1001
|
Yang Q, Zhou J. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia 2018; 67:1017-1035. [DOI: 10.1002/glia.23571] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Qiao‐qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
| | - Jia‐wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
- University of Chinese Academy of Sciences Shanghai 200031 China
| |
Collapse
|
1002
|
Shi GD, Zhang XL, Cheng X, Wang X, Fan BY, Liu S, Hao Y, Wei ZJ, Zhou XH, Feng SQ. Abnormal DNA Methylation in Thoracic Spinal Cord Tissue Following Transection Injury. Med Sci Monit 2018; 24:8878-8890. [PMID: 30531681 PMCID: PMC6295140 DOI: 10.12659/msm.913141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Spinal cord injury (SCI) is a serious disease with high disability and mortality rates, with no effective therapeutic strategies available. In SCI, abnormal DNA methylation is considered to be associated with axonal regeneration and cell proliferation. However, the roles of key genes in potential molecular mechanisms of SCI are not clear. Material/Methods Subacute spinal cord injury models were established in Wistar rats. Histological observations and motor function assessments were performed separately. Whole-genome bisulfite sequencing (WGBS) was used to detect the methylation of genes. Gene ontology (GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed using the DAVID database. Protein–protein interaction (PPI) networks were analyzed by Cytoscape software. Results After SCI, many cavities, areas of necrotic tissue, and many inflammatory cells were observed, and motor function scores were low. After the whole-genome bisulfite sequencing, approximately 96 DMGs were screened, of which 50 were hypermethylated genes and 46 were hypomethylated genes. KEGG pathway analysis highlighted the Axon Guidance pathway, Endocytosis pathway, T cell receptor signaling pathway, and Hippo signaling pathway. Expression patterns of hypermethylated genes and hypomethylated genes detected by qRT-PCR were the opposite of WGBS data, and the difference was significant. Conclusions Abnormal methylated genes and key signaling pathways involved in spinal cord injury were identified through histological observation, behavioral assessment, and bioinformatics analysis. This research can serve as a source of additional information to expand understanding of spinal cord-induced epigenetic changes.
Collapse
Affiliation(s)
- Gui-Dong Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xiao-Lei Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xin Cheng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xu Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Bao-You Fan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Shen Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Yan Hao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Zhi-Jian Wei
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xian-Hu Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Shi-Qing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| |
Collapse
|
1003
|
Zuidema JM, Gilbert RJ, Gottipati MK. Biomaterial Approaches to Modulate Reactive Astroglial Response. Cells Tissues Organs 2018; 205:372-395. [PMID: 30517922 PMCID: PMC6397084 DOI: 10.1159/000494667] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/18/2018] [Indexed: 11/19/2022] Open
Abstract
Over several decades, biomaterial scientists have developed materials to spur axonal regeneration and limit secondary injury and tested these materials within preclinical animal models. Rarely, though, are astrocytes examined comprehensively when biomaterials are placed into the injury site. Astrocytes support neuronal function in the central nervous system. Following an injury, astrocytes undergo reactive gliosis and create a glial scar. The astrocytic glial scar forms a dense barrier which restricts the extension of regenerating axons through the injury site. However, there are several beneficial effects of the glial scar, including helping to reform the blood-brain barrier, limiting the extent of secondary injury, and supporting the health of regenerating axons near the injury site. This review provides a brief introduction to the role of astrocytes in the spinal cord, discusses astrocyte phenotypic changes that occur following injury, and highlights studies that explored astrocyte changes in response to biomaterials tested within in vitro or in vivo environments. Overall, we suggest that in order to improve biomaterial designs for spinal cord injury applications, investigators should more thoroughly consider the astrocyte response to such designs.
Collapse
Affiliation(s)
- Jonathan M Zuidema
- Department of Chemistry and Biochemistry, University of California, San Diego, California, USA
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Manoj K Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, USA,
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, USA,
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA,
| |
Collapse
|
1004
|
Jones JM, DePaul MA, Gregory CR, Lang BT, Xie H, Zhu M, Rutten MJ, Mays RW, Busch SA, Pati S, Gregory KW. Multipotent Adult Progenitor Cells, but Not Tissue Inhibitor of Matrix Metalloproteinase-3, Increase Tissue Sparing and Reduce Urological Complications following Spinal Cord Injury. J Neurotrauma 2018; 36:1416-1427. [PMID: 30251917 DOI: 10.1089/neu.2018.5727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Following spinal cord injury (SCI), inflammation amplifies damage beyond the initial insult, providing an opportunity for targeted treatments. An ideal protective therapy would reduce both edema within the lesion area and the activation/infiltration of detrimental immune cells. Previous investigations demonstrated the efficacy of intravenous injection of multipotent adult progenitor cells (MAPC®) to modulate immune response following SCI, leading to significant improvements in tissue sparing, locomotor and urological functions. Separate studies have demonstrated that tissue inhibitor of matrix metalloproteinase-3 (TIMP3) reduces blood-brain barrier permeability following traumatic brain injury in a mouse model, leading to improved functional recovery. This study examined whether TIMP3, delivered alone or in concert with MAPC cells, improves functional recovery from a contusion SCI in a rat model. The results suggest that intravenous delivery of MAPC cell therapy 1 day following acute SCI significantly improves tissue sparing and impacts functional recovery. TIMP3 treatment provided no significant benefit, and further, when co-administered with MAPC cells, it abrogated the therapeutic effects of MAPC cell therapy. Importantly, this study demonstrated for the first time that acute treatment of SCI with MAPC cells can significantly reduce the incidence of urinary tract infection (UTI) and the use of antibiotics for UTI treatment.
Collapse
Affiliation(s)
- James M Jones
- 1 Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| | - Marc A DePaul
- 2 Department of Neurosciences, Case Western Reserve University, Cleveland, Ohio
| | - Cynthia R Gregory
- 1 Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon.,3 Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon
| | | | - Hua Xie
- 1 Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon.,5 Department of Surgery, Oregon Health and Science University, Portland, Oregon
| | - Meihua Zhu
- 1 Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| | - Michael J Rutten
- 1 Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon
| | | | | | - Shibani Pati
- 6 Department of Laboratory Medicine, University of California San Francisco, San Francisco, California
| | - Kenton W Gregory
- 1 Center for Regenerative Medicine, Oregon Health and Science University, Portland, Oregon.,7 Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
1005
|
Zanuzzi CN, Nishida F, Sisti MS, Barbeito CG, Portiansky EL. Reactivity of microglia and astrocytes after an excitotoxic injury induced by kainic acid in the rat spinal cord. Tissue Cell 2018; 56:31-40. [PMID: 30736902 DOI: 10.1016/j.tice.2018.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/16/2018] [Accepted: 11/30/2018] [Indexed: 01/20/2023]
Abstract
After injury of the nervous system glial cells react according to the stimuli by modifying their morphology and function. Glia activation was reported in different kainic acid (KA)-induced neurodegeneration models. Here, we describe glial morphometric changes occurring in an excitotoxic KA-induced cervical spinal cord injury model. Concomitant degenerative and apoptotic processes are also reported. Male rats injected at the spinal cord C5 segment either with KA or saline were euthanized at post-injection (PI) days 1, 2, 3 or 7. Anti-IBA-1 and anti-GFAP antibodies were used to identify microglia and activated astrocytes, respectively, and to morphometrically characterized them. Fluoro-Jade B staining and TUNEL reaction were used to determine neuronal and glial degeneration and apoptosis. KA-injected group showed a significant increase in microglia number at the ipsilateral side by PI day 3. Different microglia reactive phenotypes were observed. Reactive microglia was still present by PI day 7. Astrocytes in KA-injected group showed a biphasic increase in number at PI days 1 and 3. Degenerative and apoptotic events were only observed in KA-injected animals, increasing mainly by PI day 1. Understanding the compromise of glia in different neurodegenerative processes may help to define possible common or specific therapeutic approaches directed towards neurorestorative strategies.
Collapse
Affiliation(s)
- Carolina Natalia Zanuzzi
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), Buenos Aires, Argentina; National Research Council of Science and Technology (CONICET), Argentina.
| | - Fabián Nishida
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), Buenos Aires, Argentina; National Research Council of Science and Technology (CONICET), Argentina
| | - María Susana Sisti
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), Buenos Aires, Argentina; National Research Council of Science and Technology (CONICET), Argentina
| | - Claudio Gustavo Barbeito
- Laboratory of Descriptive, Experimental and Comparative, Histology and Embriology, Argentina; National Research Council of Science and Technology (CONICET), Argentina
| | - Enrique Leo Portiansky
- Image Analysis Laboratory, School of Veterinary Sciences, National University of La Plata (UNLP), Buenos Aires, Argentina; National Research Council of Science and Technology (CONICET), Argentina
| |
Collapse
|
1006
|
Kaneko N, Herranz-Pérez V, Otsuka T, Sano H, Ohno N, Omata T, Nguyen HB, Thai TQ, Nambu A, Kawaguchi Y, García-Verdugo JM, Sawamoto K. New neurons use Slit-Robo signaling to migrate through the glial meshwork and approach a lesion for functional regeneration. SCIENCE ADVANCES 2018; 4:eaav0618. [PMID: 30547091 PMCID: PMC6291311 DOI: 10.1126/sciadv.aav0618] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/11/2018] [Indexed: 06/09/2023]
Abstract
After brain injury, neural stem cell-derived neuronal precursors (neuroblasts) in the ventricular-subventricular zone migrate toward the lesion. However, the ability of the mammalian brain to regenerate neuronal circuits for functional recovery is quite limited. Here, using a mouse model for ischemic stroke, we show that neuroblast migration is restricted by reactive astrocytes in and around the lesion. To migrate, the neuroblasts use Slit1-Robo2 signaling to disrupt the actin cytoskeleton in reactive astrocytes at the site of contact. Slit1-overexpressing neuroblasts transplanted into the poststroke brain migrated closer to the lesion than did control neuroblasts. These neuroblasts matured into striatal neurons and efficiently regenerated neuronal circuits, resulting in functional recovery in the poststroke mice. These results suggest that the positioning of new neurons will be critical for functional neuronal regeneration in stem/progenitor cell-based therapies for brain injury.
Collapse
Affiliation(s)
- N. Kaneko
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - V. Herranz-Pérez
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universidad de Valencia, CIBERNED, C/Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain
- Predepartamental Unit of Medicine, Faculty of Health Sciences, Universitat Jaume I, Q-6250003-H Av. de Vicent Sos Baynat, s/n, 12071 Castelló de la Plana, Spain
| | - T. Otsuka
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - H. Sano
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
- Division of System Neurophysiology, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - N. Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - T. Omata
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - H. B. Nguyen
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
- Department of Anatomy, Faculty of Medicine, University of Medicine and Pharmacy (UMP), Ho Chi Minh City 700000, Vietnam
| | - T. Q. Thai
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - A. Nambu
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
- Division of System Neurophysiology, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Y. Kawaguchi
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8585, Japan
| | - J. M. García-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, Universidad de Valencia, CIBERNED, C/Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain
| | - K. Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
1007
|
Nori S, Khazaei M, Ahuja CS, Yokota K, Ahlfors JE, Liu Y, Wang J, Shibata S, Chio J, Hettiaratchi MH, Führmann T, Shoichet MS, Fehlings MG. Human Oligodendrogenic Neural Progenitor Cells Delivered with Chondroitinase ABC Facilitate Functional Repair of Chronic Spinal Cord Injury. Stem Cell Reports 2018; 11:1433-1448. [PMID: 30472009 PMCID: PMC6294173 DOI: 10.1016/j.stemcr.2018.10.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 01/09/2023] Open
Abstract
Treatment of chronic spinal cord injury (SCI) is challenging due to cell loss, cyst formation, and the glial scar. Previously, we reported on the therapeutic potential of a neural progenitor cell (NPC) and chondroitinase ABC (ChABC) combinatorial therapy for chronic SCI. However, the source of NPCs and delivery system required for ChABC remained barriers to clinical application. Here, we investigated directly reprogrammed human NPCs biased toward an oligodendrogenic fate (oNPCs) in combination with sustained delivery of ChABC using an innovative affinity release strategy in a crosslinked methylcellulose biomaterial for the treatment of chronic SCI in an immunodeficient rat model. This combinatorial therapy increased long-term survival of oNPCs around the lesion epicenter, facilitated greater oligodendrocyte differentiation, remyelination of the spared axons by engrafted oNPCs, enhanced synaptic connectivity with anterior horn cells and neurobehavioral recovery. This combinatorial therapy is a promising strategy to regenerate the chronically injured spinal cord. Sustained biomaterial delivery of ChABC successfully degraded CSPGs XMC-ChABC promoted differentiation of oNPCs to more oligodendrocytes XMC-ChABC increased the long-term survival and integration of grafted oNPCs XMC-ChABC and oNPC combinatorial therapy is a promising treatment for chronic SCI
Collapse
Affiliation(s)
- Satoshi Nori
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada; Department of Orthopaedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinju-ku, Tokyo 160-8582, Japan
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Christopher S Ahuja
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Kazuya Yokota
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jan-Eric Ahlfors
- New World Laboratories Inc., 500 Boulevard Cartier Quest, Laval, QC H7V 5B7, Canada
| | - Yang Liu
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Jian Wang
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, 35 Shinanomachi, Shinju-ku, Tokyo 160-8582, Japan
| | - Jonathon Chio
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Marian H Hettiaratchi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada
| | - Tobias Führmann
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, ON M5S 3E5, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON M5S 3H6, Canada; Institute of Biomaterials & Biomedical Engineering, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada; Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Surgery and Spinal Program, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Department of Surgery, Division of Anatomy, Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada.
| |
Collapse
|
1008
|
Insights into the Dual Role of Inflammation after Spinal Cord Injury. J Neurosci 2018; 37:4658-4660. [PMID: 28469010 DOI: 10.1523/jneurosci.0498-17.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/26/2017] [Accepted: 03/30/2017] [Indexed: 01/09/2023] Open
|
1009
|
Kamermans A, Planting KE, Jalink K, van Horssen J, de Vries HE. Reactive astrocytes in multiple sclerosis impair neuronal outgrowth through TRPM7-mediated chondroitin sulfate proteoglycan production. Glia 2018; 67:68-77. [PMID: 30453391 PMCID: PMC6587975 DOI: 10.1002/glia.23526] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system (CNS), characterized by inflammation‐mediated demyelination, axonal injury and neurodegeneration. The mechanisms underlying impaired neuronal function are not fully understood, but evidence is accumulating that the presence of the gliotic scar produced by reactive astrocytes play a critical role in these detrimental processes. Here, we identified astrocytic Transient Receptor Potential cation channel, subfamily M, member 7 (TRPM7), a Ca2+‐permeable nonselective cation channel, as a novel player in the formation of a gliotic scar. TRPM7 was found to be highly expressed in reactive astrocytes within well‐characterized MS lesions and upregulated in primary astrocytes under chronic inflammatory conditions. TRPM7 overexpressing astrocytes impaired neuronal outgrowth in vitro by increasing the production of chondroitin sulfate proteoglycans, a key component of the gliotic scar. These findings indicate that astrocytic TRPM7 is a critical regulator of the formation of a gliotic scar and provide a novel mechanism by which reactive astrocytes affect neuronal outgrowth.
Collapse
Affiliation(s)
- Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kirsten E Planting
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kees Jalink
- Department of Cell Biology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, MS center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
1010
|
Cunningham C, Dunne A, Lopez-Rodriguez AB. Astrocytes: Heterogeneous and Dynamic Phenotypes in Neurodegeneration and Innate Immunity. Neuroscientist 2018; 25:455-474. [PMID: 30451065 DOI: 10.1177/1073858418809941] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Astrocytes are the most numerous cell type in the brain and perform several essential functions in supporting neuronal metabolism and actively participating in neural circuit and behavioral function. They also have essential roles as innate immune cells in responding to local neuropathology, and the manner in which they respond to brain injury and degeneration is the subject of increasing attention in neuroscience. Although activated astrocytes have long been thought of as a relatively homogenous population, which alter their phenotype in a relatively stereotyped way upon central nervous system injury, the last decade has revealed substantial heterogeneity in the basal state and significant heterogeneity of phenotype during reactive astrocytosis. Thus, phenotypic diversity occurs at two distinct levels: that determined by regionality and development and that determined by temporally dynamic changes to the environment of astrocytes during pathology. These inflammatory and pathological states shape the phenotype of these cells, with different consequences for destruction or recovery of the local tissue, and thus elucidating these phenotypic changes has significant therapeutic implications. In this review, we will focus on the phenotypic heterogeneity of astrocytes in health and disease and their propensity to change that phenotype upon subsequent stimuli.
Collapse
Affiliation(s)
- Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland.,School of Medicine, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| | - Ana Belen Lopez-Rodriguez
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| |
Collapse
|
1011
|
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of Cerebrovascular Endothelial Cells: Prelude to Vascular Dementia. Front Aging Neurosci 2018; 10:376. [PMID: 30505270 PMCID: PMC6250852 DOI: 10.3389/fnagi.2018.00376] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia after Alzheimer's disease (AD), characterized by progressive cognitive impairment, memory loss, and thinking or speech problems. VaD is usually caused by cerebrovascular disease, during which, cerebrovascular endothelial cells (CECs) are vulnerable. CEC dysfunction occurs before the onset of VaD and can eventually lead to dysregulation of cerebral blood flow and blood-brain barrier damage, followed by the activation of glia and inflammatory environment in the brain. White matter, neuronal axons, and synapses are compromised in this process, leading to cognitive impairment. The present review summarizes the mechanisms underlying CEC impairment during hypoperfusion and pathological role of CECs in VaD. Through the comprehensive examination and summarization, endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway, Ras homolog gene family member A (RhoA) signaling pathway, and CEC-derived caveolin-1 (CAV-1) are proposed to serve as targets of new drugs for the treatment of VaD.
Collapse
Affiliation(s)
- Feixue Wang
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wolf Dieter Rausch
- Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
1012
|
Alqawlaq S, Flanagan JG, Sivak JM. All roads lead to glaucoma: Induced retinal injury cascades contribute to a common neurodegenerative outcome. Exp Eye Res 2018; 183:88-97. [PMID: 30447198 DOI: 10.1016/j.exer.2018.11.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
Abstract
Glaucoma describes a distinct optic neuropathy with complex etiology and a variety of associated risk factors, but with similar pathological endpoints. Risk factors such as age, increased intraocular pressure (IOP), low mean arterial pressure, and autoimmune disease, can all be associated with death of retinal ganglion cells (RGCs) and optic nerve head remodeling. Today, IOP management remains the standard of care, even though IOP elevation is not pathognomonic of glaucoma, and patients can continue to lose vision despite effective IOP control. A contemporary view of glaucoma as a complex, neurodegenerative disease has developed, along with the recognition of a need for new disease modifying retinal treatment strategies and improved outcomes. However, the distinction between risk factors triggering the disease process and retinal injury responses is not always clear. In this review, we attempt to distinguish between the various triggers, and their association with subsequent key RGC injury mechanisms. We propose that distinct glaucomatous risk factors result in similar retinal and optic nerve injury cascades, including oxidative and metabolic stress, glial reactivity, and altered inflammatory responses, which induce common molecular signals to induce RGC apoptosis. This organization forms a coherent disease framework and presents conserved targets for therapeutic intervention that are not limited to specific risk factors.
Collapse
Affiliation(s)
- Samih Alqawlaq
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - John G Flanagan
- School of Optometry and Vision Science Program, University of California at Berkeley, Berkeley, CA, USA
| | - Jeremy M Sivak
- Department of Vision Science, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Vision Science Research Program, Ophthalmology and Vision Science, University of Toronto, Toronto, ON, Canada; Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
1013
|
Weinberg RP, Koledova VV, Schneider K, Sambandan TG, Grayson A, Zeidman G, Artamonova A, Sambanthamurthi R, Fairus S, Sinskey AJ, Rha C. Palm Fruit Bioactives modulate human astrocyte activity in vitro altering the cytokine secretome reducing levels of TNFα, RANTES and IP-10. Sci Rep 2018; 8:16423. [PMID: 30401897 PMCID: PMC6219577 DOI: 10.1038/s41598-018-34763-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease, are becoming more prevalent and an increasing burden on society. Neurodegenerative diseases often arise in the milieu of neuro-inflammation of the brain. Reactive astrocytes are key regulators in the development of neuro-inflammation. This study describes the effects of Palm Fruit Bioactives (PFB) on the behavior of human astrocytes which have been activated by IL-1β. When activated, the astrocytes proliferate, release numerous cytokines/chemokines including TNFα, RANTES (CCL5), IP-10 (CXCL10), generate reactive oxygen species (ROS), and express specific cell surface biomarkers such as the Intercellular Adhesion Molecule (ICAM), Vascular Cellular Adhesion Molecule (VCAM) and the Neuronal Cellular Adhesion Molecule (NCAM). Interleukin 1-beta (IL-1β) causes activation of human astrocytes with marked upregulation of pro-inflammatory genes. We show significant inhibition of these pro-inflammatory processes when IL-1β-activated astrocytes are exposed to PFB. PFB causes a dose-dependent and time-dependent reduction in specific cytokines: TNFα, RANTES, and IP-10. We also show that PFB significantly reduces ROS production by IL-1β-activated astrocytes. Furthermore, PFB also reduces the expression of ICAM and VCAM, both in activated and naïve human astrocytes in vitro. Since reactive astrocytes play an essential role in the neuroinflammatory state preceding neurodegenerative diseases, this study suggests that PFB may have a potential role in their prevention and/or treatment.
Collapse
Affiliation(s)
- Robert P Weinberg
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| | - Vera V Koledova
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kirsten Schneider
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - T G Sambandan
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Adlai Grayson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Gal Zeidman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Anastasia Artamonova
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ravigadevi Sambanthamurthi
- Advanced Biotechnology and Breeding Centre, Malaysian Palm Oil Board, 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia
| | - Syed Fairus
- Advanced Biotechnology and Breeding Centre, Malaysian Palm Oil Board, 6, Persiaran Institusi, Bandar Baru Bangi, 43000, Kajang, Selangor, Malaysia
| | - Anthony J Sinskey
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - ChoKyun Rha
- Biomaterials Science and Engineering Laboratory, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
1014
|
A genetic deficiency in folic acid metabolism impairs recovery after ischemic stroke. Exp Neurol 2018; 309:14-22. [DOI: 10.1016/j.expneurol.2018.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/06/2018] [Accepted: 07/24/2018] [Indexed: 01/22/2023]
|
1015
|
Thornton MA, Mehta MD, Morad TT, Ingraham KL, Khankan RR, Griffis KG, Yeung AK, Zhong H, Roy RR, Edgerton VR, Phelps PE. Evidence of axon connectivity across a spinal cord transection in rats treated with epidural stimulation and motor training combined with olfactory ensheathing cell transplantation. Exp Neurol 2018; 309:119-133. [PMID: 30056160 PMCID: PMC6365019 DOI: 10.1016/j.expneurol.2018.07.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/18/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022]
Abstract
Olfactory ensheathing cells (OECs) are unique glia that support axon outgrowth in the olfactory system, and when used as cellular therapy after spinal cord injury, improve recovery and axon regeneration. Here we assessed the effects of combining OEC transplantation with another promising therapy, epidural electrical stimulation during a rehabilitative motor task. Sprague-Dawley rats received a mid-thoracic transection and transplantation of OECs or fibroblasts (FBs) followed by lumbar stimulation while climbing an inclined grid. We injected pseudorabies virus (PRV) into hindlimb muscles 7 months post-injury to assess connectivity across the transection. Analyses showed that the number of serotonergic (5-HT) axons that crossed the rostral scar border and the area of neurofilament-positive axons in the injury site were both greater in OEC- than FB-treated rats. We detected PRV-labeled cells rostral to the transection and remarkable evidence of 5-HT and PRV axons crossing the injury site in 1 OEC- and 1 FB-treated rat. The axons that crossed suggested either axon regeneration (OEC) or small areas of probable tissue sparing (FB). Most PRV-labeled thoracic neurons were detected in laminae VII or X, and ~25% expressed Chx10, a marker for V2a interneurons. These findings suggest potential regeneration or sparing of circuits that connect thoracic interneurons to lumbar somatic motor neurons. Despite evidence of axonal connectivity, no behavioral changes were detected in this small-scale study. Together these data suggest that when supplemented with epidural stimulation and climbing, OEC transplantation can increase axonal growth across the injury site and may promote recovery of propriospinal circuitry.
Collapse
Affiliation(s)
- Michael A Thornton
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States
| | - Manan D Mehta
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States
| | - Tyler T Morad
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States
| | - Kaitlin L Ingraham
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States; Molecular, Cellular and Integrative Physiology, Interdepartmental Ph.D. Program, UCLA, Los Angeles, CA 90095, United States
| | - Rana R Khankan
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States; Molecular, Cellular and Integrative Physiology, Interdepartmental Ph.D. Program, UCLA, Los Angeles, CA 90095, United States
| | - Khris G Griffis
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States; Molecular, Cellular and Integrative Physiology, Interdepartmental Ph.D. Program, UCLA, Los Angeles, CA 90095, United States
| | - Anthony K Yeung
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States
| | - Hui Zhong
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States
| | - Roland R Roy
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States; Brain Research Institute, UCLA, Los Angeles, CA 90095, United States
| | - V Reggie Edgerton
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States; Molecular, Cellular and Integrative Physiology, Interdepartmental Ph.D. Program, UCLA, Los Angeles, CA 90095, United States; Brain Research Institute, UCLA, Los Angeles, CA 90095, United States
| | - Patricia E Phelps
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095, United States; Molecular, Cellular and Integrative Physiology, Interdepartmental Ph.D. Program, UCLA, Los Angeles, CA 90095, United States; Brain Research Institute, UCLA, Los Angeles, CA 90095, United States.
| |
Collapse
|
1016
|
Tran AP, Warren PM, Silver J. The Biology of Regeneration Failure and Success After Spinal Cord Injury. Physiol Rev 2018. [PMID: 29513146 DOI: 10.1152/physrev.00017.2017] [Citation(s) in RCA: 556] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since no approved therapies to restore mobility and sensation following spinal cord injury (SCI) currently exist, a better understanding of the cellular and molecular mechanisms following SCI that compromise regeneration or neuroplasticity is needed to develop new strategies to promote axonal regrowth and restore function. Physical trauma to the spinal cord results in vascular disruption that, in turn, causes blood-spinal cord barrier rupture leading to hemorrhage and ischemia, followed by rampant local cell death. As subsequent edema and inflammation occur, neuronal and glial necrosis and apoptosis spread well beyond the initial site of impact, ultimately resolving into a cavity surrounded by glial/fibrotic scarring. The glial scar, which stabilizes the spread of secondary injury, also acts as a chronic, physical, and chemo-entrapping barrier that prevents axonal regeneration. Understanding the formative events in glial scarring helps guide strategies towards the development of potential therapies to enhance axon regeneration and functional recovery at both acute and chronic stages following SCI. This review will also discuss the perineuronal net and how chondroitin sulfate proteoglycans (CSPGs) deposited in both the glial scar and net impede axonal outgrowth at the level of the growth cone. We will end the review with a summary of current CSPG-targeting strategies that help to foster axonal regeneration, neuroplasticity/sprouting, and functional recovery following SCI.
Collapse
Affiliation(s)
- Amanda Phuong Tran
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Philippa Mary Warren
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| |
Collapse
|
1017
|
Becker CG, Becker T, Hugnot JP. The spinal ependymal zone as a source of endogenous repair cells across vertebrates. Prog Neurobiol 2018; 170:67-80. [DOI: 10.1016/j.pneurobio.2018.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/30/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
|
1018
|
Metwally MMM, Ebraheim LLM, Galal AAA. Potential therapeutic role of melatonin on STZ-induced diabetic central neuropathy: A biochemical, histopathological, immunohistochemical and ultrastructural study. Acta Histochem 2018; 120:828-836. [PMID: 30268437 DOI: 10.1016/j.acthis.2018.09.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
The aim of the present study was to assess the therapeutic potential of melatonin (Mel) in diabetic central neuropathy in a rat model of streptozotocin (STZ)-induced diabetes. The rats were injected with 60 mg/kg STZ and diabetes was confirmed by blood glucose levels (BGL) ≥ 250 mg/dL. Mel treatment (50 mg/kg) was started 72 h before the STZ injection and continued for 45 days. In addition, normal control, vehicle (5% ethanol) control, and Mel-treated non-diabetic control were also included. STZ induced a diabetic phenotype with persistent hyperglycemia and elevated oxidative stress in the brain, liver, and kidneys compared to the control groups. In addition, the diabetic rats showed severe β-cell necrosis with reduced insulin levels, cerebral neuronopathy, myelinopathy, axonopathy, microglial and astroglial activation, and vascular damage. While Mel treatment did not prevent the development of STZ-induced diabetes mellitus and had no significant effect on the BGLs of the diabetic rats, it significantly ameliorated the diabetes-induced oxidative stress and neurodegeneration. Taken together, Mel showed potent therapeutic effects against the neurological complications of hyperglycemia and therefore can be used to treat diabetic neuropathy.
Collapse
Affiliation(s)
- Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Lamiaa L M Ebraheim
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Azza A A Galal
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| |
Collapse
|
1019
|
Pivonkova H, Hermanova Z, Kirdajova D, Awadova T, Malinsky J, Valihrach L, Zucha D, Kubista M, Galisova A, Jirak D, Anderova M. The Contribution of TRPV4 Channels to Astrocyte Volume Regulation and Brain Edema Formation. Neuroscience 2018; 394:127-143. [PMID: 30367945 DOI: 10.1016/j.neuroscience.2018.10.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/01/2022]
Abstract
Transient receptor potential vanilloid type 4 (TRPV4) channels are involved in astrocyte volume regulation; however, only limited data exist about its mechanism in astrocytes in situ. We performed middle cerebral artery occlusion in adult mice, where we found twice larger edema 1 day after the insult in trpv4-/- mice compared to the controls, which was quantified using magnetic resonance imaging. This result suggests disrupted volume regulation in the brain cells in trpv4-/- mice leading to increased edema formation. The aim of our study was to elucidate whether TRPV4 channel-based volume regulation occurs in astrocytes in situ and whether the disrupted volume regulation in trpv4-/- mice might lead to higher edema formation after brain ischemia. For our experiments, we used trpv4-/- mice crossed with transgenic mice expressing enhanced green fluorescent protein (EGFP) under the control of the glial fibrillary acidic protein promoter, which leads to astrocyte visualization by EGFP expression. For quantification of astrocyte volume changes, we used two-dimensional (2D) and three-dimensional (3D) morphometrical approaches and a quantification algorithm based on fluorescence intensity changes during volume alterations induced by hypotonicity or by oxygen-glucose deprivation. In contrast to in vitro experiments, we found little evidence of the contribution of TRPV4 channels to volume regulation in astrocytes in situ in adult mice. Moreover, we only found a rare expression of TRPV4 channels in adult mouse astrocytes. Our data suggest that TRPV4 channels are not involved in astrocyte volume regulation in situ; however, they play a protective role during the ischemia-induced brain edema formation.
Collapse
Affiliation(s)
- Helena Pivonkova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Thuraya Awadova
- Department of Microscopy, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Malinsky
- Department of Microscopy, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prague, Czech Republic; TATAA Biocenter AB, Gothenburg 411 03, Sweden
| | - Andrea Galisova
- Department of Radiodiagnostic and Interventional Radiology, Institute of Clinical and Experimental Medicine, Prague, Czech Republic
| | - Daniel Jirak
- Department of Radiodiagnostic and Interventional Radiology, Institute of Clinical and Experimental Medicine, Prague, Czech Republic; Institute of Biophysics and Informatics, 1st Medicine Faculty, Charles University, Prague, Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic; Department of Neuroscience, 2nd Medical Faculty, Charles University, Prague, Czech Republic.
| |
Collapse
|
1020
|
Yao W, Zhu Y, Zhang X, Sha M, Meng X, Li Z. Semisynthesis of Chondroitin Sulfate E Tetrasaccharide from Hyaluronic Acid. J Org Chem 2018; 83:14069-14077. [DOI: 10.1021/acs.joc.8b01987] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Wang Yao
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Yong Zhu
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Xiao Zhang
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Meng Sha
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Zhongjun Li
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
1021
|
Varadarajan SG, Huberman AD. Assembly and repair of eye-to-brain connections. Curr Opin Neurobiol 2018; 53:198-209. [PMID: 30339988 DOI: 10.1016/j.conb.2018.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/24/2018] [Accepted: 10/02/2018] [Indexed: 12/31/2022]
Abstract
Vision is the sense humans rely on most to navigate the world and survive. A tremendous amount of research has focused on understanding the neural circuits for vision and the developmental mechanisms that establish them. The eye-to-brain, or 'retinofugal' pathway remains a particularly important model in these contexts because it is essential for sight, its overt anatomical features relate to distinct functional attributes and those features develop in a tractable sequence. Much progress has been made in understanding the growth of retinal axons out of the eye, their selection of targets in the brain, the development of laminar and cell type-specific connectivity within those targets, and also dendritic connectivity within the retina itself. Moreover, because the retinofugal pathway is prone to degeneration in many common blinding diseases, understanding the cellular and molecular mechanisms that establish connectivity early in life stands to provide valuable insights into approaches that re-wire this pathway after damage or loss. Here we review recent progress in understanding the development of retinofugal pathways and how this information is important for improving visual circuit regeneration.
Collapse
Affiliation(s)
- Supraja G Varadarajan
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, United States; Department of Ophthalmology, Stanford University School of Medicine, Stanford, United States; BioX, Stanford University School of Medicine, Stanford, United States; Neurosciences Institute, Stanford University School of Medicine, Stanford, United States.
| |
Collapse
|
1022
|
Ceyzériat K, Ben Haim L, Denizot A, Pommier D, Matos M, Guillemaud O, Palomares MA, Abjean L, Petit F, Gipchtein P, Gaillard MC, Guillermier M, Bernier S, Gaudin M, Aurégan G, Joséphine C, Déchamps N, Veran J, Langlais V, Cambon K, Bemelmans AP, Baijer J, Bonvento G, Dhenain M, Deleuze JF, Oliet SHR, Brouillet E, Hantraye P, Carrillo-de Sauvage MA, Olaso R, Panatier A, Escartin C. Modulation of astrocyte reactivity improves functional deficits in mouse models of Alzheimer's disease. Acta Neuropathol Commun 2018; 6:104. [PMID: 30322407 PMCID: PMC6190663 DOI: 10.1186/s40478-018-0606-1] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/24/2018] [Indexed: 11/26/2022] Open
Abstract
Astrocyte reactivity and neuroinflammation are hallmarks of CNS pathological conditions such as Alzheimer’s disease. However, the specific role of reactive astrocytes is still debated. This controversy may stem from the fact that most strategies used to modulate astrocyte reactivity and explore its contribution to disease outcomes have only limited specificity. Moreover, reactive astrocytes are now emerging as heterogeneous cells and all types of astrocyte reactivity may not be controlled efficiently by such strategies. Here, we used cell type-specific approaches in vivo and identified the JAK2-STAT3 pathway, as necessary and sufficient for the induction and maintenance of astrocyte reactivity. Modulation of this cascade by viral gene transfer in mouse astrocytes efficiently controlled several morphological and molecular features of reactivity. Inhibition of this pathway in mouse models of Alzheimer’s disease improved three key pathological hallmarks by reducing amyloid deposition, improving spatial learning and restoring synaptic deficits. In conclusion, the JAK2-STAT3 cascade operates as a master regulator of astrocyte reactivity in vivo. Its inhibition offers new therapeutic opportunities for Alzheimer’s disease.
Collapse
|
1023
|
Ghuman H, Mauney C, Donnelly J, Massensini AR, Badylak SF, Modo M. Biodegradation of ECM hydrogel promotes endogenous brain tissue restoration in a rat model of stroke. Acta Biomater 2018; 80:66-84. [PMID: 30232030 PMCID: PMC6217851 DOI: 10.1016/j.actbio.2018.09.020] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/12/2018] [Accepted: 09/14/2018] [Indexed: 12/15/2022]
Abstract
The brain is considered to have a limited capacity to repair damaged tissue and no regenerative capacity following injury. Tissue lost after a stroke is therefore not spontaneously replaced. Extracellular matrix (ECM)-based hydrogels implanted into the stroke cavity can attract endogenous cells. These hydrogels can be formulated at different protein concentrations that govern their rheological and inductive properties. We evaluated histologically 0, 3, 4 and 8 mg/mL of porcine-derived urinary bladder matrix (UBM)-ECM hydrogel concentrations implanted in a 14-day old stroke cavity. Less concentrated hydrogels (3 and 4 mg/mL) were efficiently degraded with a 95% decrease in volume by 90 days, whereas only 32% of the more concentrated and stiffer hydrogel (8 mg/mL) was resorbed. Macrophage infiltration and density within the bioscaffold progressively increased in the less concentrated hydrogels and decreased in the 8 mg/mL hydrogels. The less concentrated hydrogels showed a robust invasion of endothelial cells with neovascularization. No neovascularization occurred with the stiffer hydrogel. Invasion of neural cells increased with time in all hydrogel concentrations. Differentiation of neural progenitors into mature neurons with axonal projections was evident, as well as a robust invasion of oligodendrocytes. However, relatively few astrocytes were present in the ECM hydrogel, although some were present in the newly forming tissue between degrading scaffold patches. Implantation of an ECM hydrogel partially induced neural tissue restoration, but a more complete understanding is required to evaluate its potential therapeutic application. STATEMENT OF SIGNIFICANCE: Extracellular matrix hydrogel promotes tissue regeneration in many peripheral soft tissues. However, the brain has generally been considered to lack the potential for tissue regeneration. We here demonstrate that tissue regeneration in the brain can be achieved using implantation of ECM hydrogel into a tissue cavity. A structure-function relationship is key to promote tissue regeneration in the brain. Specifically, weaker hydrogels that were retained in the cavity underwent an efficient biodegradation within 14 days post-implantation to promote a tissue restoration within the lesion cavity. In contrast, stiffer ECM hydrogel only underwent minor biodegradation and did not lead to a tissue restoration. Inductive hydrogels weaker than brain tissue provide the appropriate condition to promote an endogenous regenerative response that restores tissue in a cavity. This approach offers new avenues for the future treatment of chronic tissue damage caused by stroke and other acute brain injuries.
Collapse
Affiliation(s)
- Harmanvir Ghuman
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, Pittsburgh, PA, USA
| | | | | | - Andre R Massensini
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Universidade Federal de Minas Gerais, Department of Physiology and Biophysics, Belo Horizonte, Brazil
| | - Stephen F Badylak
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, Pittsburgh, PA, USA; Department of Surgery, Pittsburgh, PA, USA
| | - Michel Modo
- University of Pittsburgh, McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, Pittsburgh, PA, USA; Department of Radiology, Pittsburgh, PA, USA.
| |
Collapse
|
1024
|
Herrera E, Agudo-Barriuso M, Murcia-Belmonte V. Cranial Pair II: The Optic Nerves. Anat Rec (Hoboken) 2018; 302:428-445. [DOI: 10.1002/ar.23922] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/19/2017] [Accepted: 05/14/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Eloísa Herrera
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina; Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIB-Arrixaca); Murcia Spain
| | - Verónica Murcia-Belmonte
- Instituto de Neurociencias de Alicante (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH); Av. Santiago Ramón y Cajal, s/n., 03550 Sant Joan d'Alacant Alicante Spain
| |
Collapse
|
1025
|
Curcio M, Bradke F. Axon Regeneration in the Central Nervous System: Facing the Challenges from the Inside. Annu Rev Cell Dev Biol 2018; 34:495-521. [DOI: 10.1146/annurev-cellbio-100617-062508] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
After an injury in the adult mammalian central nervous system (CNS), lesioned axons fail to regenerate. This failure to regenerate contrasts with axons’ remarkable potential to grow during embryonic development and after an injury in the peripheral nervous system (PNS). Several intracellular mechanisms—including cytoskeletal dynamics, axonal transport and trafficking, signaling and transcription of regenerative programs, and epigenetic modifications—control axon regeneration. In this review, we describe how manipulation of intrinsic mechanisms elicits a regenerative response in different organisms and how strategies are implemented to form the basis of a future regenerative treatment after CNS injury.
Collapse
Affiliation(s)
- Michele Curcio
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany;,
| | - Frank Bradke
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany;,
| |
Collapse
|
1026
|
Liu P, Mai C, Zhang K. Preparation of hydrogels with uniform and gradient chemical structures using dialdehyde cellulose and diamine by aerating ammonia gas. Front Chem Sci Eng 2018. [DOI: 10.1007/s11705-018-1718-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
1027
|
Squair JW, Tigchelaar S, Moon KM, Liu J, Tetzlaff W, Kwon BK, Krassioukov AV, West CR, Foster LJ, Skinnider MA. Integrated systems analysis reveals conserved gene networks underlying response to spinal cord injury. eLife 2018; 7:39188. [PMID: 30277459 PMCID: PMC6173583 DOI: 10.7554/elife.39188] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological condition for which there are currently no effective treatment options to restore function. A major obstacle to the development of new therapies is our fragmentary understanding of the coordinated pathophysiological processes triggered by damage to the human spinal cord. Here, we describe a systems biology approach to integrate decades of small-scale experiments with unbiased, genome-wide gene expression from the human spinal cord, revealing a gene regulatory network signature of the pathophysiological response to SCI. Our integrative analyses converge on an evolutionarily conserved gene subnetwork enriched for genes associated with the response to SCI by small-scale experiments, and whose expression is upregulated in a severity-dependent manner following injury and downregulated in functional recovery. We validate the severity-dependent upregulation of this subnetwork in rodents in primary transcriptomic and proteomic studies. Our analysis provides systems-level view of the coordinated molecular processes activated in response to SCI.
Collapse
Affiliation(s)
- Jordan W Squair
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Seth Tigchelaar
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Kyung-Mee Moon
- Centre for High-Throughput Biology, University of British Columbia, Vancouver, Canada
| | - Jie Liu
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | - Brian K Kwon
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,Department of Orthopaedics, University of British Columbia, Vancouver, Canada
| | - Andrei V Krassioukov
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,GF Strong Rehabilitation Centre, Vancouver Health Authority, Vancouver, Canada.,Department of Medicine, Division of Physical Medicine and Rehabilitation, University of British Columbia, Vancouver, Canada
| | - Christopher R West
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada.,School of Kinesiology, University of British Columbia, Vancouver, Canada
| | - Leonard J Foster
- Centre for High-Throughput Biology, University of British Columbia, Vancouver, Canada.,Department of Biochemistry and Molecular Biology and Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
| | - Michael A Skinnider
- Centre for High-Throughput Biology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
1028
|
Petrova V, Eva R. The Virtuous Cycle of Axon Growth: Axonal Transport of Growth-Promoting Machinery as an Intrinsic Determinant of Axon Regeneration. Dev Neurobiol 2018; 78:898-925. [PMID: 29989351 DOI: 10.1002/dneu.22608] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/25/2018] [Accepted: 05/26/2018] [Indexed: 02/02/2023]
Abstract
Injury to the brain and spinal cord has devastating consequences because adult central nervous system (CNS) axons fail to regenerate. Injury to the peripheral nervous system (PNS) has a better prognosis, because adult PNS neurons support robust axon regeneration over long distances. CNS axons have some regenerative capacity during development, but this is lost with maturity. Two reasons for the failure of CNS regeneration are extrinsic inhibitory molecules, and a weak intrinsic capacity for growth. Extrinsic inhibitory molecules have been well characterized, but less is known about the neuron-intrinsic mechanisms which prevent axon re-growth. Key signaling pathways and genetic/epigenetic factors have been identified which can enhance regenerative capacity, but the precise cellular mechanisms mediating their actions have not been characterized. Recent studies suggest that an important prerequisite for regeneration is an efficient supply of growth-promoting machinery to the axon; however, this appears to be lacking from non-regenerative axons in the adult CNS. In the first part of this review, we summarize the evidence linking axon transport to axon regeneration. We discuss the developmental decline in axon regeneration capacity in the CNS, and comment on how this is paralleled by a similar decline in the selective axonal transport of regeneration-associated receptors such as integrins and growth factor receptors. In the second part, we discuss the mechanisms regulating selective polarized transport within neurons, how these relate to the intrinsic control of axon regeneration, and whether they can be targeted to enhance regenerative capacity. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 00: 000-000, 2018.
Collapse
Affiliation(s)
- Veselina Petrova
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 OPY, United Kingdom
| | - Richard Eva
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 OPY, United Kingdom
| |
Collapse
|
1029
|
Cheng J, Korte N, Nortley R, Sethi H, Tang Y, Attwell D. Targeting pericytes for therapeutic approaches to neurological disorders. Acta Neuropathol 2018; 136:507-523. [PMID: 30097696 PMCID: PMC6132947 DOI: 10.1007/s00401-018-1893-0] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022]
Abstract
Many central nervous system diseases currently lack effective treatment and are often associated with defects in microvascular function, including a failure to match the energy supplied by the blood to the energy used on neuronal computation, or a breakdown of the blood–brain barrier. Pericytes, an under-studied cell type located on capillaries, are of crucial importance in regulating diverse microvascular functions, such as angiogenesis, the blood–brain barrier, capillary blood flow and the movement of immune cells into the brain. They also form part of the “glial” scar isolating damaged parts of the CNS, and may have stem cell-like properties. Recent studies have suggested that pericytes play a crucial role in neurological diseases, and are thus a therapeutic target in disorders as diverse as stroke, traumatic brain injury, migraine, epilepsy, spinal cord injury, diabetes, Huntington’s disease, Alzheimer’s disease, diabetes, multiple sclerosis, glioma, radiation necrosis and amyotrophic lateral sclerosis. Here we report recent advances in our understanding of pericyte biology and discuss how pericytes could be targeted to develop novel therapeutic approaches to neurological disorders, by increasing blood flow, preserving blood–brain barrier function, regulating immune cell entry to the CNS, and modulating formation of blood vessels in, and the glial scar around, damaged regions.
Collapse
Affiliation(s)
- Jinping Cheng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Rd, Guangzhou, 510120, People's Republic of China
| | - Nils Korte
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ross Nortley
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Huma Sethi
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Xi Rd, Guangzhou, 510120, People's Republic of China.
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
1030
|
Liu B, Mosienko V, Vaccari Cardoso B, Prokudina D, Huentelman M, Teschemacher AG, Kasparov S. Glio- and neuro-protection by prosaposin is mediated by orphan G-protein coupled receptors GPR37L1 and GPR37. Glia 2018; 66:2414-2426. [PMID: 30260505 PMCID: PMC6492175 DOI: 10.1002/glia.23480] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/30/2018] [Accepted: 06/04/2018] [Indexed: 02/01/2023]
Abstract
Discovery of neuroprotective pathways is one of the major priorities for neuroscience. Astrocytes are natural neuroprotectors and it is likely that brain resilience can be enhanced by mobilizing their protective potential. Among G‐protein coupled receptors expressed by astrocytes, two highly related receptors, GPR37L1 and GPR37, are of particular interest. Previous studies suggested that these receptors are activated by a peptide Saposin C and its neuroactive fragments (prosaptide TX14(A)), which were demonstrated to be neuroprotective in various animal models by several groups. However, pairing of Saposin C or prosaptides with GPR37L1/GPR37 has been challenged and presently GPR37L1/GPR37 have regained their orphan status. Here, we demonstrate that in their natural habitat, astrocytes, these receptors mediate a range of effects of TX14(A), including protection from oxidative stress. The Saposin C/GPR37L1/GPR37 pathway is also involved in the neuroprotective effect of astrocytes on neurons subjected to oxidative stress. The action of TX14(A) is at least partially mediated by Gi‐proteins and the cAMP‐PKA axis. On the other hand, when recombinant GPR37L1 or GPR37 are expressed in HEK293 cells, they are not functional and do not respond to TX14(A), which explains unsuccessful attempts to confirm the ligand‐receptor pairing. Therefore, this study identifies GPR37L1/GPR37 as the receptors for TX14(A), and, by extension of Saposin C, and paves the way for the development of neuroprotective therapeutics acting via these receptors. A video abstract of this article can be found at: https://www.youtube.com/watch?v=qTn13My9Sz8
Collapse
Affiliation(s)
- Beihui Liu
- Department of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Valentina Mosienko
- Department of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Barbara Vaccari Cardoso
- Department of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | | | | | - Anja G Teschemacher
- Department of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Sergey Kasparov
- Department of Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
1031
|
Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ, Medeiros R. Inflammation: the link between comorbidities, genetics, and Alzheimer's disease. J Neuroinflammation 2018; 15:276. [PMID: 30249283 PMCID: PMC6154824 DOI: 10.1186/s12974-018-1313-3] [Citation(s) in RCA: 364] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder, most cases of which lack a clear causative event. This has made the disease difficult to characterize and, thus, diagnose. Although some cases are genetically linked, there are many diseases and lifestyle factors that can lead to an increased risk of developing AD, including traumatic brain injury, diabetes, hypertension, obesity, and other metabolic syndromes, in addition to aging. Identifying common factors and trends between these conditions could enhance our understanding of AD and lead to the development of more effective treatments. Although the immune system is one of the body’s key defense mechanisms, chronic inflammation has been increasingly linked with several age-related diseases. Moreover, it is now well accepted that chronic inflammation has an important role in the onset and progression of AD. In this review, the different inflammatory signals associated with AD and its risk factors will be outlined to demonstrate how chronic inflammation may be influencing individual susceptibility to AD. Our goal is to bring attention to potential shared signals presented by the immune system during different conditions that could lead to the development of successful treatments.
Collapse
Affiliation(s)
- Estella A Newcombe
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| | - Judith Camats-Perna
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Mallone L Silva
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Nicholas Valmas
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Tee Jong Huat
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.,Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Rodrigo Medeiros
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| |
Collapse
|
1032
|
He X, Liu Y, Lin X, Yuan F, Long D, Zhang Z, Wang Y, Xuan A, Yang GY. Netrin-1 attenuates brain injury after middle cerebral artery occlusion via downregulation of astrocyte activation in mice. J Neuroinflammation 2018; 15:268. [PMID: 30227858 PMCID: PMC6145326 DOI: 10.1186/s12974-018-1291-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/26/2018] [Indexed: 12/21/2022] Open
Abstract
Background Netrin-1 functions largely via combined receptors and downstream effectors. Evidence has shown that astrocytes express netrin-1 receptors, including DCC and UNC5H2. However, whether netrin-1 influences the function of astrocytes was previously unknown. Methods Lipopolysaccharide was used to stimulate the primary cultured astrocytes; interleukin release was used to track astrocyte activation. In vivo, shRNA and netrin-1 protein were injected in the mouse brain. Infarct volume, astrocyte activation, and interleukin release were used to observe the function of netrin-1 in neuroinflammation and brain injury after middle cerebral artery occlusion. Results Our results demonstrated that netrin-1 reduced lipopolysaccharide-induced interleukin-1β and interleukin-12β release in cultured astrocytes, and blockade of the UNC5H2 receptor with an antibody reversed this effect. Additionally, netrin-1 increased p-AKT and PPAR-γ expression in primary cultured astrocytes. In vivo studies showed that knockdown of netrin-1 increased astrocyte activation in the mouse brain after middle cerebral artery occlusion (p < 0.05). Moreover, injection of netrin-1 attenuated GFAP expression (netrin-1 0.27 ± 0.06 vs. BSA 0.62 ± 0.04, p < 0.001) and the release of interleukins and reduced infarct volume after brain ischemia (netrin-1 0.27 ± 0.06 vs. BSA 0.62 ± 0.04 mm3, p < 0.05). Conclusion Our results indicate that netrin-1 is an important molecule in regulating astrocyte activation and neuroinflammation in cerebral ischemia and provides a potential target for ischemic stroke therapy. Electronic supplementary material The online version of this article (10.1186/s12974-018-1291-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaosong He
- Key Laboratory of Neuroscience, the Second Affiliated Hospital Guangzhou Medical University, Guangzhou, China.,Department of Anatomy, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yanqun Liu
- Department of Neurology, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Xiaohong Lin
- Key Laboratory of Neuroscience, the Second Affiliated Hospital Guangzhou Medical University, Guangzhou, China.,Department of Anatomy, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Falei Yuan
- Hailisheng Biomarine Research Institute, Zhoushan, China
| | - Dahong Long
- Key Laboratory of Neuroscience, the Second Affiliated Hospital Guangzhou Medical University, Guangzhou, China.,Department of Anatomy, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Aiguo Xuan
- Key Laboratory of Neuroscience, the Second Affiliated Hospital Guangzhou Medical University, Guangzhou, China. .,Department of Anatomy, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China. .,Department of Anatomy, Guangzhou Medical college, Guangzhou, 511546, China.
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China. .,Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China. .,Med-X Research Institute and School of Biomedical Engineering, 1954 Hua-shan Road, Shanghai, 200030, China.
| |
Collapse
|
1033
|
Hausott B, Klimaschewski L. Sprouty2-a Novel Therapeutic Target in the Nervous System? Mol Neurobiol 2018; 56:3897-3903. [PMID: 30225774 PMCID: PMC6505497 DOI: 10.1007/s12035-018-1338-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/29/2018] [Indexed: 02/08/2023]
Abstract
Clinical trials applying growth factors to alleviate symptoms of patients with neurological disorders have largely been unsuccessful in the past. As an alternative approach, growth factor receptors or components of their signal transduction machinery may be targeted directly. In recent years, the search for intracellular signaling integrator downstream of receptor tyrosine kinases provided valuable novel substrates. Among them are the Sprouty proteins which mainly act as inhibitors of growth factor-dependent neuronal and glial signaling pathways. In this review, we summarize the role of Sprouties in the lesioned central and peripheral nervous system with particular reference to Sprouty2 that is upregulated in various experimental models of neuronal degeneration and regeneration. Increased synthesis under pathological conditions makes Sprouty2 an attractive pharmacological target to enhance intracellular signaling activities, notably the ERK pathway, in affected neurons or activated astrocytes. Interestingly, high Sprouty2 levels are also found in malignant glioma cells. We recently demonstrated that abrogating Sprouty2 function strongly inhibits intracranial tumor growth and leads to significantly prolonged survival of glioblastoma bearing mice by induction of ERK-dependent DNA replication stress. On the contrary, knockdown of Sprouty proteins increases proliferation of activated astrocytes and, consequently, reduces secondary brain damage in neuronal lesion models such as kainic acid-induced epilepsy or endothelin-induced ischemia. Furthermore, downregulation of Sprouty2 improves nerve regeneration in the lesioned peripheral nervous system. Taken together, targeting Sprouties as intracellular inhibitors of the ERK pathway holds great promise for the treatment of various neurological disorders including gliomas. Since the protein lacks enzymatic activities, it will be difficult to develop chemical compounds capable to directly and specifically modulate Sprouty functions. However, interfering with Sprouty expression by gene therapy or siRNA treatment provides a realistic approach to evaluate the therapeutic potential of indirectly stimulating ERK activities in neurological disease.
Collapse
Affiliation(s)
- Barbara Hausott
- Department of Anatomy, Histology and Embyrology, Division of Neuroanatomy, Medical University Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria
| | - Lars Klimaschewski
- Department of Anatomy, Histology and Embyrology, Division of Neuroanatomy, Medical University Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria. .,Division for Neuroanatomy, Medical University of Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria.
| |
Collapse
|
1034
|
Patel R, Muir M, Cvetkovic C, Krencik R. Concepts toward directing human astroplasticity to promote neuroregeneration. Dev Dyn 2018; 248:21-33. [DOI: 10.1002/dvdy.24655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
| | | | - Caroline Cvetkovic
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery; Houston Methodist Research Institute; Houston Texas
| |
Collapse
|
1035
|
Wen J, Maxwell RR, Wolf AJ, Spira M, Gulinello ME, Cole PD. Methotrexate causes persistent deficits in memory and executive function in a juvenile animal model. Neuropharmacology 2018; 139:76-84. [PMID: 29990472 PMCID: PMC6089371 DOI: 10.1016/j.neuropharm.2018.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/27/2018] [Accepted: 07/05/2018] [Indexed: 01/22/2023]
Abstract
Methotrexate is a dihydrofolate reductase inhibitor widely employed in curative treatment for children with acute lymphoblastic leukemia (ALL). However, methotrexate administration is also associated with persistent cognitive deficits among long-term childhood cancer survivors. Animal models of methotrexate-induced cognitive deficits have primarily utilized adult animals. The purpose of present study is to investigate the neurotoxicity of methotrexate in juvenile rats and its relevant mechanisms. The doses and schedule of systemic and intrathecal methotrexate, given from post-natal age 3-7 weeks, were chosen to model the effects of repeated methotrexate dosing on the developing brains of young children with ALL. This methotrexate regimen had no visible acute toxicity and no effect on growth. At 15 weeks of age (8 weeks after the last methotrexate dose) both spatial pattern memory and visual recognition memory were impaired. In addition, methotrexate-treated animals demonstrated impaired performance in the set-shifting assay, indicating decreased cognitive flexibility. Histopathological analysis demonstrated decreased cell proliferation in methotrexate-treated animals compared to controls, as well as changes in length and thickness of the corpus callosum. Moreover, methotrexate suppressed microglia activation and RANTES production. In conclusion, our study demonstrated that a clinically relevant regimen of systemic and intrathecal methotrexate induces persistent deficits in spatial pattern memory, visual recognition memory and executive function, lasting at least 8 weeks after the last injection. The mechanisms behind methotrexate-induced deficits are likely multifactorial and may relate to suppression of neurogenesis, alterations in neuroinflammation and microglial activation, and structural changes in the corpus callosum.
Collapse
Affiliation(s)
- Jing Wen
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rochelle R Maxwell
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alexander J Wolf
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Menachem Spira
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Maria E Gulinello
- Behavioral Core Facility, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Peter D Cole
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA; Rutgers Cancer Institute of New Jersey, Division of Pediatric Hematology/Oncology, New Brunswick, NJ, USA.
| |
Collapse
|
1036
|
Wollenberg AL, O'Shea TM, Kim JH, Czechanski A, Reinholdt LG, Sofroniew MV, Deming TJ. Injectable polypeptide hydrogels via methionine modification for neural stem cell delivery. Biomaterials 2018; 178:527-545. [PMID: 29657091 PMCID: PMC6054810 DOI: 10.1016/j.biomaterials.2018.03.057] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/11/2018] [Accepted: 03/31/2018] [Indexed: 12/21/2022]
Abstract
Injectable hydrogels with tunable physiochemical and biological properties are potential tools for improving neural stem/progenitor cell (NSPC) transplantation to treat central nervous system (CNS) injury and disease. Here, we developed injectable diblock copolypeptide hydrogels (DCH) for NSPC transplantation that contain hydrophilic segments of modified l-methionine (Met). Multiple Met-based DCH were fabricated by post-polymerization modification of Met to various functional derivatives, and incorporation of different amino acid comonomers into hydrophilic segments. Met-based DCH assembled into self-healing hydrogels with concentration and composition dependent mechanical properties. Mechanical properties of non-ionic Met-sulfoxide formulations (DCHMO) were stable across diverse aqueous media while cationic formulations showed salt ion dependent stiffness reduction. Murine NSPC survival in DCHMO was equivalent to that of standard culture conditions, and sulfoxide functionality imparted cell non-fouling character. Within serum rich environments in vitro, DCHMO was superior at preserving NSPC stemness and multipotency compared to cell adhesive materials. NSPC in DCHMO injected into uninjured forebrain remained local and, after 4 weeks, exhibited an immature astroglial phenotype that integrated with host neural tissue and acted as cellular substrates that supported growth of host-derived axons. These findings demonstrate that Met-based DCH are suitable vehicles for further study of NSPC transplantation in CNS injury and disease models.
Collapse
Affiliation(s)
- A L Wollenberg
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1600, USA
| | - T M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA
| | - J H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA
| | - A Czechanski
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | - M V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA
| | - T J Deming
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1600, USA.
| |
Collapse
|
1037
|
It is time to remove the stigma from glial scars: re: Lentivirus-mediated silencing of the CTGF gene suppresses the formation of glial scar tissue in a rat model of spinal cord injury. Spine J 2018; 18:1724-1725. [PMID: 30220540 DOI: 10.1016/j.spinee.2018.05.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/01/2018] [Indexed: 02/03/2023]
|
1038
|
Anderson MA, O'Shea TM, Burda JE, Ao Y, Barlatey SL, Bernstein AM, Kim JH, James ND, Rogers A, Kato B, Wollenberg AL, Kawaguchi R, Coppola G, Wang C, Deming TJ, He Z, Courtine G, Sofroniew MV. Required growth facilitators propel axon regeneration across complete spinal cord injury. Nature 2018; 561:396-400. [PMID: 30158698 PMCID: PMC6151128 DOI: 10.1038/s41586-018-0467-6] [Citation(s) in RCA: 349] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022]
Abstract
Transected axons fail to regrow across anatomically complete spinal cord injuries (SCI) in adults. Diverse molecules can partially facilitate or attenuate axon growth during development or after injury1-3, but efficient reversal of this regrowth failure remains elusive4. Here we show that three factors that are essential for axon growth during development but are attenuated or lacking in adults-(i) neuron intrinsic growth capacity2,5-9, (ii) growth-supportive substrate10,11 and (iii) chemoattraction12,13-are all individually required and, in combination, are sufficient to stimulate robust axon regrowth across anatomically complete SCI lesions in adult rodents. We reactivated the growth capacity of mature descending propriospinal neurons with osteopontin, insulin-like growth factor 1 and ciliary-derived neurotrophic factor before SCI14,15; induced growth-supportive substrates with fibroblast growth factor 2 and epidermal growth factor; and chemoattracted propriospinal axons with glial-derived neurotrophic factor16,17 delivered via spatially and temporally controlled release from biomaterial depots18,19, placed sequentially after SCI. We show in both mice and rats that providing these three mechanisms in combination, but not individually, stimulated robust propriospinal axon regrowth through astrocyte scar borders and across lesion cores of non-neural tissue that was over 100-fold greater than controls. Stimulated, supported and chemoattracted propriospinal axons regrew a full spinal segment beyond lesion centres, passed well into spared neural tissue, formed terminal-like contacts exhibiting synaptic markers and conveyed a significant return of electrophysiological conduction capacity across lesions. Thus, overcoming the failure of axon regrowth across anatomically complete SCI lesions after maturity required the combined sequential reinstatement of several developmentally essential mechanisms that facilitate axon growth. These findings identify a mechanism-based biological repair strategy for complete SCI lesions that could be suitable to use with rehabilitation models designed to augment the functional recovery of remodelling circuits.
Collapse
Affiliation(s)
- Mark A Anderson
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Timothy M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joshua E Burda
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sabry L Barlatey
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Alexander M Bernstein
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jae H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nicholas D James
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Alexandra Rogers
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Brian Kato
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexander L Wollenberg
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Departments of Psychiatry and Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Giovanni Coppola
- Departments of Psychiatry and Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chen Wang
- F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy J Deming
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gregoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
1039
|
Grainger AI, King MC, Nagel DA, Parri HR, Coleman MD, Hill EJ. In vitro Models for Seizure-Liability Testing Using Induced Pluripotent Stem Cells. Front Neurosci 2018; 12:590. [PMID: 30233290 PMCID: PMC6127295 DOI: 10.3389/fnins.2018.00590] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/06/2018] [Indexed: 12/14/2022] Open
Abstract
The brain is the most complex organ in the body, controlling our highest functions, as well as regulating myriad processes which incorporate the entire physiological system. The effects of prospective therapeutic entities on the brain and central nervous system (CNS) may potentially cause significant injury, hence, CNS toxicity testing forms part of the “core battery” of safety pharmacology studies. Drug-induced seizure is a major reason for compound attrition during drug development. Currently, the rat ex vivo hippocampal slice assay is the standard option for seizure-liability studies, followed by primary rodent cultures. These models can respond to diverse agents and predict seizure outcome, yet controversy over the relevance, efficacy, and cost of these animal-based methods has led to interest in the development of human-derived models. Existing platforms often utilize rodents, and so lack human receptors and other drug targets, which may produce misleading data, with difficulties in inter-species extrapolation. Current electrophysiological approaches are typically used in a low-throughput capacity and network function may be overlooked. Human-derived induced pluripotent stem cells (iPSCs) are a promising avenue for neurotoxicity testing, increasingly utilized in drug screening and disease modeling. Furthermore, the combination of iPSC-derived models with functional techniques such as multi-electrode array (MEA) analysis can provide information on neuronal network function, with increased sensitivity to neurotoxic effects which disrupt different pathways. The use of an in vitro human iPSC-derived neural model for neurotoxicity studies, combined with high-throughput techniques such as MEA recordings, could be a suitable addition to existing pre-clinical seizure-liability testing strategies.
Collapse
Affiliation(s)
| | - Marianne C King
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - David A Nagel
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - H Rheinallt Parri
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Michael D Coleman
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Eric J Hill
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| |
Collapse
|
1040
|
Yu B, Gu X. Combination of biomaterial transplantation and genetic enhancement of intrinsic growth capacities to promote CNS axon regeneration after spinal cord injury. Front Med 2018; 13:131-137. [PMID: 30159670 DOI: 10.1007/s11684-018-0642-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/26/2018] [Indexed: 12/16/2022]
Abstract
The inhibitory environment that surrounds the lesion site and the lack of intrinsic regenerative capacity of the adult mammalian central nervous system (CNS) impede the regrowth of injured axons and thereby the reestablishment of neural circuits required for functional recovery after spinal cord injuries (SCI). To circumvent these barriers, biomaterial scaffolds are applied to bridge the lesion gaps for the regrowing axons to follow, and, often by combining stem cell transplantation, to enable the local environment in the growth-supportive direction. Manipulations, such as the modulation of PTEN/mTOR pathways, can also enhance intrinsic CNS axon regrowth after injury. Given the complex pathophysiology of SCI, combining biomaterial scaffolds and genetic manipulation may provide synergistic effects and promote maximal axonal regrowth. Future directions will primarily focus on the translatability of these approaches and promote therapeutic avenues toward the functional rehabilitation of patients with SCIs.
Collapse
Affiliation(s)
- Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
1041
|
Janowska J, Gargas J, Ziemka-Nalecz M, Zalewska T, Buzanska L, Sypecka J. Directed glial differentiation and transdifferentiation for neural tissue regeneration. Exp Neurol 2018; 319:112813. [PMID: 30171864 DOI: 10.1016/j.expneurol.2018.08.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 02/06/2023]
Abstract
Glial cells which are indispensable for the central nervous system development and functioning, are proven to be vulnerable to a harmful influence of pathological cues and tissue misbalance. However, they are also highly sensitive to both in vitro and in vivo modulation of their commitment, differentiation, activity and even the fate-switch by different types of bioactive molecules. Since glial cells (comprising macroglia and microglia) are an abundant and heterogeneous population of neural cells, which are almost uniformly distributed in the brain and the spinal cord parenchyma, they all create a natural endogenous reservoir of cells for potential neurogenerative processes required to be initiated in response to pathophysiological cues present in the local tissue microenvironment. The past decade of intensive investigation on a spontaneous and enforced conversion of glial fate into either alternative glial (for instance from oligodendrocytes to astrocytes) or neuronal phenotypes, has considerably extended our appreciation of glial involvement in restoring the nervous tissue cytoarchitecture and its proper functions. The most effective modulators of reprogramming processes have been identified and tested in a series of pre-clinical experiments. A list of bioactive compounds which are potent in guiding in vivo cell fate conversion and driving cell differentiation includes a selection of transcription factors, microRNAs, small molecules, exosomes, morphogens and trophic factors, which are helpful in boosting the enforced neuro-or gliogenesis and promoting the subsequent cell maturation into desired phenotypes. Herein, an issue of their utility for a directed glial differentiation and transdifferentiation is discussed in the context of elaborating future therapeutic options aimed at restoring the diseased nervous tissue.
Collapse
Affiliation(s)
- Justyna Janowska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Justyna Gargas
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Malgorzata Ziemka-Nalecz
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Teresa Zalewska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Leonora Buzanska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Stem Cell Bioengineering Unit, 5, Pawinskiego str., 02-106 Warsaw, Poland
| | - Joanna Sypecka
- Mossakowski Medical Research Centre, Polish Academy of Sciences, NeuroRepair Department, 5, Pawinskiego str., 02-106 Warsaw, Poland.
| |
Collapse
|
1042
|
Wu Q, Wang Q, Li Z, Li X, Zang J, Wang Z, Xu C, Gong Y, Cheng J, Li H, Shen G, Dong C. Human menstrual blood-derived stem cells promote functional recovery in a rat spinal cord hemisection model. Cell Death Dis 2018; 9:882. [PMID: 30158539 PMCID: PMC6115341 DOI: 10.1038/s41419-018-0847-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 12/21/2022]
Abstract
Spinal cord injury (SCI) is associated with a dismal prognosis including severe voluntary motor and sensory deficits in the presence of the current therapies, thus new and efficient treatment strategies are desperately required. Along with several advantages, such as easy accessibility, high-yield, potential of enormous proliferation, menstrual blood-derived mesenchymal stem cells (MenSCs) have been proposed as a promising strategy in regeneration medicine. In this study, the MenSCs were transplanted into incomplete thoracic (T10) spinal cord injury (SCI) rats, all rats were sacrificed at 7, 14, and 28 days after surgery. Based on the results, we found that MenSCs transplantation improved the hind limb motor function. Besides, H&E staining showed that MenSCs treatment markedly reduced cavity formation in the lesion site. Furthermore, treatment by MenSCs showed more MAP2-positive mature neurons, as well as axonal regeneration manifested by NF-200 and less expression of chondroitin sulfate proteoglycans (CSPGs) than the non-treatment in the lesion site. Additionally, immunofluorescence, Western blot, and qRT-PCR methods showed that levels of brain-derived neurotrophic factor (BDNF) were significantly higher in the injured spinal cord after implantation of MenSCs. Results of qRT-PCR indicated that inflammatory factors, including TNF-α and IL-1β were inhibited after MenSCs transplantation. The improved motor function of hind limb and the increased cell body area of motor neurons were suppressed by blocking of the BDNF-TrkB signaling. It was eventually revealed that MenSCs implantation had beneficial therapeutic effects on the rehabilitation of the rat spinal cord hemisection model, mainly by enhancing the expression of BDNF. MenSCs transplantation may provide a novel therapeutic strategy for patients with SCI in the future.
Collapse
Affiliation(s)
- Qinfeng Wu
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China.,Department of Rehabilitation Medicine, Suzhou Hospital affiliated to Nanjing Medical University, Suzhou Science & Technology Town Hospital, 215153, Suzhou, Jiangsu Province, China
| | - Qinghua Wang
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Zhangjie Li
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu Province, China.,Department of Rehabilitation Medicine, Zhangjiagang First People's Hospital, 215600, Zhangjiagang, Jiangsu Province, China
| | - Xiangzhe Li
- Department of Rehabilitation Medicine, Suzhou Hospital affiliated to Nanjing Medical University, Suzhou Science & Technology Town Hospital, 215153, Suzhou, Jiangsu Province, China
| | - Jing Zang
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu Province, China
| | - Zhangwei Wang
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Chen Xu
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yujia Gong
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Jiaqi Cheng
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Haoming Li
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Guangyu Shen
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, 226001, Nantong, Jiangsu Province, China
| | - Chuanming Dong
- Department of Anatomy, Medical School of Nantong University, Laboratory Animal Center of Nantong University, Nantong, Jiangsu Province, 226001, China.
| |
Collapse
|
1043
|
Schiweck J, Eickholt BJ, Murk K. Important Shapeshifter: Mechanisms Allowing Astrocytes to Respond to the Changing Nervous System During Development, Injury and Disease. Front Cell Neurosci 2018; 12:261. [PMID: 30186118 PMCID: PMC6111612 DOI: 10.3389/fncel.2018.00261] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/31/2018] [Indexed: 12/30/2022] Open
Abstract
Astrocytes are the most prevalent glial cells in the brain. Historically considered as “merely supporting” neurons, recent research has shown that astrocytes actively participate in a large variety of central nervous system (CNS) functions including synaptogenesis, neuronal transmission and synaptic plasticity. During disease and injury, astrocytes efficiently protect neurons by various means, notably by sealing them off from neurotoxic factors and repairing the blood-brain barrier. Their ramified morphology allows them to perform diverse tasks by interacting with synapses, blood vessels and other glial cells. In this review article, we provide an overview of how astrocytes acquire their complex morphology during development. We then move from the developing to the mature brain, and review current research on perisynaptic astrocytic processes, with a particular focus on how astrocytes engage synapses and modulate their formation and activity. Comprehensive changes have been reported in astrocyte cell shape in many CNS pathologies. Factors influencing these morphological changes are summarized in the context of brain pathologies, such as traumatic injury and degenerative conditions. We provide insight into the molecular, cellular and cytoskeletal machinery behind these shape changes which drive the dynamic remodeling in astrocyte morphology during injury and the development of pathologies.
Collapse
Affiliation(s)
- Juliane Schiweck
- Institute for Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Britta J Eickholt
- Institute for Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Kai Murk
- Institute for Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
1044
|
Li X, Li J, Xiao Z, Dai J. [The role of glial scar on axonal regeneration after spinal cord injury]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:973-978. [PMID: 30238720 DOI: 10.7507/1002-1892.201806093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The 'glial scar' has been widely studied in the regeneration of spinal cord injury (SCI). For decades, mainstream scientific concept considers glial scar as a 'physical barrier' to impede axonal regeneration after SCI. Moreover, some extracellular molecules produced by glial scar are also regarded as axonal growth inhibitors. With the development of technology and the progress of research, multiple lines of new evidence challenge the pre-existing traditional notions in SCI repair, including the role of glial scar. This review briefly reviewed the history, advance, and controversy of glial scar research in SCI repair since 1930s, hoping to recognize the roles of glial scar and crack the international problem of SCI regeneration.
Collapse
Affiliation(s)
- Xing Li
- Institute of Genetics and Development Biology, Chinese Academy of Sciences, Beijing, 100101, P.R.China
| | - Jiayin Li
- Institute of Genetics and Development Biology, Chinese Academy of Sciences, Beijing, 100101, P.R.China
| | - Zhifeng Xiao
- Institute of Genetics and Development Biology, Chinese Academy of Sciences, Beijing, 100101, P.R.China
| | - Jianwu Dai
- Institute of Genetics and Development Biology, Chinese Academy of Sciences, Beijing, 100101,
| |
Collapse
|
1045
|
Noristani HN, Saint-Martin GP, Cardoso M, Sidiboulenouar R, Catteau M, Coillot C, Goze-Bac C, Perrin FE. Longitudinal Magnetic Resonance Imaging Analysis and Histological Characterization after Spinal Cord Injury in Two Mouse Strains with Different Functional Recovery: Gliosis as a Key Factor. J Neurotrauma 2018; 35:2924-2940. [PMID: 29877129 DOI: 10.1089/neu.2017.5613] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injuries (SCI) are disastrous neuropathologies causing permanent disabilities. The availability of different strains of mice is valuable for studying the pathophysiological mechanisms involved in SCI. However, strain differences have a profound effect on spontaneous functional recovery after SCI. CX3CR1+/eGFP and Aldh1l1-EGFP mice that express green fluorescent protein in microglia/monocytes and astrocytes, respectively, are particularly useful to study glial reactivity. Whereas CX3CR1+/eGFP mice have C57BL/6 background, Aldh1l1-EGFP are in Swiss Webster background. We first assessed spontaneous functional recovery in CX3CR1+/eGFP and Aldh1l1-EGFP mice over 6 weeks after lateral spinal cord hemisection. Second, we carried out a longitudinal follow-up of lesion evolution using in vivo T2-weighted magnetic resonance imaging (MRI). Finally, we performed in-depth analysis of the spinal cord tissue using ex vivo T2-weighted MRI as well as detailed histology. We demonstrate that CX3CR1+/eGFP mice have improved functional recovery and reduced anxiety after SCI compared with Aldh1l1-EGFP mice. We also found a strong correlation between in vivo MRI, ex vivo MRI, and histological analyses of the injured spinal cord in both strain of mice. All three modalities revealed no difference in lesion extension and volume between the two strains of mice. Importantly, histopathological analysis identified decreased gliosis and increased serotonergic axons in CX3CR1+/eGFP compared with Aldh1l1-EGFP mice following SCI. These results thus suggest that the strain-dependent improved functional recovery after SCI may be linked with reduced gliosis and increased serotonergic innervation.
Collapse
Affiliation(s)
- Harun N Noristani
- 1 INSERM U1198, University of Montpellier, Montpellier, France.,2 INSERM U1051, University of Montpellier, Montpellier, France
| | - Guillaume P Saint-Martin
- 1 INSERM U1198, University of Montpellier, Montpellier, France.,3 UMR 5221 CNRS, University of Montpellier, Montpellier, France
| | - Maïda Cardoso
- 2 INSERM U1051, University of Montpellier, Montpellier, France.,3 UMR 5221 CNRS, University of Montpellier, Montpellier, France
| | | | | | | | | | - Florence E Perrin
- 1 INSERM U1198, University of Montpellier, Montpellier, France.,2 INSERM U1051, University of Montpellier, Montpellier, France.,3 UMR 5221 CNRS, University of Montpellier, Montpellier, France
| |
Collapse
|
1046
|
Saba J, Turati J, Ramírez D, Carniglia L, Durand D, Lasaga M, Caruso C. Astrocyte truncated tropomyosin receptor kinase B mediates brain-derived neurotrophic factor anti-apoptotic effect leading to neuroprotection. J Neurochem 2018; 146:686-702. [DOI: 10.1111/jnc.14476] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/22/2018] [Accepted: 05/25/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Julieta Saba
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET; Paraguay 2155; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Juan Turati
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET; Paraguay 2155; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Delia Ramírez
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET; Paraguay 2155; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Lila Carniglia
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET; Paraguay 2155; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET; Paraguay 2155; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET; Paraguay 2155; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas (INBIOMED) UBA-CONICET; Paraguay 2155; Facultad de Medicina; Universidad de Buenos Aires; Buenos Aires Argentina
| |
Collapse
|
1047
|
Wang Q, Zhang H, Xu H, Zhao Y, Li Z, Li J, Wang H, Zhuge D, Guo X, Xu H, Jones S, Li X, Jia X, Xiao J. Novel multi-drug delivery hydrogel using scar-homing liposomes improves spinal cord injury repair. Am J Cancer Res 2018; 8:4429-4446. [PMID: 30214630 PMCID: PMC6134929 DOI: 10.7150/thno.26717] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/11/2018] [Indexed: 12/15/2022] Open
Abstract
Proper selection and effective delivery of combination drugs targeting multiple pathophysiological pathways key to spinal cord injury (SCI) hold promise to address the thus far scarce clinical therapeutics for improving recovery after SCI. In this study, we aim to develop a clinically feasible way for targeted delivery of multiple drugs with different physiochemical properties to the SCI site, detail the underlying mechanism of neural recovery, and detect any synergistic effect related to combination therapy. Methods: Liposomes (LIP) modified with a scar-targeted tetrapeptide (cysteine-alanine-glutamine-lysine, CAQK) were first constructed to simultaneously encapsulate docetaxel (DTX) and brain-derived neurotrophic factor (BDNF) and then were further added into a thermosensitive heparin-modified poloxamer hydrogel (HP) with affinity-bound acidic fibroblast growth factor (aFGF-HP) for local administration into the SCI site (CAQK-LIP-GFs/DTX@HP) in a rat model. In vivo fluorescence imaging was used to examine the specificity of CAQK-LIP-GFs/DTX binding to the injured site. Multiple comprehensive evaluations including biotin dextran amine anterograde tracing and magnetic resonance imaging were used to detect any synergistic effects and the underlying mechanisms of CAQK-LIP-GFs/DTX@HP both in vivo (rat SCI model) and in vitro (primary neuron). Results: The multiple drugs were effectively delivered to the injured site. The combined application of GFs and DTX supported neuro-regeneration by improving neuronal survival and plasticity, rendering a more permissive extracellular matrix environment with improved regeneration potential. In addition, our combination therapy promoted axonal regeneration via moderation of microtubule function and mitochondrial transport along the regenerating axon. Conclusion: This novel multifunctional therapeutic strategy with a scar-homing delivery system may offer promising translational prospects for the clinical treatment of SCI.
Collapse
|
1048
|
Topological remodeling of cortical perineuronal nets in focal cerebral ischemia and mild hypoperfusion. Matrix Biol 2018; 74:121-132. [PMID: 30092283 DOI: 10.1016/j.matbio.2018.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/02/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022]
Abstract
Despite the crucial role of perineuronal nets (PNNs) in neural plasticity and neurological disorders, their ultrastructural organization remains largely unresolved. We have developed a novel approach combining superresolution structured illumination microscopy (SR-SIM) and mathematical reconstruction that allows for quantitative analysis of PNN topology. Since perineuronal matrix is capable to restrict neural plasticity but at the same time is necessary to maintain synapses, we hypothesized that a beneficial post stroke recovery requires a reversible loosening of PNNs. Our results indicated that focal cerebral ischemia induces partial depletion of PNNs and that mild hypoperfusion not associated with ischemic injury can induce ultra-structural rearrangements in visually intact meshworks. In line with the activation of neural plasticity under mild stress stimuli, we provide evidence that topological conversion of PNNs can support post stroke neural rewiring.
Collapse
|
1049
|
Liu CB, Yang DG, Zhang X, Zhang WH, Li DP, Zhang C, Qin C, Du LJ, Li J, Gao F, Zhang J, Zuo ZT, Yang ML, Li JJ. Degeneration of white matter and gray matter revealed by diffusion tensor imaging and pathological mechanism after spinal cord injury in canine. CNS Neurosci Ther 2018; 25:261-272. [PMID: 30076687 DOI: 10.1111/cns.13044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/07/2018] [Accepted: 07/11/2018] [Indexed: 12/13/2022] Open
Abstract
AIM Exploration of the mechanism of spinal cord degeneration may be the key to treatment of spinal cord injury (SCI). This study aimed to investigate the degeneration of white matter and gray matter and pathological mechanism in canine after SCI. METHODS Diffusion tensor imaging (DTI) was performed on canine models with normal (n = 5) and injured (n = 7) spinal cords using a 3.0T MRI scanner at precontusion and 3 hours, 24 hours, 6 weeks, and 12 weeks postcontusion. The tissue sections were stained using H&E and immunohistochemistry. RESULTS For white matter, fractional anisotropy (FA) values significantly decreased in lesion epicenter, caudal segment 1 cm away from epicenter, and caudal segment 2 cm away from epicenter (P = 0.003, P = 0.004, and P = 0.013, respectively) after SCI. Apparent diffusion coefficient (ADC) values were initially decreased and then increased in lesion epicenter and caudal segment 1 cm away from epicenter (P < 0.001 and P = 0.010, respectively). There are no significant changes in FA and ADC values in rostral segments (P > 0.05). For gray matter, ADC values decreased initially and then increased in lesion epicenter (P < 0.001), and overall trend decreased in caudal segment 1 cm away from epicenter (P = 0.039). FA values did not change significantly (P > 0.05). Pathological examination confirmed the dynamic changes of DTI parameters. CONCLUSION Diffusion tensor imaging is more sensitive to degeneration of white matter than gray matter, and the white matter degeneration may be not symmetrical which meant the caudal degradation appeared to be more severe than the rostral one.
Collapse
Affiliation(s)
- Chang-Bin Liu
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - De-Gang Yang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Xin Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Wen-Hao Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Da-Peng Li
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Chao Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Chuan Qin
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Liang-Jie Du
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Jun Li
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Feng Gao
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Jie Zhang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Zhen-Tao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,The Innovation Center of Excellence on Brain Science, Chinese Academy of Sciences, Beijing, China.,Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Ming-Liang Yang
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Jian-Jun Li
- School of Rehabilitation Medicine, Capital Medical University, Beijing, China.,Department of Spinal and Neural Function Reconstruction, China Rehabilitation Research Center, Beijing, China.,Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.,China Rehabilitation Science Institute, Beijing, China.,Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| |
Collapse
|
1050
|
NG2/CSPG4 and progranulin in the posttraumatic glial scar. Matrix Biol 2018; 68-69:571-588. [DOI: 10.1016/j.matbio.2017.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/17/2022]
|