angélica Vega-García
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico
| | - Elisa García-Vences
- Centro de Investigación en Ciencias de la Salud, Universidad Anahuac México Campus Norte, Mexico City, Mexico
| | - Stephanie Sánchez-Torres
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico
| | - Ismael Jiménez-Estrada
- Departamento de Fisiología, Biofísica y Neurociencias, CINVESTAV, IPN, Mexico City, Mexico
| | - Gabriel Guizar-Sahagún
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico.,Departamento de Cirugía Experimental, Proyecto Camina AC, Mexico City, Mexico
| | - Jose Mondragon-Caso
- Centro de Investigación en Ciencias de la Salud, Universidad Anahuac México Campus Norte, Mexico City, Mexico
| | | | - Ignacio Madrazo
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico
| | - Israel Grijalva
- Hospital de Especialidades CMN Siglo XXI IMSS, Unidad de Investigación Médica en Enfermedades Neurologicas, Mexico City, Mexico
| |
Collapse
|
810
|
Liu XY, Guo JW, Kou JQ, Sun YL, Zheng XJ. Repair mechanism of astrocytes and non-astrocytes in spinal cord injury. World J Clin Cases 2020; 8:854-863. [PMID: 32190622 PMCID: PMC7062612 DOI: 10.12998/wjcc.v8.i5.854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/30/2019] [Accepted: 02/09/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a destructive disease that incurs huge personal and social costs, and there is no effective treatment. Although the pathogenesis and treatment mechanism of SCI has always been a strong scientific focus, the pathogenesis of SCI is still under investigation.
AIM To determine the key genes based on the modularization of in-depth analysis, in order to identify the repair mechanism of astrocytes and non-astrocytes in SCI.
METHODS Firstly, the differences between injured and non-injured spinal cord of astrocyte (HA), injured and non-injured spinal cord of non-astrocyte (FLOW), injured spinal cord of non-injured astrocyte (HA) and non-injured spinal cord of non-astrocyte (FLOW), and non-injured spinal cord of astrocyte (HA) and non-astrocyte (FLOW) were analyzed. The total number of differentially expressed genes was obtained by merging the four groups of differential results. Secondly, the genes were co-expressed and clustered. Then, the enrichment of GO function and KEGG pathway of module genes was analyzed. Finally, non-coding RNA, transcription factors and drugs that regulate module genes were predicted using hypergeometric tests.
RESULTS In summary, we obtained 19 expression modules involving 5216 differentially expressed genes. Among them, miR-494, XIST and other genes were differentially expressed in SCI patients, and played an active regulatory role in dysfunction module, and these genes were recognized as the driving genes of SCI. Enrichment results showed that module genes were significantly involved in the biological processes of inflammation, oxidation and apoptosis. Signal pathways such as NF-kappa B/A20, AMPK and MAPK were significantly regulated. In addition, non-coding RNA pivot (including miR-136-5p and let-7d-5p, etc.) and transcription factor pivot (including NFKB1, MYC, etc.) were identified as significant regulatory dysfunction modules.
CONCLUSION Overall, this study uncovered a co-expression network of key genes involved in astrocyte and non-astrocyte regulation in SCI. These findings helped to reveal the core dysfunction modules, potential regulatory factors and driving genes of the disease, and to improve our understanding of its pathogenesis.
Collapse
Affiliation(s)
- Xiang-Yun Liu
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jian-Wei Guo
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jian-Qiang Kou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Yuan-Liang Sun
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Xiu-Jun Zheng
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| |
Collapse
|
811
|
Sardari M, Dzyubenko E, Schmermund B, Yin D, Qi Y, Kleinschnitz C, Hermann DM. Dose-Dependent Microglial and Astrocytic Responses Associated With Post-ischemic Neuroprotection After Lipopolysaccharide-Induced Sepsis-Like State in Mice. Front Cell Neurosci 2020; 14:26. [PMID: 32116567 PMCID: PMC7029732 DOI: 10.3389/fncel.2020.00026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/29/2020] [Indexed: 12/25/2022] Open
Abstract
In contrast to lipopolysaccharide (LPS)-induced preconditioning, which has repeatedly been examined in the past, the effects of post-ischemic LPS-induced sepsis, although clinically considerably more important, have not systemically been studied. We exposed mice to transient intraluminal middle cerebral artery occlusion (MCAO) and examined the effects of intraperitoneal LPS (0.1 or 1 mg/kg) which was administered 24 h post-ischemia. Post-ischemic glial reactivity, neuronal survival and neurological outcome were differently modulated by the higher and the lower LPS dose. Although both doses promoted neuronal survival after 72 h, the underlying mechanisms were not similar. Mice receiving 1 mg/kg LPS exhibited transient hypothermia at 1 and 3 hours post sepsis (hps), followed by reduced focal neurological deficits at 24, 48 and 72 hps. The lower dose (0.1 mg/kg) did not induce hypothermia, but reduced microglia/macrophage activation with the appearance of an anti-inflammatory CD206 positive cell phenotype in the brain parenchyma. Together, our results indicate a novel, dose-dependent modulation of microglial cells that is intricately involved in brain protection.
Collapse
Affiliation(s)
- Maryam Sardari
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Egor Dzyubenko
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Ben Schmermund
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Dongpei Yin
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Yachao Qi
- Department of Neurology, University Hospital Essen, Essen, Germany
| | | | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany
| |
Collapse
|
812
|
Chen C, Zhong X, Smith DK, Tai W, Yang J, Zou Y, Wang LL, Sun J, Qin S, Zhang CL. Astrocyte-Specific Deletion of Sox2 Promotes Functional Recovery After Traumatic Brain Injury. Cereb Cortex 2020; 29:54-69. [PMID: 29161339 DOI: 10.1093/cercor/bhx303] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/19/2017] [Indexed: 12/19/2022] Open
Abstract
Injury to the adult brain induces activation of local astrocytes, which serves as a compensatory response that modulates tissue damage and recovery. However, the mechanism governing astrocyte activation during brain injury remains largely unknown. Here we provide in vivo evidence that SOX2, a transcription factor critical for stem cells and brain development, is also required for injury-induced activation of adult cortical astrocytes. Genome-wide chromatin immunoprecipitation-seq analysis of mouse cortical tissues reveals that SOX2 binds to regulatory regions of genes associated with signaling pathways that control glial cell activation, such as Nr2e1, Mmd2, Wnt7a, and Akt2. Astrocyte-specific deletion of Sox2 in adult mice greatly diminishes glial response to controlled cortical impact injury and, most unexpectedly, dampens injury-induced cortical loss and benefits behavioral recovery of mice after injury. Together, these results uncover an essential role of SOX2 in somatic cells under pathological conditions and indicate that SOX2-dependent astrocyte activation could be targeted for functional recovery after traumatic brain injury.
Collapse
Affiliation(s)
- Chunhai Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Xiaoling Zhong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Derek K Smith
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Jianjing Yang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Lei-Lei Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Jiahong Sun
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Song Qin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Center of Neural Injury and Repair, Shanghai Tenth People's Hospital Affiliated with Tongji University, Shanghai, China
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| |
Collapse
|
813
|
Han Q, Xie Y, Ordaz JD, Huh AJ, Huang N, Wu W, Liu N, Chamberlain KA, Sheng ZH, Xu XM. Restoring Cellular Energetics Promotes Axonal Regeneration and Functional Recovery after Spinal Cord Injury. Cell Metab 2020; 31:623-641.e8. [PMID: 32130884 PMCID: PMC7188478 DOI: 10.1016/j.cmet.2020.02.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 11/24/2019] [Accepted: 01/31/2020] [Indexed: 01/30/2023]
Abstract
Axonal regeneration in the central nervous system (CNS) is a highly energy-demanding process. Extrinsic insults and intrinsic restrictions lead to an energy crisis in injured axons, raising the question of whether recovering energy deficits facilitates regeneration. Here, we reveal that enhancing axonal mitochondrial transport by deleting syntaphilin (Snph) recovers injury-induced mitochondrial depolarization. Using three CNS injury mouse models, we demonstrate that Snph-/- mice display enhanced corticospinal tract (CST) regeneration passing through a spinal cord lesion, accelerated regrowth of monoaminergic axons across a transection gap, and increased compensatory sprouting of uninjured CST. Notably, regenerated CST axons form functional synapses and promote motor functional recovery. Administration of the bioenergetic compound creatine boosts CST regenerative capacity in Snph-/- mice. Our study provides mechanistic insights into intrinsic regeneration failure in CNS and suggests that enhancing mitochondrial transport and cellular energetics are promising strategies to promote regeneration and functional restoration after CNS injuries.
Collapse
Affiliation(s)
- Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuxiang Xie
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Josue D Ordaz
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew J Huh
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ning Huang
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Naikui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kelly A Chamberlain
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
814
|
Warnock A, Toomey LM, Wright AJ, Fisher K, Won Y, Anyaegbu C, Fitzgerald M. Damage Mechanisms to Oligodendrocytes and White Matter in Central Nervous System Injury: The Australian Context. J Neurotrauma 2020; 37:739-769. [DOI: 10.1089/neu.2019.6890] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Andrew Warnock
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Lillian M. Toomey
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Alexander J. Wright
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Katherine Fisher
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Yerim Won
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Chidozie Anyaegbu
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| |
Collapse
|
815
|
Li C, Zhu X, Lee CM, Wu Z, Cheng L. A mouse model of complete-crush transection spinal cord injury made by two operations. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:210. [PMID: 32309357 PMCID: PMC7154420 DOI: 10.21037/atm.2020.01.58] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background More and more studies have focused on the treatment of spinal cord injury (SCI) by tissue engineering, but there is still no ideal animal model that can genuinely and objectively simulate the real pathological process in clinical practice. Also, given the increasing availability and use of genetically modified animals in basic science research, it has become essential to develop clinically related models for SCI for use in mice. Methods Forty-eight C57BL/6 mice were divided into three groups (injured/sham/uninjured). We determined the scar range made by the first crush injury by specimen observation, hematoxylin and eosin (HE) staining, and immunofluorescence staining. Transection to completely remove a 2-mm spinal cord segment centered on the lesion core was completed 6 weeks after the first injury in injured groups, whereas the sham group only underwent re-exposure of the spinal cord without transection injury. The characteristics of this SCI model were fully ascertained by specimen observation, HE staining, immunofluorescence staining, and quantitative real-time polymerase chain reaction (qRT-PCR). Results No mice died after the first injury. Histopathological findings suggested a scar range of 2 mm. After the second operation, 2 mice of the injured group and 1 mouse of the sham group died. The Basso Mouse Scale (BMS) score and motor evoked potential (MEP) results showed that the neurological function of mice did not recover. Immunostaining showed that there were no neurons or neurofilament residues in the lesion core 4 weeks after the second injury. Astrocytes encapsulated immune cells to form dense glial scars. Most immune cells were confined to the core of the lesion and formed fibrous scars with the fibroblasts. At the same time, there was considerable angiogenesis in the lesion core and around the injury. The results of qRT-PCR showed that Ptprc was highly expressed in the lesion core, while Gfap, nestin, Cnp, and Sv2b were highly expressed in the adjacent region. This suggests that the lesion core is a highly inflammatory zone, but there may be spontaneous neurogenesis adjacent to the lesion core. Conclusions The mouse crash-complete transection SCI model made by the two operations has good simulation, high feasibility, and high reproducibility; it will be a useful tool for pre-clinical testing of SCI treatment.
Collapse
Affiliation(s)
- Chen Li
- Division of Spine Surgery, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration, Tongji University, Ministry of Education, Shanghai 200065, China.,Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Xingfei Zhu
- Division of Spine Surgery, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration, Tongji University, Ministry of Education, Shanghai 200065, China
| | - Chia-Ming Lee
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Zhourui Wu
- Division of Spine Surgery, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration, Tongji University, Ministry of Education, Shanghai 200065, China
| | - Liming Cheng
- Division of Spine Surgery, Department of Orthopaedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration, Tongji University, Ministry of Education, Shanghai 200065, China
| |
Collapse
|
816
|
Zhou X, Wahane S, Friedl MS, Kluge M, Friedel CC, Avrampou K, Zachariou V, Guo L, Zhang B, He X, Friedel RH, Zou H. Microglia and macrophages promote corralling, wound compaction and recovery after spinal cord injury via Plexin-B2. Nat Neurosci 2020; 23:337-350. [PMID: 32112058 PMCID: PMC7412870 DOI: 10.1038/s41593-020-0597-7] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tissue repair after spinal cord injury requires the mobilization of immune and glial cells to form a protective barrier that seals the wound and facilitates debris clearing, inflammatory containment and matrix compaction. This process involves corralling, wherein phagocytic immune cells become confined to the necrotic core, which is surrounded by an astrocytic border. Here we elucidate a temporally distinct gene signature in injury-activated microglia and macrophages (IAMs) that engages axon guidance pathways. Plexin-B2 is upregulated in IAMs and is required for motor sensory recovery after spinal cord injury. Plexin-B2 deletion in myeloid cells impairs corralling, leading to diffuse tissue damage, inflammatory spillover and hampered axon regeneration. Corralling begins early and requires Plexin-B2 in both microglia and macrophages. Mechanistically, Plexin-B2 promotes microglia motility, steers IAMs away from colliding cells and facilitates matrix compaction. Our data therefore establish Plexin-B2 as an important link that integrates biochemical cues and physical interactions of IAMs with the injury microenvironment during wound healing.
Collapse
Affiliation(s)
- Xiang Zhou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shalaka Wahane
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marie-Sophie Friedl
- Institut für Informatik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Kluge
- Institut für Informatik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Caroline C Friedel
- Institut für Informatik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kleopatra Avrampou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lei Guo
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xijing He
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an International Medical Center, Xi'an, China
| | - Roland H Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neurosurgery, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neurosurgery, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
817
|
Miyamoto N, Magami S, Inaba T, Ueno Y, Hira K, Kijima C, Nakajima S, Yamashiro K, Urabe T, Hattori N. The effects of A1/A2 astrocytes on oligodendrocyte linage cells against white matter injury under prolonged cerebral hypoperfusion. Glia 2020; 68:1910-1924. [DOI: 10.1002/glia.23814] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Nobukazu Miyamoto
- Department of NeurologyJuntendo University School of Medicine Tokyo Japan
| | - Shunsuke Magami
- Department of NeurosurgeryJuntendo University School of Medicine Tokyo Japan
| | - Toshiki Inaba
- Department of NeurologyJuntendo University Urayasu Hospital Chiba Japan
| | - Yuji Ueno
- Department of NeurologyJuntendo University School of Medicine Tokyo Japan
| | - Kenichiro Hira
- Department of NeurologyJuntendo University School of Medicine Tokyo Japan
| | - Chikage Kijima
- Department of NeurologyJuntendo University School of Medicine Tokyo Japan
| | - Sho Nakajima
- Department of NeurologyJuntendo University School of Medicine Tokyo Japan
| | - Kazuo Yamashiro
- Department of NeurologyJuntendo University School of Medicine Tokyo Japan
| | - Takao Urabe
- Department of NeurologyJuntendo University Urayasu Hospital Chiba Japan
| | - Nobutaka Hattori
- Department of NeurologyJuntendo University School of Medicine Tokyo Japan
| |
Collapse
|
818
|
Zhong K, Wang RX, Qian XD, Yu P, Zhu XY, Zhang Q, Ye YL. Neuroprotective effects of saffron on the late cerebral ischemia injury through inhibiting astrogliosis and glial scar formation in rats. Biomed Pharmacother 2020; 126:110041. [PMID: 32113053 DOI: 10.1016/j.biopha.2020.110041] [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: 08/28/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
This study is to explore the neuroprotective effects and involved glial scar of saffron (Crocus sativus L.) on the late cerebral ischemia in rats. Focal cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in Sprague Dawley rats that were randomly divided into sham group, MCAO group, edaravone group (as a positive control) and saffron groups (saffron extract 30, 100, 300 mg/kg). Saffron was administered orally at 2 h at the first day and once daily from day 2 to 42 after ischemia. Behavioral changes were detected from day 43 to 46 after ischemia to evaluate the effects of saffron. Infarct volume, survival neuron density, activated astrocyte, and the thickness of glial scar were also detected. GFAP, neurocan, phosphocan, neurofilament expressions and inflammatory cytokine contents were detected by Western-blotting and ELISA methods, respectively. Saffron improved the body weight loss, neurological deficit and spontaneous activity. It also ameliorated anxiety-like state and cognitive dysfunction, which were detected by elevated plus maze (EPM), marble burying test (MBT) and novel object recognition test (NORT). Toluidine blue staining found that saffron treatment decreased the infarct volume and increased the neuron density in cortex in the ischemic boundary zone. The activated astrocyte number and the thickness of glial scar in the penumbra zone reduced after saffron treatment. Additionally, saffron decreased the contents of IL-6 and IL-1β, increased the content of IL-10 in the ischemic boundary zone. GFAP, neurocan, and phosphocan expressions in ischemic boundary zone and ischemic core zone all decreased after saffron treatment. Saffron exerted neuroprotective effects on late cerebral ischemia, associating with attenuating astrogliosis and glial scar formation after ischemic injury.
Collapse
Affiliation(s)
- Kai Zhong
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Rou-Xin Wang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | - Ping Yu
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xin-Ying Zhu
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qi Zhang
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yi-Lu Ye
- School of Basic Medical Sciences & Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
819
|
Santiago AR, Madeira MH, Boia R, Aires ID, Rodrigues-Neves AC, Santos PF, Ambrósio AF. Keep an eye on adenosine: Its role in retinal inflammation. Pharmacol Ther 2020; 210:107513. [PMID: 32109489 DOI: 10.1016/j.pharmthera.2020.107513] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adenosine is an endogenous purine nucleoside ubiquitously distributed throughout the body that interacts with G protein-coupled receptors, classified in four subtypes: A1R, A2AR, A2BR and A3R. Among the plethora of functions of adenosine, it has been increasingly recognized as a key mediator of the immune response. Neuroinflammation is a feature of chronic neurodegenerative diseases and contributes to the pathophysiology of several retinal degenerative diseases. Animal models of retinal diseases are helping to elucidate the regulatory roles of adenosine receptors in the development and progression of those diseases. Mounting evidence demonstrates that the adenosinergic system is altered in the retina during pathological conditions, compromising retinal physiology. This review focuses on the roles played by adenosine and the elements of the adenosinergic system (receptors, enzymes, transporters) in the neuroinflammatory processes occurring in the retina. An improved understanding of the molecular and cellular mechanisms of the signalling pathways mediated by adenosine underlying the onset and progression of retinal diseases will pave the way towards the identification of new therapeutic approaches.
Collapse
Affiliation(s)
- Ana Raquel Santiago
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548 Coimbra, Portugal.
| | - Maria H Madeira
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548 Coimbra, Portugal
| | - Raquel Boia
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Inês Dinis Aires
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Catarina Rodrigues-Neves
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Paulo Fernando Santos
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - António Francisco Ambrósio
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548 Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
820
|
Fang J, Hsueh YY, Soto J, Sun W, Wang J, Gu Z, Khademhosseini A, Li S. Engineering Biomaterials with Micro/Nanotechnologies for Cell Reprogramming. ACS NANO 2020; 14:1296-1318. [PMID: 32011856 PMCID: PMC10067273 DOI: 10.1021/acsnano.9b04837] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cell reprogramming is a revolutionized biotechnology that offers a powerful tool to engineer cell fate and function for regenerative medicine, disease modeling, drug discovery, and beyond. Leveraging advances in biomaterials and micro/nanotechnologies can enhance the reprogramming performance in vitro and in vivo through the development of delivery strategies and the control of biophysical and biochemical cues. In this review, we present an overview of the state-of-the-art technologies for cell reprogramming and highlight the recent breakthroughs in engineering biomaterials with micro/nanotechnologies to improve reprogramming efficiency and quality. Finally, we discuss future directions and challenges for reprogramming technologies and clinical translation.
Collapse
Affiliation(s)
- Jun Fang
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Yuan-Yu Hsueh
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Division of Plastic Surgery, Department of Surgery, College of Medicine , National Cheng Kung University Hospital , Tainan 70456 , Taiwan
| | - Jennifer Soto
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Wujin Sun
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
| | - Jinqiang Wang
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
| | - Zhen Gu
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
- Jonsson Comprehensive Cancer Center , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Ali Khademhosseini
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
- Department of Chemical and Biomolecular Engineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Radiology , University of California, Los Angeles , Los Angeles , California 90095 , United States
| | - Song Li
- Department of Bioengineering , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Department of Medicine , University of California, Los Angeles , Los Angeles , California 90095 , United States
- Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute , University of California, Los Angeles , Los Angles , California 90095 , United States
| |
Collapse
|
821
|
Lindsay SL, McCanney GA, Willison AG, Barnett SC. Multi-target approaches to CNS repair: olfactory mucosa-derived cells and heparan sulfates. Nat Rev Neurol 2020; 16:229-240. [PMID: 32099190 DOI: 10.1038/s41582-020-0311-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.
Collapse
Affiliation(s)
- Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - George A McCanney
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alice G Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
822
|
Chang CY, Liang MZ, Wu CC, Huang PY, Chen HI, Yet SF, Tsai JW, Kao CF, Chen L. WNT3A Promotes Neuronal Regeneration upon Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21041463. [PMID: 32098078 PMCID: PMC7073099 DOI: 10.3390/ijms21041463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 01/12/2023] Open
Abstract
The treatment of traumatic brain injury (TBI) remains a challenge due to limited knowledge about the mechanisms underlying neuronal regeneration. This current study compared the expression of WNT genes during regeneration of injured cortical neurons. Recombinant WNT3A showed positive effect in promoting neuronal regeneration via in vitro, ex vivo, and in vivo TBI models. Intranasal administration of WNT3A protein to TBI mice increased the number of NeuN+ neurons without affecting GFAP+ glial cells, compared to control mice, as well as retained motor function based on functional behavior analysis. Our findings demonstrated that WNT3A, 8A, 9B, and 10A promote regeneration of injured cortical neurons. Among these WNTs, WNT3A showed the most promising regenerative potential in vivo, ex vivo, and in vitro.
Collapse
Affiliation(s)
- Chu-Yuan Chang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Min-Zong Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Ching-Chih Wu
- Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Pei-Yuan Huang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Hong-I Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan 35053, Taiwan;
| | - Jin-Wu Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Cheng-Fu Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11574, Taiwan
- Correspondence: (C.-F.K.); (L.C.); Tel.: +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (C.-Y.C.); (M.-Z.L.); (P.-Y.H.); (H.-I.C.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: (C.-F.K.); (L.C.); Tel.: +886-3-574-2775 (L.C.); Fax: +886-3-571-5934 (L.C.)
| |
Collapse
|
823
|
Guillemaud O, Ceyzériat K, Saint-Georges T, Cambon K, Petit F, Ben Haim L, Carrillo-de Sauvage MA, Guillermier M, Bernier S, Hérard AS, Joséphine C, Bémelmans AP, Brouillet E, Hantraye P, Bonvento G, Escartin C. Complex roles for reactive astrocytes in the triple transgenic mouse model of Alzheimer disease. Neurobiol Aging 2020; 90:135-146. [PMID: 32171592 DOI: 10.1016/j.neurobiolaging.2020.02.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/29/2022]
Abstract
In Alzheimer disease (AD), astrocytes undergo complex changes and become reactive. The consequences of this reaction are still unclear. To evaluate the net impact of reactive astrocytes in AD, we developed viral vectors targeting astrocytes that either activate or inhibit the Janus kinase-signal transducer and activator of transcription 3 (JAK2-STAT3) pathway, a central cascade controlling astrocyte reaction. We aimed to evaluate whether reactive astrocytes contribute to tau as well as amyloid pathologies in the hippocampus of 3xTg-AD mice, an AD model that develops tau hyper-phosphorylation and amyloid deposition. JAK2-STAT3 pathway-mediated modulation of reactive astrocytes in 25% of the hippocampus of 3xTg-AD mice did not significantly influence tau phosphorylation or amyloid processing and deposition at early, advanced, and terminal disease stage. Interestingly, inhibition of the JAK2-STAT3 pathway in hippocampal astrocytes did not improve spatial memory in the Y maze but it did reduce anxiety in the elevated plus maze. Our unique approach to specifically manipulate reactive astrocytes in situ show they may impact behavioral outcomes without influencing tau or amyloid pathology.
Collapse
Affiliation(s)
- Océane Guillemaud
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Kelly Ceyzériat
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Thomas Saint-Georges
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Karine Cambon
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Fanny Petit
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Lucile Ben Haim
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | | | - Martine Guillermier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Sueva Bernier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Anne-Sophie Hérard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Charlène Joséphine
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Alexis Pierre Bémelmans
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Emmanuel Brouillet
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| |
Collapse
|
824
|
Astrocytic YAP Promotes the Formation of Glia Scars and Neural Regeneration after Spinal Cord Injury. J Neurosci 2020; 40:2644-2662. [PMID: 32066583 DOI: 10.1523/jneurosci.2229-19.2020] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 12/15/2022] Open
Abstract
Yes-associated protein (YAP) transcriptional coactivator is negatively regulated by the Hippo pathway and functions in controlling the size of multiple organs, such as liver during development. However, it is not clear whether YAP signaling participates in the process of the formation of glia scars after spinal cord injury (SCI). In this study, we found that YAP was upregulated and activated in astrocytes of C57BL/6 male mice after SCI in a Hippo pathway-dependent manner. Conditional knockout (KO) of yap in astrocytes significantly inhibited astrocytic proliferation, impaired the formation of glial scars, inhibited the axonal regeneration, and impaired the behavioral recovery of C57BL/6 male mice after SCI. Mechanistically, the bFGF was upregulated after SCI and induced the activation of YAP through RhoA pathways, thereby promoting the formation of glial scars. Additionally, YAP promoted bFGF-induced proliferation by negatively controlling nuclear distribution of p27Kip1 mediated by CRM1. Finally, bFGF or XMU-MP-1 (an inhibitor of Hippo kinase MST1/2 to activate YAP) injection indeed activated YAP signaling and promoted the formation of glial scars and the functional recovery of mice after SCI. These findings suggest that YAP promotes the formation of glial scars and neural regeneration of mice after SCI, and that the bFGF-RhoA-YAP-p27Kip1 pathway positively regulates astrocytic proliferation after SCI.SIGNIFICANCE STATEMENT Glial scars play critical roles in neuronal regeneration of CNS injury diseases, such as spinal cord injury (SCI). Here, we provide evidence for the function of Yes-associated protein (YAP) in the formation of glial scars after SCI through regulation of astrocyte proliferation. As a downstream of bFGF (which is upregulated after SCI), YAP promotes the proliferation of astrocytes through negatively controlling nuclear distribution of p27Kip1 mediated by CRM1. Activation of YAP by bFGF or XMU-MP-1 injection promotes the formation of glial scar and the functional recovery of mice after SCI. These results suggest that the bFGF-RhoA-YAP-p27Kip1 axis for the formation of glial scars may be a potential therapeutic strategy for SCI patients.
Collapse
|
825
|
Scheib J, Byrd-Jacobs C. Zebrafish Astroglial Morphology in the Olfactory Bulb Is Altered With Repetitive Peripheral Damage. Front Neuroanat 2020; 14:4. [PMID: 32116575 PMCID: PMC7026507 DOI: 10.3389/fnana.2020.00004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Zebrafish do not possess the typical astrocytes that are found in mammalian systems. In some brain areas, this teleost has radial glia that appears to perform astrocyte-like functions, but these cells have not been described in the zebrafish olfactory bulb. Mammalian astrocytes facilitate neuroplasticity and undergo astrogliosis after insult. The role of these cells in the zebrafish olfactory system after the damage has been poorly explored. This is important to examine because zebrafish have a high degree of neuroplasticity and the olfactory bulb is a brain area renowned for plasticity. The goal of this study was to explore the potential role of zebrafish astrocytes in the olfactory bulb damage response, with a goal to exploit the high level of regeneration in this system. We found that anti-glial fibrillary acidic protein (GFAP) labels numerous processes in the zebrafish olfactory bulb that are concentrated in the nerve and glomerular layers (GL) and do not show radial glial-like morphology. We propose to term this astroglia, since their location and response to damage suggests that they are similar in function to the mammalian astrocyte. To induce repetitive peripheral damage to the olfactory organ, a wax plug was inserted into the nasal cavity of adult zebrafish every 12 h for up to 7 days; this crushes the olfactory organ and leads to degradation of olfactory sensory neuron axons that project to the olfactory bulb. After 1 day, we found a significant increase in astroglial labeling in the affected bulb when compared to the internal control bulb and astroglial branches appeared to increase in number and size. By the third day of plug insertions there was no significant difference in astroglial labeling between the affected bulb and the internal control bulb. These data lead us to believe that astrogliosis does occur in the presence of peripheral damage, but this process attenuates within 1 week and no glial scar is evident upon recovery from the damage. Further exploration of astrocytes in zebrafish, in particular this apparent attenuation of astrogliosis, has the potential to elucidate key differences in glial function between teleosts and mammals.
Collapse
Affiliation(s)
- Jackson Scheib
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, United States
| | - Christine Byrd-Jacobs
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, United States
| |
Collapse
|
826
|
Yu X, Nagai J, Khakh BS. Improved tools to study astrocytes. Nat Rev Neurosci 2020; 21:121-138. [DOI: 10.1038/s41583-020-0264-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
|
827
|
Immune cell regulation of glia during CNS injury and disease. Nat Rev Neurosci 2020; 21:139-152. [PMID: 32042145 DOI: 10.1038/s41583-020-0263-9] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
|
828
|
Sun MK, Passaro AP, Latchoumane CF, Spellicy SE, Bowler M, Goeden M, Martin WJ, Holmes PV, Stice SL, Karumbaiah L. Extracellular Vesicles Mediate Neuroprotection and Functional Recovery after Traumatic Brain Injury. J Neurotrauma 2020; 37:1358-1369. [PMID: 31774030 DOI: 10.1089/neu.2019.6443] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The lack of effective therapies for moderate-to-severe traumatic brain injuries (TBIs) leaves patients with lifelong disabilities. Neural stem cells (NSCs) have demonstrated great promise for neural repair and regeneration. However, direct evidence to support their use as a cell replacement therapy for neural injuries is currently lacking. We hypothesized that NSC-derived extracellular vesicles (NSC EVs) mediate repair indirectly after TBI by enhancing neuroprotection and therapeutic efficacy of endogenous NSCs. We evaluated the short-term effects of acute intravenous injections of NSC EVs immediately following a rat TBI. Male NSC EV-treated rats demonstrated significantly reduced lesion sizes, enhanced presence of endogenous NSCs, and attenuated motor function impairments 4 weeks post-TBI, when compared with vehicle- and TBI-only male controls. Although statistically not significant, we observed a therapeutic effect of NSC EVs on brain lesion volume, nestin expression, and behavioral recovery in female subjects. Our study demonstrates the neuroprotective and functional benefits of NSC EVs for treating TBI and points to gender-dependent effects on treatment outcomes, which requires further investigation.
Collapse
Affiliation(s)
- Min Kyoung Sun
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
| | - Austin P Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
| | - Charles-Francois Latchoumane
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia, USA
| | - Samantha E Spellicy
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
| | - Michael Bowler
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Morgan Goeden
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - William J Martin
- Animal Health Research Center, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Philip V Holmes
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
- Department of Psychology, University of Georgia, Athens, Georgia, USA
| | - Steven L Stice
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia, USA
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, USA
- Interdisciplinary Neuroscience Program, University of Georgia, Athens, Georgia, USA
- Department of Animal and Dairy Science, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
829
|
Wang P, Wang H, Ma K, Wang S, Yang C, Mu N, Yang F, Feng H, Chen T. Novel cytokine-loaded PCL-PEG scaffold composites for spinal cord injury repair. RSC Adv 2020; 10:6306-6314. [PMID: 35495987 PMCID: PMC9049693 DOI: 10.1039/c9ra10385f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/20/2020] [Indexed: 12/14/2022] Open
Abstract
Severe spinal cord injury (SCI) always leads to permanent sensory and motor dysfunction. However, the therapeutic effects of current treatment methods, including high dose methylprednisolone, surgical interventions and rehabilitative care, are far from satisfactory. In recent years, cellular, molecular, tissue engineering and rehabilitative training have shown promising results in animal models. Poly-ε-caprolacton (PCL) - based hydrogel composite system has been considered as a promising strategy to direct the axon growth and mimic the properties of natural extracellular matrix. In this study, we found the addition of the fibroblast growth factor 2 (FGF2) and epidermal growth factor (EGF) to the hydrogel induces the production of axon growth-supportive substrates. The addition of the glial-derived neurotrophic factor (GDNF) to the hydrogel further induces axon directional growth. This "five-in-one" composite scaffold, referred to as PCL/PEG/FGF2/EGF/GDNF, improved the locomotor function in rats 8 weeks after spinal cord injury (SCI) after implantation in transected spinal cord. Furthermore, histological assessment indicated that the designed composite scaffold guided the neuronal regeneration and promoted the production of axon growth-supportive substrates, providing a favorable biological microenvironment. Our novel composite scaffold provides a promising therapeutic method for SCI.
Collapse
Affiliation(s)
- Pangbo Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Hufei Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Kang Ma
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Shi Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Chuanyan Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Ning Mu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Fei Yang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Tunan Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| |
Collapse
|
830
|
Griffin JM, Fackelmeier B, Clemett CA, Fong DM, Mouravlev A, Young D, O'Carroll SJ. Astrocyte-selective AAV-ADAMTS4 gene therapy combined with hindlimb rehabilitation promotes functional recovery after spinal cord injury. Exp Neurol 2020; 327:113232. [PMID: 32044329 DOI: 10.1016/j.expneurol.2020.113232] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 01/06/2023]
Abstract
Chondroitin sulphate proteoglycans (CSPGs) are inhibitors to axon regeneration and plasticity. A disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS4) is a human enzyme that catalyses the proteolysis of CSPG protein cores. Infusion of ADAMTS4 into the damaged spinal cord was previously shown to improve functional recovery SCI, however, this therapy is limited in its enzyme form. Adeno-associated viral (AAV) vector gene therapy has emerged as the vector of choice for safe, robust and long-term transgene expression in the central nervous system. Here, an AAV expression cassette containing ADAMTS4 under the control of the astrocytic GfaABC1D promoter was packaged into an AAV5 vector. Sustained expression of ADAMTS4 was achieved in vitro and in vivo leading to degradation of CSPGs. Compared to a contusion only group, AAV-ADAMTS4 resulted in significantly decreased lesion size, increased sprouting of hindlimb corticospinal tract axons, increased serotonergic fiber density caudal to a contusive spinal cord injury. Hindlimb-specific exercise rehabilitation was used to drive neuroplasticity towards improving functional connections. The combination of hindlimb rehabilitation with AAV-ADAMTS4 led to functional recovery after SCI compared to a contusion only group. Thus, long-term degradation of CSPGs through AAV-ADAMTS4 gene therapy in a combinational approach with rehabilitation represents a candidate for further preclinical development.
Collapse
Affiliation(s)
- Jarred M Griffin
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand; Centre for Brain Research, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand.
| | - Barbara Fackelmeier
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand; Centre for Brain Research, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand.
| | - Connor A Clemett
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand; Centre for Brain Research, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand.
| | - Dahna M Fong
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand; Centre for Brain Research, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand.
| | - Alexandre Mouravlev
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand; Centre for Brain Research, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand.
| | - Deborah Young
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand; Centre for Brain Research, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand.
| | - Simon J O'Carroll
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand; Centre for Brain Research, University of Auckland, Auckland, 85 Park Road, Grafton, New Zealand.
| |
Collapse
|
831
|
Yao M, Ventura PB, Jiang Y, Rodriguez FJ, Wang L, Perry JSA, Yang Y, Wahl K, Crittenden RB, Bennett ML, Qi L, Gong CC, Li XN, Barres BA, Bender TP, Ravichandran KS, Janes KA, Eberhart CG, Zong H. Astrocytic trans-Differentiation Completes a Multicellular Paracrine Feedback Loop Required for Medulloblastoma Tumor Growth. Cell 2020; 180:502-520.e19. [PMID: 31983537 PMCID: PMC7259679 DOI: 10.1016/j.cell.2019.12.024] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/16/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) is critical for tumor progression. However, the establishment and function of the TME remain obscure because of its complex cellular composition. Using a mouse genetic system called mosaic analysis with double markers (MADMs), we delineated TME evolution at single-cell resolution in sonic hedgehog (SHH)-activated medulloblastomas that originate from unipotent granule neuron progenitors in the brain. First, we found that astrocytes within the TME (TuAstrocytes) were trans-differentiated from tumor granule neuron precursors (GNPs), which normally never differentiate into astrocytes. Second, we identified that TME-derived IGF1 promotes tumor progression. Third, we uncovered that insulin-like growth factor 1 (IGF1) is produced by tumor-associated microglia in response to interleukin-4 (IL-4) stimulation. Finally, we found that IL-4 is secreted by TuAstrocytes. Collectively, our studies reveal an evolutionary process that produces a multi-lateral network within the TME of medulloblastoma: a fraction of tumor cells trans-differentiate into TuAstrocytes, which, in turn, produce IL-4 that stimulates microglia to produce IGF1 to promote tumor progression.
Collapse
Affiliation(s)
- Maojin Yao
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - P Britten Ventura
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ying Jiang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Fausto J Rodriguez
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Justin S A Perry
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yibo Yang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kelsey Wahl
- Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Rowena B Crittenden
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
| | - Mariko L Bennett
- Department of Neurobiology, Stanford University, Palo Alto, CA 94305, USA
| | - Lin Qi
- Brain Tumor Program, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cong-Cong Gong
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xiao-Nan Li
- Brain Tumor Program, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University, Palo Alto, CA 94305, USA
| | - Timothy P Bender
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA; VIB-UGent Center for Inflammation Research and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Charles G Eberhart
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
832
|
Zhu S, Chen M, Deng L, Zhang J, Ni W, Wang X, Yao F, Li X, Xu H, Xu J, Xiao J. The repair and autophagy mechanisms of hypoxia-regulated bFGF-modified primary embryonic neural stem cells in spinal cord injury. Stem Cells Transl Med 2020; 9:603-619. [PMID: 32027101 PMCID: PMC7180297 DOI: 10.1002/sctm.19-0282] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022] Open
Abstract
There is no effective strategy for the treatment of spinal cord injury (SCI), a devastating condition characterized by severe hypoxia and ischemic insults. In this study, we investigated the histology and pathophysiology of the SCI milieu in a rat model and found that areas of hypoxia were unevenly interspersed in compressed SCI. With this new knowledge, we generated embryonic neural stem cells (NSCs) expressing basic fibroblast growth factor (bFGF) under the regulation of five hypoxia-responsive elements (5HRE) using a lentiviral vector (LV-5HRE-bFGF-NSCs) to specifically target these hypoxic loci. SCI models treated with bFGF expressed by the LV-5HRE-bFGF-NSCs viral vector demonstrated improved recovery, increased neuronal survival, and inhibited autophagy in spinal cord lesions in the rat model due to the reversal of hypoxic conditions at day 42 after injury. Furthermore, improved functional restoration of SCI with neuron regeneration was achieved in vivo, accompanied by glial scar inhibition and the evidence of axon regeneration across the scar boundary. This is the first study to illustrate the presence of hypoxic clusters throughout the injury site of compressed SCI and the first to show that the transplantation of LV-5HRE-bFGF-NSCs to target this hypoxic microenvironment enhanced the recovery of neurological function after SCI in rats; LV-5HRE-bFGF-NSCs may therefore be a good candidate to evaluate cellular SCI therapy in humans.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Molecular Laboratory, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Min Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Liancheng Deng
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jinjing Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenfei Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Felix Yao
- Molecular Laboratory, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Xiaokun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Laboratory, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Jian Xiao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
833
|
Smethurst P, Risse E, Tyzack GE, Mitchell JS, Taha DM, Chen YR, Newcombe J, Collinge J, Sidle K, Patani R. Distinct responses of neurons and astrocytes to TDP-43 proteinopathy in amyotrophic lateral sclerosis. Brain 2020; 143:430-440. [PMID: 32040555 PMCID: PMC7009461 DOI: 10.1093/brain/awz419] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/12/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disease caused by motor neuron loss, resulting in muscle wasting, paralysis and eventual death. A key pathological feature of ALS is cytoplasmically mislocalized and aggregated TDP-43 protein in >95% of cases, which is considered to have prion-like properties. Historical studies have predominantly focused on genetic forms of ALS, which represent ∼10% of cases, leaving the remaining 90% of sporadic ALS relatively understudied. Additionally, the role of astrocytes in ALS and their relationship with TDP-43 pathology is also not currently well understood. We have therefore used highly enriched human induced pluripotent stem cell (iPSC)-derived motor neurons and astrocytes to model early cell type-specific features of sporadic ALS. We first demonstrate seeded aggregation of TDP-43 by exposing human iPSC-derived motor neurons to serially passaged sporadic ALS post-mortem tissue (spALS) extracts. Next, we show that human iPSC-derived motor neurons are more vulnerable to TDP-43 aggregation and toxicity compared with their astrocyte counterparts. We demonstrate that these TDP-43 aggregates can more readily propagate from motor neurons into astrocytes in co-culture paradigms. We next found that astrocytes are neuroprotective to seeded aggregation within motor neurons by reducing (mislocalized) cytoplasmic TDP-43, TDP-43 aggregation and cell toxicity. Furthermore, we detected TDP-43 oligomers in these spALS spinal cord extracts, and as such demonstrated that highly purified recombinant TDP-43 oligomers can reproduce this observed cell-type specific toxicity, providing further support to a protein oligomer-mediated toxicity hypothesis in ALS. In summary, we have developed a human, clinically relevant, and cell-type specific modelling platform that recapitulates key aspects of sporadic ALS and uncovers both an initial neuroprotective role for astrocytes and the cell type-specific toxic effect of TDP-43 oligomers.
Collapse
Affiliation(s)
- Phillip Smethurst
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emmanuel Risse
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, Courtauld Building, 33 Cleveland Street, London W1W 7FF
| | - Giulia E Tyzack
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jamie S Mitchell
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Doaa M Taha
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Yun-Ru Chen
- Genomics Research Center, Academia Sinica, 128, Academia Road, Section 2, Nankang District, Taipei 115, Taiwan
| | - Jia Newcombe
- NeuroResource, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - John Collinge
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, Courtauld Building, 33 Cleveland Street, London W1W 7FF
| | - Katie Sidle
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
834
|
Wheeler MA, Clark IC, Tjon EC, Li Z, Zandee SEJ, Couturier CP, Watson BR, Scalisi G, Alkwai S, Rothhammer V, Rotem A, Heyman JA, Thaploo S, Sanmarco LM, Ragoussis J, Weitz DA, Petrecca K, Moffitt JR, Becher B, Antel JP, Prat A, Quintana FJ. MAFG-driven astrocytes promote CNS inflammation. Nature 2020; 578:593-599. [PMID: 32051591 PMCID: PMC8049843 DOI: 10.1038/s41586-020-1999-0] [Citation(s) in RCA: 306] [Impact Index Per Article: 61.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/16/2019] [Indexed: 01/14/2023]
Abstract
Multiple sclerosis is a chronic inflammatory disease of the CNS1. Astrocytes contribute to the pathogenesis of multiple sclerosis2, but little is known about the heterogeneity of astrocytes and its regulation. Here we report the analysis of astrocytes in multiple sclerosis and its preclinical model experimental autoimmune encephalomyelitis (EAE) by single-cell RNA sequencing in combination with cell-specific Ribotag RNA profiling, assay for transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunoprecipitation with sequencing (ChIP-seq), genome-wide analysis of DNA methylation and in vivo CRISPR-Cas9-based genetic perturbations. We identified astrocytes in EAE and multiple sclerosis that were characterized by decreased expression of NRF2 and increased expression of MAFG, which cooperates with MAT2α to promote DNA methylation and represses antioxidant and anti-inflammatory transcriptional programs. Granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling in astrocytes drives the expression of MAFG and MAT2α and pro-inflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, multiple sclerosis. Our results identify candidate therapeutic targets in multiple sclerosis.
Collapse
Affiliation(s)
- Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Iain C Clark
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie E J Zandee
- Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Charles P Couturier
- Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Brianna R Watson
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Giulia Scalisi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Alkwai
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Assaf Rotem
- Department of Physics, Harvard University, Cambridge, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - John A Heyman
- Department of Physics, Harvard University, Cambridge, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Shravan Thaploo
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liliana M Sanmarco
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiannis Ragoussis
- McGill University and Genome Quebec Innovation Centre, Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - David A Weitz
- Department of Physics, Harvard University, Cambridge, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Kevin Petrecca
- Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Jeffrey R Moffitt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
835
|
Lu A, Baker-Nigh A, Sun P. Operation spinal cord regeneration: Patterning information residing in extracellular matrix glycosaminoglycans. Brain Behav 2020; 10:e01531. [PMID: 31944630 PMCID: PMC7010577 DOI: 10.1002/brb3.1531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/11/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Spinal cord injuries are devastating, with many complications beyond paralysis and loss of sensory function. Although spinal cord regeneration can revolutionize treatment for spinal cord injuries, the goal has not yet been achieved. The regenerative mechanism of axolotls demonstrates that the regeneration is a repeat of developmental process that all animals have all the genes, but axolotls have both the genes and the patterning information to do it at the adult stage. METHODS A narrative review was conducted. Relevant studies were collected via an English-language PubMed database search and those known to the authors. RESULTS Research during the past 30 years reveals that growth factors, along with spinal cord extracellular matrix, especially glycosaminoglycans, regulates axonal regrowth. Degrading chondroitin sulfate glycosaminoglycans by injecting the bacterial enzyme chondroitinase improves axonal sprouting and functional recovery after spinal cord injury in both rodents and rhesus monkeys. Furthermore, the brain is one of the first organs to develop during the embryonic period, and heparan sulfate glycosaminoglycans are key molecules required for brain development. CONCLUSIONS Patterning information residing in glycosaminoglycans might be key elements in restricting spinal cord regeneration. A recommended solution is not to edit the human genome, considering the conserved signaling pathways between animals, but to take advantage of the regenerative mechanism of axolotls and the current knowledge about the pattern-forming glycosaminoglycans for successful spinal cord regeneration and clinical applications.
Collapse
Affiliation(s)
- Alexander Lu
- Department of Biology, Saint Louis University, St. Louis, Missouri.,Program in Neuroscience, Saint Louis University, St. Louis, Missouri
| | - Alaina Baker-Nigh
- Department of Biology, Saint Louis University, St. Louis, Missouri.,Program in Neuroscience, Saint Louis University, St. Louis, Missouri
| | - Peng Sun
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
836
|
Yang Z, Bao Y, Chen W, He Y. Melatonin exerts neuroprotective effects by attenuating astro- and microgliosis and suppressing inflammatory response following spinal cord injury. Neuropeptides 2020; 79:102002. [PMID: 31902595 DOI: 10.1016/j.npep.2019.102002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/15/2019] [Accepted: 12/20/2019] [Indexed: 01/17/2023]
Abstract
Reactive gliosis and inflammatory reaction are common pathological change to spinal cord injury (SCI). Whereas, the effects of melatonin (MT) on the astro- and microgliosis and their related inflammatory response in the injured spinal cord are not fully understood. In this study, MT's effects on the accumulation and proliferation of microglia and astrocytes and their related inflammatory response were investigated in an acute SCI model. The effects of MT on oxidative stress, neuronal survival and behavioral performance were also tested. It was found that MT treatment significantly suppressed the accumulation and the proliferation of microglia and astrocytes. Quantitative PCR data showed that MT significantly down-regulated the pro-inflammatory markers iNOS, IL-1β and TNF-α expressions. The data showed that MT led to the rise in SOD, CAT and GSH-Px contents and the decrease in MDA content. Western blotting analysis verified that MT significantly down-regulated caspase-3, Bax and GFAP expressions, up-regulated Bcl-2 expression. Compared with the SCI vehicle-treated group, the SCI MT-treated group exhibited a greater Basso Mouse Scale (BMS) locomotor rating score. On the whole, these findings implied that MT exerts its neuroprotective effects by suppressing the accumulation and the proliferation of microglia and astrocytes and reducing the release of pro-inflammatory cytokines, which might be one of the underlying mechanisms of the MT's neuroprotective effect after SCI. Accordingly, MT may be a promising therapeutic candidate for acute SCI.
Collapse
Affiliation(s)
- Zhijie Yang
- School of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Yingying Bao
- School of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Weigang Chen
- School of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuqin He
- School of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China.
| |
Collapse
|
837
|
Goetzl EJ, Yaffe K, Peltz CB, Ledreux A, Gorgens K, Davidson B, Granholm AC, Mustapic M, Kapogiannis D, Tweedie D, Greig NH. Traumatic brain injury increases plasma astrocyte-derived exosome levels of neurotoxic complement proteins. FASEB J 2020; 34:3359-3366. [PMID: 31916313 PMCID: PMC7459190 DOI: 10.1096/fj.201902842r] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 01/16/2023]
Abstract
Possible involvement of complement (C) systems in the pathogenesis of traumatic brain injury (TBI) was investigated by quantifying Cproteins in plasma astrocyte-derived exosomes (ADEs) of subjects with sports-related TBI (sTBI) and TBI in military veterans (mtTBI) without cognitive impairment. All sTBI subjects (n = 24) had mild injuries, whereas eight of the mtTBI subjects had moderate, and 17 had mild injuries. Plasma levels of ADEs were decreased after acute sTBI and returned to normal within months. Cprotein levels in ADEs were from 12- to 35-fold higher than the corresponding levels in neuron-derived exosomes. CD81 exosome marker-normalized ADE levels of classical pathway C4b, alternative pathway factor D and Bb, lectin pathway mannose-binding lectin (MBL), and shared neurotoxic effectors C3b and C5b-9 terminal C complex were significantly higher and those of C regulatory proteins CR1 and CD59 were lower in the first week of acute sTBI (n = 12) than in controls (n = 12). Most C abnormalities were no longer detected in chronic sTBI at 3-12 months after acute sTBI, except for elevated levels of factor D, Bb, and MBL. In contrast, significant elevations of ADE levels of C4b, factor D, Bb, MBL, C3b and C5b-9 terminal C complex, and depressions of CR1 and CD59 relative to those of controls were observed after 1-4 years in early chronic mtTBI (n = 10) and persisted for decades except for normalization of Bb, MBL, and CD59 in late chronic mtTBI (n = 15). Complement inhibitors may be useful therapeutically in acute TBI and post-concussion syndrome.
Collapse
Affiliation(s)
- Edward J. Goetzl
- Department of Medicine, University of California Medical Center, San Francisco, CA, USA
| | - Kristine Yaffe
- Neurology-Psychiatry, University of California Medical Center, San Francisco, CA, USA
- Department of Psychiatry, San Francisco VA Medical Center, San Francisco, CA, USA
| | - Carrie B. Peltz
- Department of Psychiatry, San Francisco VA Medical Center, San Francisco, CA, USA
- Northern California Institute for Research and Education, San Francisco, CA, USA
| | - Aurélie Ledreux
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Kim Gorgens
- Graduate School of Professional Psychology, University of Denver, Denver, CO, USA
| | - Bradley Davidson
- Department of Mechanical and Materials Engineering, University of Denver, Denver, CO, USA
| | | | - Maja Mustapic
- Laboratory for Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory for Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - David Tweedie
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Nigel H. Greig
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| |
Collapse
|
838
|
Wang J, Li D, Liang C, Wang C, Zhou X, Ying L, Tao Y, Xu H, Shu J, Huang X, Gong Z, Xia K, Li F, Chen Q, Tang J, Shen Y. Scar Tissue-Targeting Polymer Micelle for Spinal Cord Injury Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1906415. [PMID: 32003924 DOI: 10.1002/smll.201906415] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/07/2020] [Indexed: 06/10/2023]
Abstract
Spinal cord injury (SCI) is a devastating disorder, leading to permanent motor and sensory deficit. Despite recent advances in neurosciences, the treatment efficacy on SCI patients remains unsatisfactory, mainly due to the poor accumulation, short retention, and lack of controlled release of therapeutics in lesion tissue. Herein, an injured spinal cord targeting prodrug polymer micelle is built. An esterase-responsive bond is used to link apocynin (APO) monomer, because of the enhanced esterase activity found in microglia cells after activation, which ensures a controlled degradation of APO prodrug (Allyloxypolyethyleneglycol-b-poly [2-(((4-acetyl-2-methoxyphenoxy)carbonyl)oxy)ethyl methacrylate], APEG-PAPO or PAPO) by activated microglia cells. A scar tissue-homing peptide (cysteine-alanine-glutamine-lysine, CAQK) is introduced to the PAPO to endow the polymer micelle the lesion tissue-targeting ability. As a result, this CAQK-modified prodrug micelle (cPAM) exhibits an improved accumulation and prolonged retention in lesion tissue compared to the control micelle. The cPAM also leads to superior tissue protection and sustained motor function recovery than the control groups in a mouse model of SCI. In conclusion, the cPAM induces an effective treatment of SCI by the lesion tissue specific delivery of the prodrug polymer via its robust scar binding effect, making the scar tissue a drug releasing platform for sustained treatment of SCI.
Collapse
Affiliation(s)
- Jingkai Wang
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Dongdong Li
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Chengzhen Liang
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Chenggui Wang
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Xiaopeng Zhou
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Liwei Ying
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Yiqing Tao
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Hongxia Xu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Jiawei Shu
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Xianpeng Huang
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Zhe Gong
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Kaishun Xia
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Fangcai Li
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Qixin Chen
- Department of Orthopedics, Second affiliated hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, P. R. China
| | - Jianbin Tang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| | - Youqing Shen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, P. R. China
| |
Collapse
|
839
|
Ma D, Zhao Y, Huang L, Xiao Z, Chen B, Shi Y, Shen H, Dai J. A novel hydrogel-based treatment for complete transection spinal cord injury repair is driven by microglia/macrophages repopulation. Biomaterials 2020; 237:119830. [PMID: 32036301 DOI: 10.1016/j.biomaterials.2020.119830] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 12/28/2019] [Accepted: 01/25/2020] [Indexed: 12/17/2022]
Abstract
Microglia/macrophage mediated-inflammation, a main contributor to the microenvironment after spinal cord injury (SCI), persists for a long period of time and affects SCI repair. However, the effects of microglia/macrophage mediated-inflammation on neurogenic differentiation of endogenous neural stem/progenitor cells (NSPCs) are not well understood. In this study, to attenuate activated microglia/macrophage mediated-inflammation in the spinal cord of complete transection SCI mice, a combination of photo-crosslinked hydrogel transplantation and CSF1R inhibitor (PLX3397) treatment was used to replace the prolonged, activated microglia/macrophages via cell depletion and repopulation. This combined treatment in SCI mice produced a significant reduction in CD68-positive reactive microglia/macrophages and mRNA levels of pro-inflammatory factors, and a substantial increase in the number of Tuj1-positive neurons in the lesion area compared with single treatment methods. Moreover, most of the newborn Tuj1-positive neurons were confirmed to be generated from endogenous NSPCs using a genetic fate mapping mouse line (Nestin-CreERT2; LSL-tdTomato) that can label and trace NSPC marker-nestin expressing cells and their progenies. Collectively, our findings show that the combined treatment method for inhibiting microglia/macrophage mediated-inflammation promotes endogenous NSPC neurogenesis and improves functional recovery, which provides a promising therapeutic strategy for complete transection SCI.
Collapse
Affiliation(s)
- Dezun Ma
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Lei Huang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, PR China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, PR China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Beijing, 100101, PR China.
| |
Collapse
|
840
|
Vismara I, Papa S, Veneruso V, Mauri E, Mariani A, De Paola M, Affatato R, Rossetti A, Sponchioni M, Moscatelli D, Sacchetti A, Rossi F, Forloni G, Veglianese P. Selective Modulation of A1 Astrocytes by Drug-Loaded Nano-Structured Gel in Spinal Cord Injury. ACS NANO 2020; 14:360-371. [PMID: 31887011 DOI: 10.1021/acsnano.9b05579] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Astrogliosis has a very dynamic response during the progression of spinal cord injury, with beneficial or detrimental effects on recovery. It is therefore important to develop strategies to target activated astrocytes and their harmful molecular mechanisms so as to promote a protective environment to counteract the progression of the secondary injury. The challenge is to formulate an effective therapy with maximum protective effects, but reduced side effects. In this study, a functionalized nanogel-based nanovector was selectively internalized in activated mouse or human astrocytes. Rolipram, an anti-inflammatory drug, when administered by these nanovectors limited the inflammatory response in A1 astrocytes, reducing iNOS and Lcn2, which in turn reverses the toxic effect of proinflammatory astrocytes on motor neurons in vitro, showing advantages over conventionally administered anti-inflammatory therapy. When tested acutely in a spinal cord injury mouse model, it improved motor performance, but only in the early stage after injury, reducing the astrocytosis and preserving neuronal cells.
Collapse
Affiliation(s)
- Irma Vismara
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| | - Simonetta Papa
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| | - Valeria Veneruso
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Emanuele Mauri
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Alessandro Mariani
- Department of Environmental Health Sciences , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milan , Italy
| | - Massimiliano De Paola
- Department of Environmental Health Sciences , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milan , Italy
| | - Roberta Affatato
- Department of Oncology , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milan , Italy
| | - Arianna Rossetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Mattia Sponchioni
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Davide Moscatelli
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Alessandro Sacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Filippo Rossi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta" , Politecnico di Milano , via Mancinelli 7 , 20131 Milano , Italy
| | - Gianluigi Forloni
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| | - Pietro Veglianese
- Department of Neuroscience , Istituto di Ricerche Farmacologiche Mario Negri IRCCS , via Mario Negri 2 , 20156 Milano , Italy
| |
Collapse
|
841
|
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: 2.0] [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
|
842
|
The use of bioactive matrices in regenerative therapies for traumatic brain injury. Acta Biomater 2020; 102:1-12. [PMID: 31751809 DOI: 10.1016/j.actbio.2019.11.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 01/01/2023]
Abstract
Functional deficits due to neuronal loss are a common theme across multiple neuropathologies, including traumatic brain injury (TBI). Apart from mitigating cell death, another approach to treating brain injuries involves re-establishing the neural circuitry at the lesion site by utilizing exogeneous and/or endogenous stem cells to achieve functional recovery. While there has been limited success, the emergence of new bioactive matrices that promote neural repair introduces new perspectives on the development of regenerative therapies for TBI. This review briefly discusses current development on cell-based therapies and the use of bioactive matrices, hydrogels in particular, when incorporated in regenerative therapies. Desirable characteristics of bioactive matrices that have been shown to augment neural repair in TBI models were identified and further discussed. Understanding the relative outcomes of newly developed biomaterials implanted in vivo can better guide the development of biomaterials as a therapeutic strategy, for biomaterial-based cellular therapies are still in their nascent stages. Nonetheless, the value of bioactive matrices as a treatment for acute brain injuries should be appreciated and further developed. STATEMENT OF SIGNIFICANCE: Cell-based therapies have received attention as an alternative therapeutic strategy to improve clinical outcome post-traumatic brain injury but have achieved limited success. Whilst the incorporation of newly developed biomaterials in regenerative therapies has shown promise in augmenting neural repair, studies have revealed new hurdles which must be overcome to improve their therapeutic efficacy. This review discusses the recent development of cell-based therapies with a specific focus on the use of bioactive matrices in the form of hydrogels, to complement cell transplantation within the injured brain. Moreover, this review consolidates in vivo animal studies that demonstrate relative functional outcome upon the implantation of different biomaterials to highlight their desirable traits to guide their development for regenerative therapies in traumatic brain injury.
Collapse
|
843
|
Mohammed R, Opara K, Lall R, Ojha U, Xiang J. Evaluating the effectiveness of anti-Nogo treatment in spinal cord injuries. Neural Dev 2020; 15:1. [PMID: 31918754 PMCID: PMC6953157 DOI: 10.1186/s13064-020-0138-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 01/05/2020] [Indexed: 02/08/2023] Open
Abstract
As humans, we cannot regenerate axons within the central nervous system (CNS), therefore, making any damage to it permanent. This leads to the loss of sensory and motor function below the site of injury and can be crippling to a person’s health. Spontaneous recovery can occur from plastic changes, but it is minimal. The absence of regeneration is due to the inhibitory environment of the CNS as well as the inherent inability of CNS axons to form growth cones. Amongst many factors, one of the major inhibitory signals of the CNS environment is the myelin-associated Nogo pathway. Nogo-A, Nogo-B and Nogo-C (Nogo), stimulate the Nogo receptor, inhibiting neurite outgrowth by causing growth cones to collapse through activation of Rho Kinase (ROCK). Antibodies can be used to target this signalling pathway by binding to Nogo and thus promote the outgrowth of neuronal axons in the CNS. This use of anti-Nogo antibodies has been shown to upregulate CNS regeneration as well as drastically improve sensory and motor function in both rats and primates when coupled with adequate training. Here, we evaluate whether the experimental success of anti-Nogo at improving CNS regeneration can be carried over into the clinical setting to treat spinal cord injuries (SCI) and their symptoms successfully. Furthermore, we also discuss potential methods to improve the current treatment and any developmental obstacles.
Collapse
Affiliation(s)
- Raihan Mohammed
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Hills Rd, Cambridge, CB2 0SP, UK.
| | - Kaesi Opara
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Hills Rd, Cambridge, CB2 0SP, UK
| | - Rahul Lall
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Hills Rd, Cambridge, CB2 0SP, UK
| | - Utkarsh Ojha
- Faculty of Medicine, Imperial College London, London, UK
| | - Jinpo Xiang
- Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
844
|
Krucoff MO, Miller JP, Saxena T, Bellamkonda R, Rahimpour S, Harward SC, Lad SP, Turner DA. Toward Functional Restoration of the Central Nervous System: A Review of Translational Neuroscience Principles. Neurosurgery 2020; 84:30-40. [PMID: 29800461 DOI: 10.1093/neuros/nyy128] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/15/2018] [Indexed: 01/09/2023] Open
Abstract
Injury to the central nervous system (CNS) can leave patients with devastating neurological deficits that may permanently impair independence and diminish quality of life. Recent insights into how the CNS responds to injury and reacts to critically timed interventions are being translated into clinical applications that have the capacity to drastically improve outcomes for patients suffering from permanent neurological deficits due to spinal cord injury, stroke, or other CNS disorders. The translation of such knowledge into practical and impactful treatments involves the strategic collaboration between neurosurgeons, clinicians, therapists, scientists, and industry. Therefore, a common understanding of key neuroscientific principles is crucial. Conceptually, current approaches to CNS revitalization can be divided by scale into macroscopic (systems-circuitry) and microscopic (cellular-molecular). Here we review both emerging and well-established tenets that are being utilized to enhance CNS recovery on both levels, and we explore the role of neurosurgeons in developing therapies moving forward. Key principles include plasticity-driven functional recovery, cellular signaling mechanisms in axonal sprouting, critical timing for recovery after injury, and mechanisms of action underlying cellular replacement strategies. We then discuss integrative approaches aimed at synergizing interventions across scales, and we make recommendations for the basis of future clinical trial design. Ultimately, we argue that strategic modulation of microscopic cellular behavior within a macroscopic framework of functional circuitry re-establishment should provide the foundation for most neural restoration strategies, and the early involvement of neurosurgeons in the process will be crucial to successful clinical translation.
Collapse
Affiliation(s)
- Max O Krucoff
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Jonathan P Miller
- Department of Neurosurgery, Case Western Reserve University, Cleve-land, Ohio
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Ravi Bellamkonda
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Shervin Rahimpour
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Stephen C Harward
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Shivanand P Lad
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Mechan-ical Engineering and Material Sciences, Pratt School of Engineering, Duke Uni-versity, Durham, North Carolina.,Duke Institute for Brain Sciences, Duke Univer-sity, Durham, North Carolina.,Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina
| | - Dennis A Turner
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Biomedical Engineering, Duke University, Durham, North Carolina.,Depart-ment of Neurobiology, Duke University, Durham, North Carolina.,Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina
| |
Collapse
|
845
|
Blochet C, Buscemi L, Clément T, Gehri S, Badaut J, Hirt L. Involvement of caveolin-1 in neurovascular unit remodeling after stroke: Effects on neovascularization and astrogliosis. J Cereb Blood Flow Metab 2020; 40:163-176. [PMID: 30354902 PMCID: PMC6928561 DOI: 10.1177/0271678x18806893] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Complex cellular and molecular events occur in the neurovascular unit after stroke, such as blood-brain barrier (BBB) dysfunction and inflammation that contribute to neuronal death, neurological deterioration and mortality. Caveolin-1 (Cav-1) has distinct physiological functions such as caveolae formation associated with endocytosis and transcytosis as well as in signaling pathways. Cav-1 has been proposed to be involved in BBB dysfunction after brain injury; however, its precise role is poorly understood. The goal of this study was to characterize the expression and effect of Cav-1 deletion on outcome in the first week in a transient Middle Cerebral Artery Occlusion stroke model. We found increased Cav-1 expression in new blood vessels in the lesion and in reactive astrocytes in the peri-lesion areas. In Cav-1 KO mice, the lesion volume was larger and the behavioral outcome worse than in WT mice. Cav-1 KO mice exhibited reduced neovascularization and modified astrogliosis, without formation of a proper glial scar around the lesion at three days post injury, coinciding with aggravated outcomes. Altogether, these results point towards a potential protective role of endogenous Cav-1 in the first days after ischemia by promoting neovascularization, astrogliosis and scar formation.
Collapse
Affiliation(s)
- Camille Blochet
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland.,Brain Molecular Imaging Lab, CNRS UMR 5287, INCIA, University of Bordeaux, Bordeaux, France
| | - Lara Buscemi
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Tifenn Clément
- Brain Molecular Imaging Lab, CNRS UMR 5287, INCIA, University of Bordeaux, Bordeaux, France
| | - Sabrina Gehri
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| | - Jérôme Badaut
- Brain Molecular Imaging Lab, CNRS UMR 5287, INCIA, University of Bordeaux, Bordeaux, France.,Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Lorenz Hirt
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland
| |
Collapse
|
846
|
Zhou C, Chen H, Zheng JF, Guo ZD, Huang ZJ, Wu Y, Zhong JJ, Sun XC, Cheng CJ. Pentraxin 3 contributes to neurogenesis after traumatic brain injury in mice. Neural Regen Res 2020; 15:2318-2326. [PMID: 32594056 PMCID: PMC7749468 DOI: 10.4103/1673-5374.285001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence indicates that pentraxin 3 is an acute-phase protein that is linked with the immune response to inflammation. It is also a newly discovered marker of anti-inflammatory A2 reactive astrocytes, and potentially has multiple protective effects in stroke; however, its role in the adult brain after traumatic brain injury is unknown. In the present study, a moderate model of traumatic brain injury in mice was established using controlled cortical impact. The models were intraventricularly injected with recombinant pentraxin 3 (the recombinant pentraxin 3 group) or an equal volume of vehicle (the control group). The sham-operated mice underwent craniotomy, but did not undergo the controlled cortical impact. The potential neuroprotective and neuroregenerative roles of pentraxin 3 were investigated on days 14 and 21 after traumatic brain injury. Western blot assay showed that the expression of endogenous pentraxin 3 was increased after traumatic brain injury in mice. Furthermore, the neurological severity test and wire grip test revealed that recombinant pentraxin 3 treatment reduced the neurological severity score and increased the wire grip score, suggesting an improved recovery of sensory-motor functions. The Morris water maze results demonstrated that recombinant pentraxin 3 treatment reduced the latency to the platform, increased the time spent in the correct quadrant, and increased the number of times traveled across the platform, thus suggesting an improved recovery of cognitive function. In addition, to investigate the effects of pentraxin 3 on astrocytes, specific markers of A2 astrocytes were detected in primary astrocyte cultures in vitro using western blot assay. The results demonstrated that pentraxin 3 administration activates A2 astrocytes. To explore the protective mechanisms of pentraxin 3, immunofluorescence staining was used. Intraventricular injection of recombinant pentraxin 3 increased neuronal maintenance in the peri-injured cortex and ipsilateral hippocampus, increased the number of doublecortin-positive neural progenitor cells in the subventricular and subgranular zones, and increased the number of bromodeoxyuridine (proliferation) and neuronal nuclear antigen (mature neuron) double-labeled cells in the hippocampus and peri-injured cortex. Pentraxin 3 administration also increased the number of neurospheres and the number of bromodeoxyuridine and doublecortin double-labeled cells in neurospheres, and enhanced the proliferation of neural progenitor cells in primary neural progenitor cell cultures in vitro. In conclusion, recombinant pentraxin 3 administration activated A2 astrocytes, and consequently improved the recovery of neural function by increasing neuronal survival and enhancing neurogenesis. All experiments were approved by the Animal Ethics Committee of the First Affiliated Hospital of Chongqing Medical University, China on March 1, 2016.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Feng Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zong-Duo Guo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Jian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Jun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Chuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chong-Jie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
847
|
Xiong W, Zhang C, Lyu X, Zhou H, Chang W, Bo Y, Chen E, Shen Z, Lu H. Synthesis of modifiable photo-responsive polypeptides bearing allyloxyazobenzene side-chains. Polym Chem 2020. [DOI: 10.1039/c9py01106d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A photo-responsive and modifiable polypeptide with stable helical conformation was synthesized. The self-assembly and liquid crystalline phase structure were subsequently studied.
Collapse
Affiliation(s)
- Wei Xiong
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Chong Zhang
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Xiaolin Lyu
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Hantao Zhou
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Wenying Chang
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Yu Bo
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Erqiang Chen
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Zhihao Shen
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| | - Hua Lu
- Beijing National Laboratory for Molecular Sciences
- Center for Soft Matter Science and Engineering
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education
- College of Chemistry and Molecular Engineering
- Peking University
| |
Collapse
|
848
|
Planas-Fontánez TM, Dreyfus CF, Saitta KS. Reactive Astrocytes as Therapeutic Targets for Brain Degenerative Diseases: Roles Played by Metabotropic Glutamate Receptors. Neurochem Res 2020; 45:541-550. [PMID: 31983009 PMCID: PMC7058558 DOI: 10.1007/s11064-020-02968-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/13/2020] [Accepted: 01/18/2020] [Indexed: 02/06/2023]
Abstract
Astrocytes are well known to play critical roles in the development and maintenance of the central nervous system (CNS). Moreover, recent reports indicate that these cells are heterogeneous with respect to the molecules they express and the functions they exhibit in the quiescent or activated state. Because astrocytes also contribute to pathology, promising new results raise the possibility of manipulating specific astroglial populations for therapeutic roles. In this mini-review, we highlight the function of metabotropic glutamate receptors (mGluRs), in particular mGluR3 and mGluR5, in reactive astrocytes and relate these to three degenerative CNS diseases: multiple sclerosis, Alzheimer's disease and Amyotrophic Lateral Sclerosis. Previous studies demonstrate that effects of these receptors may be beneficial, but this varies depending on the subtype of receptor, the state of the astrocytes, and the specific disease to which they are exposed. Elucidating the role of mGluRs on astrocytes at specific times during development and disease will provide novel insights in understanding how to best use these to serve as therapeutic targets.
Collapse
Affiliation(s)
- Talia M. Planas-Fontánez
- grid.430387.b0000 0004 1936 8796Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ USA ,grid.430387.b0000 0004 1936 8796Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ USA
| | - Cheryl F. Dreyfus
- grid.430387.b0000 0004 1936 8796Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ USA ,grid.430387.b0000 0004 1936 8796Robert Wood Johnson Medical School, 683 Hoes Lane West, Room 361, Piscataway, NJ 08854 USA
| | - Kyle S. Saitta
- grid.430387.b0000 0004 1936 8796Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ USA ,grid.430387.b0000 0004 1936 8796Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ USA
| |
Collapse
|
849
|
Deftu AF, Suter MR. Glia and Pain in Spinal Cord. THE SENSES: A COMPREHENSIVE REFERENCE 2020:235-248. [DOI: 10.1016/b978-0-12-809324-5.24214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
850
|
Zhang BY, Chang PY, Zhu QS, Zhu YH. Decoding epigenetic codes: new frontiers in exploring recovery from spinal cord injury. Neural Regen Res 2020; 15:1613-1622. [PMID: 32209760 PMCID: PMC7437595 DOI: 10.4103/1673-5374.276323] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury that results in severe neurological disability is often incurable. The poor clinical outcome of spinal cord injury is mainly caused by the failure to reconstruct the injured neural circuits. Several intrinsic and extrinsic determinants contribute to this inability to reconnect. Epigenetic regulation acts as the driving force for multiple pathological and physiological processes in the central nervous system by modulating the expression of certain critical genes. Recent studies have demonstrated that post-SCI alteration of epigenetic landmarks is strongly associated with axon regeneration, glial activation and neurogenesis. These findings not only establish a theoretical foundation for further exploration of spinal cord injury, but also provide new avenues for the clinical treatment of spinal cord injury. This review focuses on the epigenetic regulation in axon regeneration and secondary spinal cord injury. Together, these discoveries are a selection of epigenetic-based prognosis biomarkers and attractive therapeutic targets in the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Bo-Yin Zhang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Peng-Yu Chang
- Department of Radiotherapy, The First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qing-San Zhu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yu-Hang Zhu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | -
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|