851
|
Rudlong J, Cheng A, Johnson GVW. The role of transglutaminase 2 in mediating glial cell function and pathophysiology in the central nervous system. Anal Biochem 2019; 591:113556. [PMID: 31866289 DOI: 10.1016/j.ab.2019.113556] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/04/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022]
Abstract
The ubiquitously expressed transglutaminase 2 (TG2) has diverse functions in virtually all cell types, with its role depending not only on cell type, but also on specific subcellular localization. In the central nervous system (CNS) different types of glial cells, such as astrocytes, microglia, and oligodendrocytes and their precursor cells (OPCs), play pivotal supportive functions. This review is focused on what is currently known about the role of TG2 in each type of glial cell, in the context of normal function and pathophysiology. For example, astrocytic TG2 facilitates their migration and proliferation, but hinders their ability to protect neurons after CNS injury. The review also examines the interactions between glial cell types, and how TG2 in one cell type may affect another, as well as implications for specific TG2 populations as therapeutic targets in CNS pathology.
Collapse
Affiliation(s)
- Jacob Rudlong
- Department of Anesthesiology and Perioperative Medicine and the Neuroscience Graduate Program, University of Rochester, Rochester, NY, 14620, USA
| | - Anson Cheng
- Department of Anesthesiology and Perioperative Medicine and the Neuroscience Graduate Program, University of Rochester, Rochester, NY, 14620, USA
| | - Gail V W Johnson
- Department of Anesthesiology and Perioperative Medicine and the Neuroscience Graduate Program, University of Rochester, Rochester, NY, 14620, USA.
| |
Collapse
|
852
|
Zarei-Kheirabadi M, Hesaraki M, Kiani S, Baharvand H. In vivo conversion of rat astrocytes into neuronal cells through neural stem cells in injured spinal cord with a single zinc-finger transcription factor. Stem Cell Res Ther 2019; 10:380. [PMID: 31842989 PMCID: PMC6916443 DOI: 10.1186/s13287-019-1448-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/23/2019] [Accepted: 10/09/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) results in glial scar formation and irreversible neuronal loss, which finally leads to functional impairments and long-term disability. Our previous studies have demonstrated that the ectopic expression of Zfp521 reprograms fibroblasts and astrocytes into induced neural stem cells (iNSCs). However, it remains unclear whether treatment with Zfp521 also affects endogenous astrocytes, thus promoting further functional recovery following SCI. METHODS Rat astrocytes were transdifferentiated into neural stem cells in vitro by ZFP521 or Sox2. Then, ZFP521 was applied to the spinal cord injury site of a rat. Transduction, real-time PCR, immunohistofluorescence, and function assessments were performed at 6 weeks post-transduction to evaluate improvement and in vivo lineage reprogramming of astrocytes. RESULTS Here, we show that Zfp521 is more efficient in reprogramming cultured astrocytes compared with Sox2. In the injured spinal cord of an adult rat, resident astrocytes can be reprogrammed into neurons through a progenitor stage by Zfp521. Importantly, this treatment improves the functional abilities of the rats as evaluated by the Basso, Beattie, and Bresnahan (BBB) locomotor rating scale and further by calculation of its subscores. There was enhanced locomotor activity in the hind limbs, step length, toe spread, foot length, and paw area. In addition, motor evoked potential recordings demonstrated the functional integrity of the spinal cord. CONCLUSIONS These results have indicated that the generation of iNSCs or neurons from endogenous astrocytes by in situ reprogramming might be a potential strategy for SCI repair.
Collapse
Affiliation(s)
- Masoumeh Zarei-Kheirabadi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Sahar Kiani
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, 1461968151, Iran.
| |
Collapse
|
853
|
Toro CA, Das DK, Cai D, Cardozo CP. Elucidating the Role of Apolipoprotein E Isoforms in Spinal Cord Injury-Associated Neuropathology. J Neurotrauma 2019; 36:3317-3322. [PMID: 31218915 DOI: 10.1089/neu.2018.6334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating, life-altering, neurological event that affects ∼300,000 individuals in the United States. Currently, there are no effective treatments to reverse the neurological impairments caused by the lesion. Until a cure is available, there is an urgent need for strategies that can either spare injured neurons or promote neuroplasticity and functional recovery. Genetic links to outcomes after SCI may provide insights into the pathological mechanisms, and possible new avenues for drug development. In the present review, we discuss the current knowledge linking apolipoprotein E genotypes with better or worse functional outcomes after an SCI, and the possible molecular mechanisms that may contribute to this association.
Collapse
Affiliation(s)
- Carlos A Toro
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
| | - Dibash K Das
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
| | - Dongming Cai
- Neurology Service, James J. Peters VA, Bronx, New York
- Department of Neurology, Icahn School of Medicine at Mount Sinai, Bronx, New York
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA, Bronx, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
- Department of Rehabilitative Medicine, Icahn School of Medicine at Mount Sinai, Bronx, New York
| |
Collapse
|
854
|
Wu R, Mao S, Wang Y, Zhou S, Liu Y, Liu M, Gu X, Yu B. Differential Circular RNA Expression Profiles Following Spinal Cord Injury in Rats: A Temporal and Experimental Analysis. Front Neurosci 2019; 13:1303. [PMID: 31920480 PMCID: PMC6916439 DOI: 10.3389/fnins.2019.01303] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI), one of the most severe types of neurological damage, results in persistent motor and sensory dysfunction and involves complex gene alterations. Circular RNAs (circRNAs) are a recently discovered class of regulatory molecules, and their roles in SCI still need to be addressed. This study comprehensively investigated circRNA alterations in rats across a set time course (days 0, 1, 3, 7, 14, 21, and 28) after hemisection SCI at the right T9 site. A total of 360 differentially expressed circRNAs were identified using RNA sequencing. From these, the functions of the exonic circRNA_01477 were further explored in cultured spinal cord astrocytes. Knockdown of circRNA_01477 significantly inhibited astrocyte proliferation and migration. The circRNA_01477/microRNAs (miRNA)/messenger RNA (mRNA) interaction network was visualized following microarray assay. Among the downregulated differentially expressed mRNAs, four of the seven validated genes were controlled by miRNA-423-5p. We then demonstrated that miRNA-423-5p is significantly upregulated after circRNA_01477 depletion. In summary, this study provides, for the first time, a systematic evaluation of circRNA alterations following SCI and an insight into the transcriptional regulation of the genes involved. It further reveals that circRNA_01477/miR-423-5p could be a key regulator involved in regulating the changeable regeneration environment that occurs during recovery from SCI.
Collapse
Affiliation(s)
- Ronghua Wu
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yaxian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shuoshuo Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaosong Gu
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
855
|
Frost GR, Jonas LA, Li YM. Friend, Foe or Both? Immune Activity in Alzheimer's Disease. Front Aging Neurosci 2019; 11:337. [PMID: 31920620 PMCID: PMC6916654 DOI: 10.3389/fnagi.2019.00337] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/21/2019] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is marked by the presence of amyloid beta (Aβ) plaques, neurofibrillary tangles (NFT), neuronal death and synaptic loss, and inflammation in the brain. AD research has, in large part, been dedicated to the understanding of Aβ and NFT deposition as well as to the pharmacological reduction of these hallmarks. However, recent GWAS data indicates neuroinflammation plays a critical role in AD development, thereby redirecting research efforts toward unveiling the complexities of AD-associated neuroinflammation. It is clear that the innate immune system is intimately associated with AD progression, however, the specific roles of glia and neuroinflammation in AD pathology remain to be described. Moreover, inflammatory processes have largely been painted as detrimental to AD pathology, when in fact, many immune mechanisms such as phagocytosis aid in the reduction of AD pathologies. In this review, we aim to outline the delicate balance between the beneficial and detrimental aspects of immune activation in AD as a more thorough understanding of these processes is critical to development of effective therapeutics for AD.
Collapse
Affiliation(s)
- Georgia R. Frost
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, Manhattan, NY, United States
| | - Lauren A. Jonas
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, Manhattan, NY, United States
- Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, Ithaca, NY, United States
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, Manhattan, NY, United States
- Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
856
|
Potassium and glutamate transport is impaired in scar-forming tumor-associated astrocytes. Neurochem Int 2019; 133:104628. [PMID: 31825815 PMCID: PMC6957761 DOI: 10.1016/j.neuint.2019.104628] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/09/2023]
Abstract
Unprovoked recurrent seizures are a serious comorbidity affecting most patients who suffer from glioma, a primary brain tumor composed of malignant glial cells. Cellular mechanisms contributing to the development of recurrent spontaneous seizures include the release of the excitatory neurotransmitter glutamate from glioma into extracellular space. Under physiological conditions, astrocytes express two high affinity glutamate transporters, Glt-1 and Glast, which are responsible for the removal of excess extracellular glutamate. In the context of neurological disease or brain injury, astrocytes become reactive which can negatively affect neuronal function, causing hyperexcitability and/or death. Using electrophysiology, immunohistochemistry, fluorescent in situ hybridization, and Western blot analysis in different orthotopic xenograft and allograft models of human and mouse gliomas, we find that peritumoral astrocytes exhibit astrocyte scar formation characterized by proliferation, cellular hypertrophy, process elongation, and increased GFAP and pSTAT3. Overall, peritumoral reactive astrocytes show a significant reduction in glutamate and potassium uptake, as well as decreased glutamine synthetase activity. A subset of peritumoral astrocytes displayed a depolarized resting membrane potential, further contributing to reduced potassium and glutamate homeostasis. These changes may contribute to the propagation of peritumoral neuronal hyperexcitability and excitotoxic death.
Collapse
|
857
|
Smith DR, Dumont CM, Ciciriello AJ, Guo A, Tatineni R, Munsell MK, Cummings BJ, Anderson AJ, Shea LD. PLG Bridge Implantation in Chronic SCI Promotes Axonal Elongation and Myelination. ACS Biomater Sci Eng 2019; 5:6679-6690. [PMID: 33423486 PMCID: PMC11283858 DOI: 10.1021/acsbiomaterials.9b01012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition that may cause permanent functional loss below the level of injury, including paralysis and loss of bladder, bowel, and sexual function. Patients are rarely treated immediately, and this delay is associated with tissue loss and scar formation that can make regeneration at chronic time points more challenging. Herein, we investigated regeneration using a poly(lactide-co-glycolide) multichannel bridge implanted into a chronic SCI following surgical resection of necrotic tissue. We characterized the dynamic injury response and noted that scar formation decreased at 4 and 8 weeks postinjury (wpi), yet macrophage infiltration increased between 4 and 8 wpi. Subsequently, the scar tissue was resected and bridges were implanted at 4 and 8 wpi. We observed robust axon growth into the bridge and remyelination at 6 months after initial injury. Axon densities were increased for 8 week bridge implantation relative to 4 week bridge implantation, whereas greater myelination, particularly by Schwann cells, was observed with 4 week bridge implantation. The process of bridge implantation did not significantly decrease the postinjury function. Collectively, this chronic model follows the pathophysiology of human SCI, and bridge implantation allows for clear demarcation of the regenerated tissue. These data demonstrate that bridge implantation into chronic SCI supports regeneration and provides a platform to investigate strategies to buttress and expand regeneration of neural tissue at chronic time points.
Collapse
Affiliation(s)
- Dominique R. Smith
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Courtney M. Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL, 33156
- Biomedical Nanotechnology Institute at University of Miami (BioNIUM), University of Miami, 1951 NW 7 Avenue Ste 475, Miami, FL, 33136
| | - Andrew J. Ciciriello
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL, 33156
- Biomedical Nanotechnology Institute at University of Miami (BioNIUM), University of Miami, 1951 NW 7 Avenue Ste 475, Miami, FL, 33136
| | - Amina Guo
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Ravindra Tatineni
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Mary K. Munsell
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Brian J. Cummings
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, 1100 Gottschalk Medical Plaza, Irvine, CA, 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, 845 Health Science Road, Irvine, CA, 92697
- Department of Anatomy and Neurobiology, University of California, 364 Med Surge II, Irvine, CA, 92697
- Department of Physical Medicine and Rehabilitation, University of California, 101 The City Drive South, Building 53, Suite 311A, Orange, CA, 92868
| | - Aileen J. Anderson
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, 1100 Gottschalk Medical Plaza, Irvine, CA, 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, 845 Health Science Road, Irvine, CA, 92697
- Department of Anatomy and Neurobiology, University of California, 364 Med Surge II, Irvine, CA, 92697
- Department of Physical Medicine and Rehabilitation, University of California, 101 The City Drive South, Building 53, Suite 311A, Orange, CA, 92868
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109
| |
Collapse
|
858
|
Metabolic Control of Astrocyte Pathogenic Activity via cPLA2-MAVS. Cell 2019; 179:1483-1498.e22. [PMID: 31813625 DOI: 10.1016/j.cell.2019.11.016] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/31/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022]
Abstract
Metabolism has been shown to control peripheral immunity, but little is known about its role in central nervous system (CNS) inflammation. Through a combination of proteomic, metabolomic, transcriptomic, and perturbation studies, we found that sphingolipid metabolism in astrocytes triggers the interaction of the C2 domain in cytosolic phospholipase A2 (cPLA2) with the CARD domain in mitochondrial antiviral signaling protein (MAVS), boosting NF-κB-driven transcriptional programs that promote CNS inflammation in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis. cPLA2 recruitment to MAVS also disrupts MAVS-hexokinase 2 (HK2) interactions, decreasing HK enzymatic activity and the production of lactate involved in the metabolic support of neurons. Miglustat, a drug used to treat Gaucher and Niemann-Pick disease, suppresses astrocyte pathogenic activities and ameliorates EAE. Collectively, these findings define a novel immunometabolic mechanism that drives pro-inflammatory astrocyte activities, outlines a new role for MAVS in CNS inflammation, and identifies candidate targets for therapeutic intervention.
Collapse
|
859
|
Ma C, Zhang P, Shen Y. Progress in research into spinal cord injury repair: Tissue engineering scaffolds and cell transdifferentiation. JOURNAL OF NEURORESTORATOLOGY 2019; 7:196-206. [DOI: doi 10.26599/jnr.2019.9040024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025] Open
|
860
|
He Y, Liu X, Chen Z. Glial Scar-a Promising Target for Improving Outcomes After CNS Injury. J Mol Neurosci 2019; 70:340-352. [PMID: 31776856 DOI: 10.1007/s12031-019-01417-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Abstract
After central nervous system (CNS) injury, a series of stress responses induce astrocytes activation. Reactive astrocytes, which are typically different from astrocytes in normal conditions in altered morphology and gene expression, combine with extracellular matrix (ECM) components to form a glial scar at the lesion site, which walls of the injured region from neighboring healthier tissue. However, as a physical and molecular barrier, glial scar can impede patients' functional recovery in the late period of CNS injury. Thus, inhibiting glial scar formation in the chronic stage after CNS injury may be a promising target to improve outcomes. Since the therapeutic strategies targeting on mediating glial scar formation are regarded as an important part on improving functional recovery after CNS injury, in this review, we focus on the regulating effects of related signaling pathways and other molecules on glial scar, and the process of glial scar formation and the roles that it plays during the acute and chronic stages are also expounded in this article. We hope to get a comprehensive understanding of glial scar during CNS injury based on current researches and to open new perspectives for the therapies to promote functional recovery after CNS injury.
Collapse
Affiliation(s)
- Yuanyuan He
- Department of Pharmacy, Xuyi People's Hospital, 28 Hongwu Road, Xuyi, 211700, Jiangsu, People's Republic of China
| | - Xiaoyan Liu
- Department of Pharmacy, Xuyi People's Hospital, 28 Hongwu Road, Xuyi, 211700, Jiangsu, People's Republic of China
| | - Zhongying Chen
- Department of Pharmacy, Xuyi People's Hospital, 28 Hongwu Road, Xuyi, 211700, Jiangsu, People's Republic of China.
| |
Collapse
|
861
|
Connexins-Based Hemichannels/Channels and Their Relationship with Inflammation, Seizures and Epilepsy. Int J Mol Sci 2019; 20:ijms20235976. [PMID: 31783599 PMCID: PMC6929063 DOI: 10.3390/ijms20235976] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
Connexins (Cxs) are a family of 21 protein isoforms, eleven of which are expressed in the central nervous system, and they are found in neurons and glia. Cxs form hemichannels (connexons) and channels (gap junctions/electric synapses) that permit functional and metabolic coupling between neurons and astrocytes. Altered Cx expression and function is involved in inflammation and neurological diseases. Cxs-based hemichannels and channels have a relevance to seizures and epilepsy in two ways: First, this pathological condition increases the opening probability of hemichannels in glial cells to enable gliotransmitter release, sustaining the inflammatory process and exacerbating seizure generation and epileptogenesis, and second, the opening of channels favors excitability and synchronization through coupled neurons. These biological events highlight the global pathological mechanism of epilepsy, and the therapeutic potential of Cxs-based hemichannels and channels. Therefore, this review describes the role of Cxs in neuroinflammation and epilepsy and examines how the blocking of channels and hemichannels may be therapeutic targets of anti-convulsive and anti-epileptic treatments.
Collapse
|
862
|
Yu B, Yao C, Wang Y, Mao S, Wang Y, Wu R, Feng W, Chen Y, Yang J, Xue C, Liu D, Ding F, Gu X. The Landscape of Gene Expression and Molecular Regulation Following Spinal Cord Hemisection in Rats. Front Mol Neurosci 2019; 12:287. [PMID: 31824262 PMCID: PMC6883948 DOI: 10.3389/fnmol.2019.00287] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/12/2019] [Indexed: 01/25/2023] Open
Abstract
Spinal cord injury (SCI) is a challenging clinical problem worldwide. The cellular state and molecular expression in spinal cord tissue after injury are extremely complex and closely related to functional recovery. However, the spatial and temporal changes of gene expression and regulation in various cell types after SCI are still unclear. Here, we collected the rostral and caudal regions to the lesion at 11 time points over a period of 28 days after rat hemisection SCI. Combining whole-transcriptome sequencing and bioinformatic analysis, we identified differentially expressed genes (DEGs) between spinal cord tissue from injured and sham-operated animals. Significantly altered biological processes were enriched from DEGs in astrocytes, microglia, oligodendrocytes, immune cells, and vascular systems after SCI. We then identified dynamic trends in these processes using the average expression profiles of DEGs. Gene expression and regulatory networks for selected biological processes were also constructed to illustrate the complicate difference between rostral and caudal tissues. Finally, we validated the expressions of some key genes from these networks, including α-synuclein, heme oxygenase 1, bone morphogenetic protein 2, activating transcription factor 3, and leukemia inhibitory factor. Collectively, we provided a comprehensive network of gene expression and regulation to shed light on the molecular characteristics of critical biological processes that occur after SCI, which will broaden the understanding of SCI and facilitate clinical therapeutics for SCI.
Collapse
Affiliation(s)
- Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yaxian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ronghua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Wei Feng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yanping Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jian Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chengbin Xue
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Dong Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
863
|
Molina-Gonzalez I, Miron VE. Astrocytes in myelination and remyelination. Neurosci Lett 2019; 713:134532. [PMID: 31589903 DOI: 10.1016/j.neulet.2019.134532] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/13/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
Astrocytes are known to play critical roles in central nervous system development, homeostasis, and response to injury. In addition to well-defined functions in synaptic signalling and blood-brain barrier control, astrocytes are now emerging as important contributors to white matter health. Here, we review the roles of astrocytes in myelin formation and regeneration (remyelination), focusing on both direct interactions with oligodendrocyte lineage cells, and indirect influences via crosstalk with central nervous system resident macrophages, microglia.
Collapse
Affiliation(s)
- Irene Molina-Gonzalez
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
864
|
Hutson TH, Di Giovanni S. The translational landscape in spinal cord injury: focus on neuroplasticity and regeneration. Nat Rev Neurol 2019; 15:732-745. [DOI: 10.1038/s41582-019-0280-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2019] [Indexed: 12/22/2022]
|
865
|
Xu S, Sun Q, Fan J, Jiang Y, Yang W, Cui Y, Yu Z, Jiang H, Li B. Role of Astrocytes in Post-traumatic Epilepsy. Front Neurol 2019; 10:1149. [PMID: 31798512 PMCID: PMC6863807 DOI: 10.3389/fneur.2019.01149] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022] Open
Abstract
Traumatic brain injury, a common cause of acquired epilepsy, is typical to find necrotic cell death within the injury core. The dynamic changes in astrocytes surrounding the injury core contribute to epileptic seizures associated with intense neuronal firing. However, little is known about the molecular mechanisms that activate astrocytes during traumatic brain injury or the effect of functional changes of astrocytes on seizures. In this comprehensive review, we present our cumulated understanding of the complex neurological affection in astrocytes after traumatic brain injury. We approached the problem through describing the changes of cell morphology, neurotransmitters, biochemistry, and cytokines in astrocytes during post-traumatic epilepsy. In addition, we also discussed the relationship between dynamic changes in astrocytes and seizures and the current pharmacologic agents used for treatment. Hopefully, this review will provide a more detailed knowledge from which better therapeutic strategies can be developed to treat post-traumatic epilepsy.
Collapse
Affiliation(s)
- Songbai Xu
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
| | - Qihan Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jie Fan
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Yuanyuan Jiang
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Yifeng Cui
- Department of Pediatrics, Yanbian Maternal and Child Health Hospital, Yanji, China
| | - Zhenxiang Yu
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
| | - Huiyi Jiang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
866
|
Nikolic L, Nobili P, Shen W, Audinat E. Role of astrocyte purinergic signaling in epilepsy. Glia 2019; 68:1677-1691. [DOI: 10.1002/glia.23747] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/08/2019] [Accepted: 10/25/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Ljiljana Nikolic
- Institute for Biological Research Siniša Stanković, University of Belgrade Serbia
| | | | - Weida Shen
- Zhejiang University City College Zhejiang Hangzhou China
| | - Etienne Audinat
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM Montpellier France
| |
Collapse
|
867
|
Hachem LD, Mothe AJ, Tator CH. Unlocking the paradoxical endogenous stem cell response after spinal cord injury. Stem Cells 2019; 38:187-194. [PMID: 31648407 DOI: 10.1002/stem.3107] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/21/2019] [Accepted: 10/08/2019] [Indexed: 11/08/2022]
Abstract
Nearly a century ago, the concept of the secondary injury in spinal cord trauma was first proposed to explain the complex cascade of molecular and cellular events leading to widespread neuronal and glial cell death after trauma. In recent years, it has been established that the ependymal region of the adult mammalian spinal cord contains a population of multipotent neural stem/progenitor cells (NSPCs) that are activated after spinal cord injury (SCI) and likely play a key role in endogenous repair and regeneration. How these cells respond to the various components of the secondary injury remains poorly understood. Emerging evidence suggests that many of the biochemical components of the secondary injury cascade which have classically been viewed as deleterious to host neuronal and glial cells may paradoxically trigger NSPC activation, proliferation, and differentiation thus challenging our current understanding of secondary injury mechanisms in SCI. Herein, we highlight new findings describing the response of endogenous NSPCs to spinal cord trauma, redefining the secondary mechanisms of SCI through the lens of the endogenous population of stem/progenitor cells. Moreover, we outline how these insights can fuel novel stem cell-based therapeutic strategies to repair the injured spinal cord.
Collapse
Affiliation(s)
- Laureen D Hachem
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Andrea J Mothe
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Charles H Tator
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
868
|
Ago T. [Why are pericytes important for brain functions?]. Rinsho Shinkeigaku 2019; 59:707-715. [PMID: 31656270 DOI: 10.5692/clinicalneurol.cn-001357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pericytes are mural cells embedded in the basal membrane surrounding endothelial cells in capillary and small vessels (from precapillary arterioles to postcapillary venules). They exist with a high coverage ratio to endothelial cells in the brain and play crucial roles in the formation and maintenance of the blood-brain barrier and the control of blood flow through a close interaction with endothelial cells. Thus, intactness of pericyte is absolutely needed for neuronal/brain functions. Ageing, life-style diseases, hypoperfusion/ischemia, drugs, and genetic factors can primarily cause pericyte dysfunctions, thereby leading to the development or progression of various brain disorders, including cerebrovascular diseases. Because pericytes also play an important role in tissue repair after brain injuries, they have received much attention as a therapeutic target even from the standpoint of functional recovery.
Collapse
Affiliation(s)
- Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University
| |
Collapse
|
869
|
Mizuno GO, Wang Y, Shi G, Wang Y, Sun J, Papadopoulos S, Broussard GJ, Unger EK, Deng W, Weick J, Bhattacharyya A, Chen CY, Yu G, Looger LL, Tian L. Aberrant Calcium Signaling in Astrocytes Inhibits Neuronal Excitability in a Human Down Syndrome Stem Cell Model. Cell Rep 2019; 24:355-365. [PMID: 29996097 PMCID: PMC6631348 DOI: 10.1016/j.celrep.2018.06.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/04/2018] [Accepted: 06/07/2018] [Indexed: 11/25/2022] Open
Abstract
Down syndrome (DS) is a genetic disorder that causes cognitive impairment. The staggering effects associated with an extra copy of human chromosome 21 (HSA21) complicates mechanistic understanding of DS pathophysiology. We examined the neuron-astrocyte interplay in a fully recapitulated HSA21 trisomy cellular model differentiated from DS-patient-derived induced pluripotent stem cells (iPSCs). By combining calcium imaging with genetic approaches, we discovered the functional defects of DS astroglia and their effects on neuronal excitability. Compared with control isogenic astroglia, DS astroglia exhibited more-frequent spontaneous calcium fluctuations, which reduced the excitability of co-cultured neurons. Furthermore, suppressed neuronal activity could be rescued by abolishing astrocytic spontaneous calcium activity either chemically by blocking adenosine-mediated signaling or genetically by knockdown of inositol triphosphate (IP3) receptors or S100B, a calcium binding protein coded on HSA21. Our results suggest a mechanism by which DS alters the function of astrocytes, which subsequently disturbs neuronal excitability. To understand how Down syndrome (DS) affects neural networks, Mizuno et al. used a DS-patient-derived stem cell model and calcium imaging to investigate the functional defects of DS astrocytes and their effects on neuronal excitability. Their study reveals that DS astroglia exhibited more frequent spontaneous calcium fluctuations, which impair neuronal excitability.
Collapse
Affiliation(s)
- Grace O Mizuno
- Department of Biochemistry and Molecular Medicine, Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
| | - Yinxue Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Guilai Shi
- Department of Biochemistry and Molecular Medicine, Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
| | - Yizhi Wang
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Junqing Sun
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Stelios Papadopoulos
- Department of Biochemistry and Molecular Medicine, Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
| | - Gerard J Broussard
- Department of Biochemistry and Molecular Medicine, Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
| | - Elizabeth K Unger
- Department of Biochemistry and Molecular Medicine, Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, Shriner's Hospital, University of California, Davis, Davis, CA, USA
| | - Jason Weick
- Department of Neuroscience, University of New Mexico, Albuquerque, NM, USA
| | | | - Chao-Yin Chen
- Department of Pharmacology, University of California, Davis, Davis, CA, USA
| | - Guoqiang Yu
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
870
|
Pearson CS, Solano AG, Tilve SM, Mencio CP, Martin KR, Geller HM. Spatiotemporal distribution of chondroitin sulfate proteoglycans after optic nerve injury in rodents. Exp Eye Res 2019; 190:107859. [PMID: 31705897 DOI: 10.1016/j.exer.2019.107859] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/16/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023]
Abstract
The accumulation of chondroitin sulfate proteoglycans (CSPGs) in the glial scar following acute damage to the central nervous system (CNS) limits the regeneration of injured axons. Given the rich diversity of CSPG core proteins and patterns of GAG sulfation, identifying the composition of these CSPGs is essential for understanding their roles in injury and repair. Differential expression of core proteins and sulfation patterns have been characterized in the brain and spinal cord of mice and rats, but a comprehensive study of these changes following optic nerve injury has not yet been performed. Here, we show that the composition of CSPGs in the optic nerve and retina following optic nerve crush (ONC) in mice and rats exhibits an increase in aggrecan, brevican, phosphacan, neurocan and versican, similar to changes following spinal cord injury. We also observe an increase in inhibitory 4-sulfated (4S) GAG chains, which suggests that the persistence of CSPGs in the glial scar opposes the growth of CNS axons, thereby contributing to the failure of regeneration and recovery of function.
Collapse
Affiliation(s)
- Craig S Pearson
- Laboratory of Developmental Neurobiology, National Heart, Lung, Blood Institute, National Institutes of Health, Bethesda, MD, USA; Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Andrea G Solano
- Laboratory of Developmental Neurobiology, National Heart, Lung, Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sharada M Tilve
- Laboratory of Developmental Neurobiology, National Heart, Lung, Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Caitlin P Mencio
- Laboratory of Developmental Neurobiology, National Heart, Lung, Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Keith R Martin
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, National Heart, Lung, Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
871
|
Leucine Zipper-Bearing Kinase Is a Critical Regulator of Astrocyte Reactivity in the Adult Mammalian CNS. Cell Rep 2019; 22:3587-3597. [PMID: 29590625 PMCID: PMC5905706 DOI: 10.1016/j.celrep.2018.02.102] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/08/2017] [Accepted: 02/26/2018] [Indexed: 01/09/2023] Open
Abstract
Reactive astrocytes influence post-injury recovery, repair, and pathogenesis of the mammalian CNS. Much of the regulation of astrocyte reactivity, however, remains to be understood. Using genetic loss and gain-of-function analyses in vivo, we show that the conserved MAP3K13 (also known as leucine zipper-bearing kinase [LZK]) promotes astrocyte reactivity and glial scar formation after CNS injury. Inducible LZK gene deletion in astrocytes of adult mice reduced astrogliosis and impaired glial scar formation, resulting in increased lesion size after spinal cord injury. Conversely, LZK overexpression in astrocytes enhanced astrogliosis and reduced lesion size. Remarkably, in the absence of injury, LZK overexpression alone induced widespread astrogliosis in the CNS and upregulated astrogliosis activators pSTAT3 and SOX9. The identification of LZK as a critical cell-intrinsic regulator of astrocyte reactivity expands our understanding of the multicellular response to CNS injury and disease, with broad translational implications for neural repair.
Collapse
|
872
|
Sabin KZ, Echeverri K. The role of the immune system during regeneration of the central nervous system. ACTA ACUST UNITED AC 2019; 7. [PMID: 32864529 DOI: 10.1016/j.regen.2019.100023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Central nervous system damage in mammals leads to neuronal cell death, axonal degeneration, and formation of a glial scar resulting in functional and behavioral defects. Other vertebrates, like fish and salamanders, have retained the ability to functionally regenerate after central nervous system injury. To date research from many research organisms has led to a more concise understanding of the response of local neural cells to injury. However, it has become clear that non-neural cells of the immune system play an important role in determining the tissue response to injury. In this review we briefly consider the mammalian response to injury compared to organisms with the natural ability to regenerate. We then discuss similarities and differences in how cells of the innate and adaptive immune system respond and contribute to tissue repair in various species.
Collapse
Affiliation(s)
- K Z Sabin
- Eugene Bell Center for Regenerative Biology & Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543 USA
| | - K Echeverri
- Eugene Bell Center for Regenerative Biology & Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02543 USA
| |
Collapse
|
873
|
Wang Z, Huang J, Liu C, Liu L, Shen Y, Shen C, Liu C. BAF45D Downregulation in Spinal Cord Ependymal Cells Following Spinal Cord Injury in Adult Rats and Its Potential Role in the Development of Neuronal Lesions. Front Neurosci 2019; 13:1151. [PMID: 31736692 PMCID: PMC6828649 DOI: 10.3389/fnins.2019.01151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
The endogenous spinal cord ependymal cells (SCECs), which form the central canal (CC), are critically involved in proliferation, differentiation and migration after spinal cord injury (SCI) and represents a repair cell source in treating SCI. Previously, we reported that BAF45D is expressed in the SCECs and the spinal cord neurons in adult mice and knockdown of BAF45D fail to induce expression of PAX6, a neurogenic fate determinant, during early neural differentiation of human embryonic stem cells. However, the effects of SCI on expression of BAF45D have not been reported. The aim of this study is to explore the expression and potential role of BAF45D in rat SCI model. In this study, adult rats were randomly divided into intact, sham, and SCI groups. We first explored expression of BAF45D in the SCECs in intact adult rats. We then explored SCI-induced loss of motor neurons and lesion of neurites in the anterior horns induced by the SCI. We also investigated whether the SCI-induced lesions in SCECs are accompanied by the motor neuron lesions. Finally, we examined the effect of BAF45D knockdown on cell growth in neuro2a cells. Our data showed that BAF45D is expressed in SCECs, neurons, and oligodendrocytes but not astrocytes in the spinal cords of intact adult rats. After SCI, the structure of CC was disrupted and the BAF45D-positive SCEC-derivatives were decreased. During the early stages of SCI, when shape of CC was affected but there was no disruption in circular structure of the SCECs, it was evident that there was a significant reduction in the number of neurites and motor neurons in the anterior horns compared with those of intact rats. In comparison, a complete loss of SCECs accompanied by further loss of motor neurons but not neurites was observed at the later stage. BAF45D knockdown was also found to inhibit cell growth in neuro2a cells. These results highlight the decreased expression of BAF45D in SCI-injured SCECs and the potential role of BAF45D downregulation in development of neuronal lesion after SCI in adult rats.
Collapse
Affiliation(s)
- Zhenzhen Wang
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Histology and Embryology, Anhui Medical University, Hefei, China
- Institute of Stem Cell and Tissue Engineering, Anhui Medical University, Hefei, China
| | - Jian Huang
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Histology and Embryology, Anhui Medical University, Hefei, China
- Institute of Stem Cell and Tissue Engineering, Anhui Medical University, Hefei, China
| | - Chang Liu
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lihua Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Cailiang Shen
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chao Liu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Department of Histology and Embryology, Anhui Medical University, Hefei, China
- Institute of Stem Cell and Tissue Engineering, Anhui Medical University, Hefei, China
| |
Collapse
|
874
|
Dexmedetomidine Protects Against Oxygen-Glucose Deprivation-Induced Injury Through Inducing Astrocytes Autophagy via TSC2/mTOR Pathway. Neuromolecular Med 2019; 22:210-217. [PMID: 31654225 PMCID: PMC7230061 DOI: 10.1007/s12017-019-08576-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 10/11/2019] [Indexed: 12/13/2022]
Abstract
Although there is an increment in stroke burden in the world, stroke therapeutic strategies are still extremely limited to a minority of patients. We previously demonstrated that dexmedetomidine (DEX) protects against focal cerebral ischemia via inhibiting neurons autophagy. Nevertheless, the role of DEX in regulating astrocytes autophagic status in oxygen–glucose deprivation, a condition that mimics cerebral ischemia, is still unknown. In this study, we have shown that DEX and DEX + RAPA (autophagy inducer) increased viability and reduced apoptosis of primary astrocytes in oxygen–glucose deprivation (OGD) model compared with DEX + 3-methyladenine (3-MA) (autophagy inhibitor). DEX induced the expression of microtubule-associated protein 1 light chain 3 (LC3) and Beclin 1, while reduced the expression of p62 in primary cultured astrocytes through induction of autophagy. In addition, DEX enhanced the expression of tuberous sclerosis complex 2 (TSC2) in primary cultured astrocytes, while reduced the expression of mammalian target of rapamycin (mTOR). In conclusion, our study suggests that DEX exerts a neuroprotection against OGD-induced astrocytes injury via activation of astrocytes autophagy by regulating the TSC2/mTOR signaling pathway, which provides a new insight into the mechanisms of DEX treatment for acute ischemic injury.
Collapse
|
875
|
Ashammakhi N, Kim HJ, Ehsanipour A, Bierman RD, Kaarela O, Xue C, Khademhosseini A, Seidlits SK. Regenerative Therapies for Spinal Cord Injury. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:471-491. [PMID: 31452463 DOI: 10.1089/ten.teb.2019.0182] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Spinal cord injury (SCI) is a serious problem that primarily affects younger and middle-aged adults at its onset. To date, no effective regenerative treatment has been developed. Over the last decade, researchers have made significant advances in stem cell technology, biomaterials, nanotechnology, and immune engineering, which may be applied as regenerative therapies for the spinal cord. Although the results of clinical trials using specific cell-based therapies have proven safe, their efficacy has not yet been demonstrated. The pathophysiology of SCI is multifaceted, complex and yet to be fully understood. Thus, combinatorial therapies that simultaneously leverage multiple approaches will likely be required to achieve satisfactory outcomes. Although combinations of biomaterials with pharmacologic agents or cells have been explored, few studies have combined these modalities in a systematic way. For most strategies, clinical translation will be facilitated by the use of minimally invasive therapies, which are the focus of this review. In addition, this review discusses previously explored therapies designed to promote neuroregeneration and neuroprotection after SCI, while highlighting present challenges and future directions. Impact Statement To date there are no effective treatments that can regenerate the spinal cord after injury. Although there have been significant preclinical advances in bioengineering and regenerative medicine over the last decade, these have not translated into effective clinical therapies for spinal cord injury. This review focuses on minimally invasive therapies, providing extensive background as well as updates on recent technological developments and current clinical trials. This review is a comprehensive resource for researchers working towards regenerative therapies for spinal cord injury that will help guide future innovation.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland.,Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | - Han-Jun Kim
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California
| | | | | | - Outi Kaarela
- Division of Plastic Surgery, Department of Surgery, Oulu University, Oulu, Finland
| | - Chengbin Xue
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong University, Nantong, P.R. China
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Department of Radiological Sciences, University of California, Los Angeles, Los Angeles, California.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, California.,Center of Nanotechnology, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Chemical and Biological Engineering, University of California, Los Angeles, California
| | - Stephanie K Seidlits
- Center for Minimally Invasive Therapeutics (C-MIT), Los Angeles, California.,California NanoSystems Institute (CNSI), Los Angeles, California.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California.,Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, California.,Brain Research Institute, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
876
|
Divolis G, Stavropoulos A, Manioudaki M, Apostolidou A, Doulou A, Gavriil A, Dafnis I, Chroni A, Mummery C, Xilouri M, Sideras P. Activation of both transforming growth factor-β and bone morphogenetic protein signalling pathways upon traumatic brain injury restrains pro-inflammatory and boosts tissue reparatory responses of reactive astrocytes and microglia. Brain Commun 2019; 1:fcz028. [PMID: 32954268 PMCID: PMC7425383 DOI: 10.1093/braincomms/fcz028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Various ligands and receptors of the transforming growth factor-β superfamily have been found upregulated following traumatic brain injury; however, the role of this signalling system in brain injury pathophysiology is not fully characterized. To address this, we utilized an acute stab wound brain injury model to demonstrate that hallmarks of transforming growth factor-β superfamily system activation, such as levels of phosphorylated Smads, ligands and target genes for both transforming growth factor-β and bone morphogenetic protein pathways, were upregulated within injured tissues. Using a bone morphogenetic protein-responsive reporter mouse model, we showed that activation of the bone morphogenetic protein signalling pathway involves primarily astrocytes that demarcate the wound area. Insights regarding the potential role of transforming growth factor-β superfamily activation in glia cells within the injured tissues were obtained indirectly by treating purified reactive astrocytes and microglia with bone morphogenetic protein-4 or transforming growth factor-β1 and characterizing changes in their transcriptional profiles. Astrocytes responded to both ligands with considerably overlapping profiles, whereas, microglia responded selectively to transforming growth factor-β1. Novel pathways, crucial for repair of tissue-injury and blood-brain barrier, such as activation of cholesterol biosynthesis and transport, production of axonal guidance and extracellular matrix components were upregulated by transforming growth factor-β1 and/or bone morphogenetic protein-4 in astrocytes. Moreover, both ligands in astrocytes and transforming growth factor-β1 in microglia shifted the phenotype of reactive glia cells towards the anti-inflammatory and tissue reparatory 'A2'-like and 'M0/M2'-like phenotypes, respectively. Increased expression of selected key components of the in vitro modulated pathways and markers of 'A2'-like astrocytes was confirmed within the wound area, suggesting that these processes could also be modulated in situ by the integrated action of transforming growth factor-β and/or bone morphogenetic protein-mediated signalling. Collectively, our study provides a comprehensive comparative analysis of transforming growth factor-β superfamily signalling in reactive astrocytes and microglia and points towards a crucial role of both transforming growth factor-β and bone morphogenetic protein pathways in modulating the inflammatory and brain injury reparatory functions of activated glia cells.
Collapse
Affiliation(s)
- Georgios Divolis
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Athanasios Stavropoulos
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Maria Manioudaki
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Anastasia Apostolidou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Athanasia Doulou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Ariana Gavriil
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Ioannis Dafnis
- Institute of Biosciences and Applications, National Center for Scientific Research-Demokritos, 15341 Athens, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research-Demokritos, 15341 Athens, Greece
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Maria Xilouri
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Paschalis Sideras
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
877
|
Hypertension and Its Impact on Stroke Recovery: From a Vascular to a Parenchymal Overview. Neural Plast 2019; 2019:6843895. [PMID: 31737062 PMCID: PMC6815533 DOI: 10.1155/2019/6843895] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Hypertension is the first modifiable vascular risk factor accounting for 10.4 million deaths worldwide; it is strongly and independently associated with the risk of stroke and is related to worse prognosis. In addition, hypertension seems to be a key player in the implementation of vascular cognitive impairment. Long-term hypertension, complicated or not by the occurrence of ischemic stroke, is often reviewed on its vascular side, and parenchymal consequences are put aside. Here, we sought to review the impact of isolated hypertension or hypertension associated to stroke on brain atrophy, neuron connectivity and neurogenesis, and phenotype modification of microglia and astrocytes. Finally, we discuss the impact of antihypertensive therapies on cell responses to hypertension and functional recovery. This attractive topic remains a focus of continued investigation and stresses the relevance of including this vascular risk factor in preclinical investigations of stroke outcome.
Collapse
|
878
|
Lassmann H. Pathology of inflammatory diseases of the nervous system: Human disease versus animal models. Glia 2019; 68:830-844. [PMID: 31605512 PMCID: PMC7065008 DOI: 10.1002/glia.23726] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022]
Abstract
Numerous recent studies have been performed to elucidate the function of microglia, macrophages, and astrocytes in inflammatory diseases of the central nervous system. Regarding myeloid cells a core pattern of activation has been identified, starting with the activation of resident homeostatic microglia followed by recruitment of blood borne myeloid cells. An initial state of proinflammatory activation is at later stages followed by a shift toward an‐anti‐inflammatory and repair promoting phenotype. Although this core pattern is similar between experimental models and inflammatory conditions in the human brain, there are important differences. Even in the normal human brain a preactivated microglia phenotype is evident, and there are disease specific and lesion stage specific differences in the contribution between resident and recruited myeloid cells and their lesion state specific activation profiles. Reasons for these findings reside in species related differences and in differential exposure to different environmental cues. Most importantly, however, experimental rodent studies on brain inflammation are mainly focused on autoimmune encephalomyelitis, while there is a very broad spectrum of human inflammatory diseases of the central nervous system, triggered and propagated by a variety of different immune mechanisms.
Collapse
Affiliation(s)
- Hans Lassmann
- Institut fur Hirnforschung, Medical University of Vienna, Wien, Austria
| |
Collapse
|
879
|
Driving Neurogenesis in Neural Stem Cells with High Sensitivity Optogenetics. Neuromolecular Med 2019; 22:139-149. [PMID: 31595404 DOI: 10.1007/s12017-019-08573-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/21/2019] [Indexed: 01/15/2023]
Abstract
Optogenetic stimulation of neural stem cells (NSCs) enables their activity-dependent photo-modulation. This provides a spatio-temporal tool for studying activity-dependent neurogenesis and for regulating the differentiation of the transplanted NSCs. Currently, this is mainly driven by viral transfection of channelrhodopsin-2 (ChR2) gene, which requires high irradiance and complex in vivo/vitro stimulation systems. Additionally, despite the extensive application of optogenetics in neuroscience, the transcriptome-level changes induced by optogenetic stimulation of NSCs have not been elucidated yet. Here, we made transformed NSCs (SFO-NSCs) stably expressing one of the step-function opsin (SFO)-variants of chimeric channelrhodopsins, ChRFR(C167A), which is more sensitive to blue light than native ChR2, via a non-viral transfection system using piggyBac transposon. We set up a simple low-irradiance optical stimulation (OS)-incubation system that induced c-fos mRNA expression, which is activity-dependent, in differentiating SFO-NSCs. More neuron-like SFO-NCSs, which had more elongated axons, were differentiated with daily OS than control cells without OS. This was accompanied by positive/negative changes in the transcriptome involved in axonal remodeling, synaptic plasticity, and microenvironment modulation with the up-regulation of several genes involved in the Ca2+-related functions. Our approach could be applied for stem cell transplantation studies in tissue with two strengths: lower carcinogenicity and less irradiance needed for tissue penetration.
Collapse
|
880
|
Liu R, Wang W, Wang S, Xie W, Li H, Ning B. microRNA-21 regulates astrocytic reaction post-acute phase of spinal cord injury through modulating TGF-β signaling. Aging (Albany NY) 2019; 10:1474-1488. [PMID: 29936495 PMCID: PMC6046223 DOI: 10.18632/aging.101484] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/14/2018] [Indexed: 01/28/2023]
Abstract
Astrogliosis following spinal cord injury (SCI) was considered as a negative factor for neural regeneration. We found that miR-21 was significantly upregulated after SCI. So, we aim to determine whether miR-21 acts in a positive manner post SCI. In vitro, we measured the proliferation, apoptosis and cytokine secretion of primary cultured astrocytes after modulating the expression of miR-21 by western blot, RT-PCR and immunofluorescence. In vivo, we performed a modified Allen's weight drop model. Manipulation of the miR-21 expression level was achieved by interfering with antagomir and agomir. Clinic score was evaluated and recorded every day. Then, western blot, immunohistochemistry, TUNEL assay and ELISA were performed to detect pathological and functional alterations. Our results demonstrate that miR-21 can modulate the secretion, proliferation and apoptosis of astrocytes to promote recovery after SCI both in vivo and in vitro. These effects are likely mediated through transforming growth factor beta mediated targeting of the PI3K/Akt/mTOR pathway. These data suggest that miR-21 can regulate astrocytic function, then promote the functional recovery after SCI. We therefore highlight the positive effects of miR-21 after SCI.
Collapse
Affiliation(s)
- Ronghan Liu
- Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, China
| | - Wenzhao Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, China
| | - Shuya Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, China
| | - Wei Xie
- Affiliated Hospital of Taishan Medical University, Taian, Shandong 271000, China
| | - Hongfei Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, China
| | - Bin Ning
- Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, China
| |
Collapse
|
881
|
Liddelow SA. Modern approaches to investigating non-neuronal aspects of Alzheimer's disease. FASEB J 2019; 33:1528-1535. [PMID: 30703873 DOI: 10.1096/fj.201802592] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The slow, continuous, devastating march of Alzheimer's disease continues to move across the globe. As a society, we are at a loss for options to treat or reverse the death of neurons-the final, apparently inescapable, hallmark of the disease. A continued focus on these dying neurons has taught us much about the disease but with no knowledge-based effective treatment in sight. A surge of interest in non-neuronal cells, including glia, blood vasculature, and immune cells, has shed new light on how we may better diagnose and treat patients. This may be our best hope to treat the millions patients with cognitive decline and memory loss.-Liddelow, S. A. Modern approaches to investigating non-neuronal aspects of Alzheimer's disease.
Collapse
Affiliation(s)
- Shane A Liddelow
- Neuroscience Institute and Department of Neuroscience and Physiology, New York University Langone Medical Center, New York, New York, USA; and.,Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
882
|
Barros Ribeiro da Silva V, Porcionatto M, Toledo Ribas V. The Rise of Molecules Able To Regenerate the Central Nervous System. J Med Chem 2019; 63:490-511. [PMID: 31518122 DOI: 10.1021/acs.jmedchem.9b00863] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Injury to the adult central nervous system (CNS) usually leads to permanent deficits of cognitive, sensory, and/or motor functions. The failure of axonal regeneration in the damaged CNS limits functional recovery. The lack of information concerning the biological mechanism of axonal regeneration and its complexity has delayed the process of drug discovery for many years compared to other drug classes. Starting in the early 2000s, the ability of many molecules to stimulate axonal regrowth was evaluated through automated screening techniques; many hits and some new mechanisms involved in axonal regeneration were identified. In this Perspective, we discuss the rise of the CNS regenerative drugs, the main biological techniques used to test these drug candidates, some of the most important screens performed so far, and the main challenges following the identification of a drug that is able to induce axonal regeneration in vivo.
Collapse
Affiliation(s)
| | - Marimélia Porcionatto
- Universidade Federal de São Paulo , Escola Paulista de Medicina, Laboratório de Neurobiologia Molecular, Departmento de Bioquímica , Rua Pedro de Toledo, 669 - third floor, 04039-032 São Paulo , São Paolo , Brazil
| | - Vinicius Toledo Ribas
- Universidade Federal de Minas Gerais , Instituto de Ciências Biológicas, Departamento de Morfologia, Laboratório de Neurobiologia Av. Antônio Carlos, 6627, room O3-245 , - Campus Pampulha, 31270-901 , Belo Horizonte , Minas Gerais , Brazil
| |
Collapse
|
883
|
Lee SJ, Zhu W, Nowicki M, Lee G, Heo DN, Kim J, Zuo YY, Zhang LG. 3D printing nano conductive multi-walled carbon nanotube scaffolds for nerve regeneration. J Neural Eng 2019; 15:016018. [PMID: 29064377 DOI: 10.1088/1741-2552/aa95a5] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Nanomaterials, such as carbon nanotubes (CNTs), have been introduced to modify the surface properties of scaffolds, thus enhancing the interaction between the neural cells and biomaterials. In addition to superior electrical conductivity, CNTs can provide nanoscale structures similar to those present in the natural neural environment. The primary objective of this study is to investigate the proliferative capability and differential potential of neural stem cells (NSCs) seeded on a CNT incorporated scaffold. APPROACH Amine functionalized multi-walled carbon nanotubes (MWCNTs) were incorporated with a PEGDA polymer to provide enhanced electrical properties as well as nanofeatures on the surface of the scaffold. A stereolithography 3D printer was employed to fabricate a well-dispersed MWCNT-hydrogel composite neural scaffold with a tunable porous structure. 3D printing allows easy fabrication of complex 3D scaffolds with extremely intricate microarchitectures and controlled porosity. MAIN RESULTS Our results showed that MWCNT-incorporated scaffolds promoted neural stem cell proliferation and early neuronal differentiation when compared to those scaffolds without the MWCNTs. Furthermore, biphasic pulse stimulation with 500 µA current promoted neuronal maturity quantified through protein expression analysis by quantitative polymerase chain reaction. SIGNIFICANCE Results of this study demonstrated that an electroconductive MWCNT scaffold, coupled with electrical stimulation, may have a synergistic effect on promoting neurite outgrowth for therapeutic application in nerve regeneration.
Collapse
Affiliation(s)
- Se-Jun Lee
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
884
|
Wang C, Gong Z, Huang X, Wang J, Xia K, Ying L, Shu J, Yu C, Zhou X, Li F, Liang C, Chen Q. An injectable heparin-Laponite hydrogel bridge FGF4 for spinal cord injury by stabilizing microtubule and improving mitochondrial function. Am J Cancer Res 2019; 9:7016-7032. [PMID: 31660084 PMCID: PMC6815951 DOI: 10.7150/thno.37601] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
Rationale: Spinal cord injury (SCI) remains a critical clinical challenge. The controlled release of FGF4, a novel neuroprotective factor, from a versatile Laponite hydrogel to the injured site was a promising strategy to promote axon regeneration and motor functional recovery after SCI. Methods: Characterization of Laponite, Laponite/Heparin (Lap/Hep) and Laponite/Heparin loaded with FGF4 (Lap/Hep@FGF4) hydrogels were measured by rheometer. Multiple comprehensive evaluations were used to detect motor functional recovery and the axonal rehabilitation after Lap/Hep@FGF4 treatment in vivo (SCI rat model). Moreover, microtubule dynamic and energy transportation, which regulated axonal regeneration was evaluated by Lap/Hep@FGF4 gel in vitro (primary neuron). Results: FGF4 released from Lap/Hep gel locally achieves strong protection and regeneration after SCI. The Lap/Hep@FGF4 group revealed remarkable motor functional recovery and axonal regrowth after SCI through suppressing inflammatory reaction, increasing remyelination and reducing glial/fibrotic scars. Furthermore, the underlying mechanism of axonal rehabilitation were demonstrated via enhancing microtubule stability and regulating mitochondrial localization after Lap/Hep@FGF4 treatment. Conclusion: This promising sustained release system provides a synergistic effective approach to enhance recovery after SCI underlying a novel mechanism of axonal rehabilitation, and shows a translational prospect for the clinical treatment of SCI.
Collapse
|
885
|
Qian D, Li L, Rong Y, Liu W, Wang Q, Zhou Z, Gu C, Huang Y, Zhao X, Chen J, Fan J, Yin G. Blocking Notch signal pathway suppresses the activation of neurotoxic A1 astrocytes after spinal cord injury. Cell Cycle 2019; 18:3010-3029. [PMID: 31530090 DOI: 10.1080/15384101.2019.1667189] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a catastrophic disease which has complicated pathogenesis including inflammation, oxidative stress and glial scar formation. Astrocytes are the most abundant cells in central nervous system and fulfill homeostatic functions. Recent studies have described a new reactive phenotype of astrocytes, A1, induced by inflammation, which may have negative effects in SCI. As the Notch signaling pathway has been linked to cell differentiation and inflammation, we aimed to investigate its potential role in the differentiation of astrocytes in SCI. Contusive SCI rat model showed elevated A1 astrocyte numbers at the damage site 28 days after SCI and the expression levels of Notch signaling and its downstream genes were upregulated parallelly. Western blotting, RT-qPCR and immunofluorescence revealed that blocking of Notch pathway using γ-secretase blocker (DAPT) suppressed the differentiation of A1 astrocytes. Flow cytometry, and TUNEL staining indicated that DAPT alleviated neuronal apoptosis and axonal damage caused by A1 astrocytes likely through the Notch-dependent release of pro-inflammatory factors. CO-IP and western blotting revealed an interaction between Notch pathway and signal transducer and activator of transcription 3 (Stat3), which played a vital role in differentiation of A1 astrocytes. We conclude that phenotypic transition of A1 astrocytes and their neurotoxity were controlled by the Notch-Stat3 axis and that Notch pathway in astrocytes may serve as a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Dingfei Qian
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Linwei Li
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Yuluo Rong
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Wei Liu
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Qian Wang
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Zheng Zhou
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Changjiang Gu
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Yifan Huang
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Xuan Zhao
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Jian Chen
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Jin Fan
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| | - Guoyong Yin
- Department of Orthopedic, The First Affiliated Hospital of Nanjing Medical University , Nanjing , P.R. China
| |
Collapse
|
886
|
Predicting Reactive Astrogliosis Propagation by Bayesian Computational Modeling: the Repeater Stations Model. Mol Neurobiol 2019; 57:879-895. [PMID: 31522382 DOI: 10.1007/s12035-019-01749-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Reactive astrogliosis occurs upon focal brain injury and in neurodegenerative diseases. The mechanisms that propagate reactive astrogliosis to distal parts of the brain, in a rapid wave that activates astrocytes and other cell types along the way, are not completely understood. It is proposed that damage-associated molecular patterns (DAMP) released by necrotic cells from the injury core have a major role in the reactive astrogliosis initiation but whether they also participate in reactive astrogliosis propagation remains to be determined. We here developed a Bayesian computational model to define the most probable model for reactive astrogliosis propagation. Starting with experimental data from GFAP-immunostained reactive astrocytes, we defined five types of astrocytes based on morphometrical cues and registered the position of each reactive astrocyte cell type in the hemisphere ipsilateral to the injured site after 3 and 7 days post-ischemia. We developed equations for the changes in DAMP concentration (due to diffusion, binding to receptors or degradation), soluble mediators secretion, and for the evolution reactive astrogliosis. We tested four predefined models based on abovementioned previous hypothesis and modifications to it. Our results showed that DAMP diffusion alone has not justified the reactive astrogliosis propagation as previously assumed. Only two models succeeded in accurately reproducing the experimentally measured data and they highlighted the role of microglia and the glial secretion of soluble mediators to sustain the reactive signal and activating neighboring astrocytes. Thus, our in silico analysis proposes that glial cells behave as repeater stations of the injury signal in order to propagate reactive astrogliosis.
Collapse
|
887
|
Verkhratsky A, Rodrigues JJ, Pivoriunas A, Zorec R, Semyanov A. Astroglial atrophy in Alzheimer’s disease. Pflugers Arch 2019; 471:1247-1261. [DOI: 10.1007/s00424-019-02310-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
|
888
|
Maysinger D, Ji J. Nanostructured Modulators of Neuroglia. Curr Pharm Des 2019; 25:3905-3916. [PMID: 31512994 DOI: 10.2174/1381612825666190912163339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/08/2019] [Indexed: 01/08/2023]
Abstract
Biological and synthetic nanostructures can influence both glia and neurons in the central nervous system. Neurons represent only a small proportion (about 10%) of cells in the brain, whereas glial cells are the most abundant cell type. Non-targeted nanomedicines are mainly internalized by glia, in particular microglia, and to a lesser extent by astrocytes. Internalized nanomedicines by glia indirectly modify the functional status of neurons. The mechanisms of biochemical, morphological and functional changes of neural cells exposed to nanomedicines are still not well-understood. This minireview provides a cross-section of morphological and biochemical changes in glial cells and neurons exposed to different classes of hard and soft nanostructures.
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec H3AOG4, Canada
| | - Jeff Ji
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec H3AOG4, Canada
| |
Collapse
|
889
|
Ong W, Pinese C, Chew SY. Scaffold-mediated sequential drug/gene delivery to promote nerve regeneration and remyelination following traumatic nerve injuries. Adv Drug Deliv Rev 2019; 149-150:19-48. [PMID: 30910595 DOI: 10.1016/j.addr.2019.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/27/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
Abstract
Neural tissue regeneration following traumatic injuries is often subpar. As a result, the field of neural tissue engineering has evolved to find therapeutic interventions and has seen promising outcomes. However, robust nerve and myelin regeneration remain elusive. One possible reason may be the fact that tissue regeneration often follows a complex sequence of events in a temporally-controlled manner. Although several other fields of tissue engineering have begun to recognise the importance of delivering two or more biomolecules sequentially for more complete tissue regeneration, such serial delivery of biomolecules in neural tissue engineering remains limited. This review aims to highlight the need for sequential delivery to enhance nerve regeneration and remyelination after traumatic injuries in the central nervous system, using spinal cord injuries as an example. In addition, possible methods to attain temporally-controlled drug/gene delivery are also discussed for effective neural tissue regeneration.
Collapse
|
890
|
Bradbury EJ, Burnside ER. Moving beyond the glial scar for spinal cord repair. Nat Commun 2019; 10:3879. [PMID: 31462640 PMCID: PMC6713740 DOI: 10.1038/s41467-019-11707-7] [Citation(s) in RCA: 432] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 07/25/2019] [Indexed: 02/08/2023] Open
Abstract
Traumatic spinal cord injury results in severe and irreversible loss of function. The injury triggers a complex cascade of inflammatory and pathological processes, culminating in formation of a scar. While traditionally referred to as a glial scar, the spinal injury scar in fact comprises multiple cellular and extracellular components. This multidimensional nature should be considered when aiming to understand the role of scarring in limiting tissue repair and recovery. In this Review we discuss recent advances in understanding the composition and phenotypic characteristics of the spinal injury scar, the oversimplification of defining the scar in binary terms as good or bad, and the development of therapeutic approaches to target scar components to enable improved functional outcome after spinal cord injury.
Collapse
Affiliation(s)
- Elizabeth J Bradbury
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK.
| | - Emily R Burnside
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), Guy's Campus, London Bridge, London, SE1 1UL, UK
| |
Collapse
|
891
|
Delarue Q, Mayeur A, Chalfouh C, Honoré A, Duclos C, Di Giovanni M, Li X, Salaun M, Dampierre J, Vaudry D, Marie JP, Guérout N. Inhibition of ADAMTS-4 Expression in Olfactory Ensheathing Cells Enhances Recovery after Transplantation within Spinal Cord Injury. J Neurotrauma 2019; 37:507-516. [PMID: 31264504 DOI: 10.1089/neu.2019.6481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) induces permanent loss of sensitive and motor functions below the injury level. To date, a wide variety of cells has been used as biotherapies to cure SCI in different animal paradigms. Specifically, olfactory ensheathing cells (OECs) is one of the most promising. Indeed, OECs have been shown to enhance recovery in many animal studies. Moreover, OECs transplantation has been applied to a paraplegic patient and have shown beneficial effects. However, it has been reported that the significant level of recovery varies among different patients. Therefore, it is of primary importance to enhance the regenerative efficiency of OECs for better translations. Recently, it has been shown that inhibiting ADAMTS4 expression in glial cells in vitro increases their synthesis of neurotrophic factors. We hypothesized that the expression of neurotrophic factors secreted by OECs can be increased by the deletion of ADAMTS4. Taking advantage of ADAMTS4-/- mouse line, we produce ADAMTS4 deficient primary OEC cultures and then we investigated their regenerative potential after SCI. By using quantitative polymerase chain reaction, bioluminescence imaging, measurement of locomotor activity, electrophysiological studies, and immunohistochemistry, our results show that ADAMTS4-/- olfactory bulb OEC (bOECs) primary cultures upregulate their trophic factor expression in vitro, and that the transplantation of ADAMTS4-/- bOECs in a severe SCI model increases functional recovery and tissue repair in vivo. Altogether, our study reveals, for the first time, that primary bOEC cultures transplantation can be potentialized by inhibition of the expression of ADAMTS4.
Collapse
Affiliation(s)
- Quentin Delarue
- Normandie Univ, UNIROUEN, GRHV EA3830; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Anne Mayeur
- Normandie Univ, UNIROUEN, GRHV EA3830; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Chaima Chalfouh
- Normandie Univ, UNIROUEN, GRHV EA3830; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Axel Honoré
- Normandie Univ, UNIROUEN, GRHV EA3830; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Célia Duclos
- Normandie Univ, UNIROUEN, GRHV EA3830; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Marine Di Giovanni
- Normandie Univ, UNIROUEN, PRIMACEN; Institute for Research and Innovation in Biomedicine (IRIB), Mont-Saint-Aignan, France
| | - Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, BioClinicum, Karolinska Institutet, 17164 Stockholm, Sweden
| | - Mathieu Salaun
- Normandie Univ, UNIROUEN, LITIS EA 4108; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Justine Dampierre
- Normandie Univ, UNIROUEN, LITIS EA 4108; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - David Vaudry
- Normandie Univ, UNIROUEN, PRIMACEN; Institute for Research and Innovation in Biomedicine (IRIB), Mont-Saint-Aignan, France
| | - Jean-Paul Marie
- Normandie Univ, UNIROUEN, GRHV EA3830; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Nicolas Guérout
- Normandie Univ, UNIROUEN, GRHV EA3830; Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| |
Collapse
|
892
|
Galloway DA, Gowing E, Setayeshgar S, Kothary R. Inhibitory milieu at the multiple sclerosis lesion site and the challenges for remyelination. Glia 2019; 68:859-877. [PMID: 31441132 DOI: 10.1002/glia.23711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/26/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
Regeneration of myelin, following injury, can occur within the central nervous system to reinstate proper axonal conductance and provide trophic support. Failure to do so renders the axons vulnerable, leading to eventual degeneration, and neuronal loss. Thus, it is essential to understand the mechanisms by which remyelination or failure to remyelinate occur, particularly in the context of demyelinating and neurodegenerative disorders. In multiple sclerosis, oligodendrocyte progenitor cells (OPCs) migrate to lesion sites to repair myelin. However, during disease progression, the ability of OPCs to participate in remyelination diminishes coincident with worsening of the symptoms. Remyelination is affected by a broad range of cues from intrinsic programming of OPCs and extrinsic local factors to the immune system and other systemic elements including diet and exercise. Here we review the literature on these diverse inhibitory factors and the challenges they pose to remyelination. Results spanning several disciplines from fundamental preclinical studies to knowledge gained in the clinic will be discussed.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Elizabeth Gowing
- Neurosciences Department, Faculty of Medicine, Centre de recherche du CHUM, Université de Montreal, Montreal, Quebec, Canada
| | - Solmaz Setayeshgar
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Department of Biochemistry, Microbiology and Immunology, and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
893
|
Escartin C, Guillemaud O, Carrillo-de Sauvage MA. Questions and (some) answers on reactive astrocytes. Glia 2019; 67:2221-2247. [PMID: 31429127 DOI: 10.1002/glia.23687] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/12/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
Astrocytes are key cellular partners for neurons in the central nervous system. Astrocytes react to virtually all types of pathological alterations in brain homeostasis by significant morphological and molecular changes. This response was classically viewed as stereotypical and is called astrogliosis or astrocyte reactivity. It was long considered as a nonspecific, secondary reaction to pathological conditions, offering no clues on disease-causing mechanisms and with little therapeutic value. However, many studies over the last 30 years have underlined the crucial and active roles played by astrocytes in physiology, ranging from metabolic support, synapse maturation, and pruning to fine regulation of synaptic transmission. This prompted researchers to explore how these new astrocyte functions were changed in disease, and they reported alterations in many of them (sometimes beneficial, mostly deleterious). More recently, cell-specific transcriptomics revealed that astrocytes undergo massive changes in gene expression when they become reactive. This observation further stressed that reactive astrocytes may be very different from normal, nonreactive astrocytes and could influence disease outcomes. To make the picture even more complex, both normal and reactive astrocytes were shown to be molecularly and functionally heterogeneous. Very little is known about the specific roles that each subtype of reactive astrocytes may play in different disease contexts. In this review, we have interrogated researchers in the field to identify and discuss points of consensus and controversies about reactive astrocytes, starting with their very name. We then present the emerging knowledge on these cells and future challenges in this field.
Collapse
Affiliation(s)
- Carole Escartin
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Univ. Paris Sud, Univ. Paris-Saclay, UMR 9199, Neurodegenerative Disease Laboratory, Fontenay-aux-Roses, France
| | - Océane Guillemaud
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Univ. Paris Sud, Univ. Paris-Saclay, UMR 9199, Neurodegenerative Disease Laboratory, Fontenay-aux-Roses, France
| | - Maria-Angeles Carrillo-de Sauvage
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Univ. Paris Sud, Univ. Paris-Saclay, UMR 9199, Neurodegenerative Disease Laboratory, Fontenay-aux-Roses, France
| |
Collapse
|
894
|
Wang Q, Gao R, Wang M, Chen Q, Xiao M, Li Z, Wang L, Chen C. Spatiotemporal expression patterns of Galectin-3 in perinatal rat hypoxic-ischemic brain injury model. Neurosci Lett 2019; 711:134439. [PMID: 31425825 DOI: 10.1016/j.neulet.2019.134439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/03/2019] [Accepted: 08/15/2019] [Indexed: 12/28/2022]
Abstract
In this research, we intended to evaluate the expression pattern, distribution and sources of Galectin-3 (Gal-3) in perinatal hypoxic-ischemic brain injury rat model. Postnatal day 3 Sprague-Dawley rat pups were subjected to right carotid artery ligation followed by 2.5 h of hypoxia (6% oxygen). Expression and distribution of Gal-3 were evaluated by western blotting and immunofluorescence. Sources of Gal-3 were evaluated by double staining with neuronic, oligodendrocytic, astrocytic, microglial and endotheliocytic markers. Our results indicated Gal-3 significantly upregulated from 12 h and maintained an increasing tendency within 72 h post injury. Although the relative expression of Gal-3 decreased after 72 h, we detected significant differences until 14d. We found Gal-3 started to distribute in cortex and thalamus area and maintained an increasing tendency. Gal-3 could be detected in cortex, thalamus, corpus callosum and hippocampus area at 72 h post injury. After that, expression of Gal-3 in cortex and thalamus area downregulated, the expression in corpus callosum and hippocampus area vanished. We found astrocyte, microglia, neuron and endotheliocyte were sources of Gal-3 in cortex area; astrocyte, microglia and endotheliocyte were sources of Gal-3 in thalamus area; oligodendrocyte precursor cell and endotheliocyte were sources of Gal-3 in corpus callosum; neuron, microglia and endotheliocyte were sources of Gal-3 in hippocampus. In conclusion, we demonstrated spatiotemporal expression patterns of Galectin-3 post perinatal hypoxic-ischemic brain injury in this research.
Collapse
Affiliation(s)
- Qian Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Ruiwei Gao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Minjie Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Qiufan Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Mili Xiao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Zhihua Li
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Laishuan Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China.
| |
Collapse
|
895
|
Epac2 Elevation Reverses Inhibition by Chondroitin Sulfate Proteoglycans In Vitro and Transforms Postlesion Inhibitory Environment to Promote Axonal Outgrowth in an Ex Vivo Model of Spinal Cord Injury. J Neurosci 2019; 39:8330-8346. [PMID: 31409666 DOI: 10.1523/jneurosci.0374-19.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Millions of patients suffer from debilitating spinal cord injury (SCI) without effective treatments. Elevating cAMP promotes CNS neuron growth in the presence of growth-inhibiting molecules. cAMP's effects on neuron growth are partly mediated by Epac, comprising Epac1 and Epac2; the latter predominantly expresses in postnatal neural tissue. Here, we hypothesized that Epac2 activation would enhance axonal outgrowth after SCI. Using in vitro assays, we demonstrated, for the first time, that Epac2 activation using a specific soluble agonist (S-220) significantly enhanced neurite outgrowth of postnatal rat cortical neurons and markedly overcame the inhibition by chondroitin sulfate proteoglycans and mature astrocytes on neuron growth. We further investigated the novel potential of Epac2 activation in promoting axonal outgrowth by an ex vivo rat model of SCI mimicking post-SCI environment in vivo and by delivering S-220 via a self-assembling Fmoc-based hydrogel that has suitable properties for SCI repair. We demonstrated that S-220 significantly enhanced axonal outgrowth across the lesion gaps in the organotypic spinal cord slices, compared with controls. Furthermore, we elucidated, for the first time, that Epac2 activation profoundly modulated the lesion environment by reducing astrocyte/microglial activation and transforming astrocytes into elongated morphology that guided outgrowing axons. Finally, we showed that S-220, when delivered by the gel at 3 weeks after contusion SCI in male adult rats, resulted in significantly better locomotor performance for up to 4 weeks after treatment. Our data demonstrate a promising therapeutic potential of S-220 in SCI, via beneficial effects on neurons and glia after injury to facilitate axonal outgrowth.SIGNIFICANCE STATEMENT During development, neuronal cAMP levels decrease significantly compared with the embryonic stage when the nervous system is established. This has important consequences following spinal cord injury, as neurons fail to regrow. Elevating cAMP levels encourages injured CNS neurons to sprout and extend neurites. We have demonstrated that activating its downstream effector, Epac2, enhances neurite outgrowth in vitro, even in the presence of an inhibitory environment. Using a novel biomaterial-based drug delivery system in the form of a hydrogel to achieve local delivery of an Epac2 agonist, we further demonstrated that specific activation of Epac2 enhances axonal outgrowth and minimizes glial activation in an ex vivo model of spinal cord injury, suggesting a new strategy for spinal cord repair.
Collapse
|
896
|
Korotkov A, Broekaart DWM, Banchaewa L, Pustjens B, van Scheppingen J, Anink JJ, Baayen JC, Idema S, Gorter JA, van Vliet EA, Aronica E. microRNA-132 is overexpressed in glia in temporal lobe epilepsy and reduces the expression of pro-epileptogenic factors in human cultured astrocytes. Glia 2019; 68:60-75. [PMID: 31408236 PMCID: PMC6899748 DOI: 10.1002/glia.23700] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/26/2022]
Abstract
Temporal lobe epilepsy (TLE) is a chronic neurological disease in humans, which is refractory to pharmacological treatment in about 30% of the patients. Reactive glial cells are thought to play a major role during the development of epilepsy (epileptogenesis) via regulation of brain inflammation and remodeling of the extracellular matrix (ECM). These processes can be regulated by microRNAs (miRs), a class of small non‐coding RNAs, which can control entire gene networks at a post‐transcriptional level. The expression of miRs is known to change dynamically during epileptogenesis. miR‐132 is one of the most commonly upregulated miRs in animal TLE models with important roles shown in neurons. However, the possible role of miR‐132 in glia remains largely unknown. The aim of this study was to characterize the cell‐type specific expression of miR‐132 in the hippocampus of patients with TLE and during epileptogenesis in a rat TLE model. Furthermore, the potential role of miR‐132 was investigated by transfection of human primary cultured astrocytes that were stimulated with the cytokines IL‐1β or TGF‐β1. We showed an increased expression of miR‐132 in the human and rat epileptogenic hippocampus, particularly in glial cells. Transfection of miR‐132 in human primary astrocytes reduced the expression of pro‐epileptogenic COX‐2, IL‐1β, TGF‐β2, CCL2, and MMP3. This suggests that miR‐132, particularly in astrocytes, represents a potential therapeutic target that warrants further in vivo investigation.
Collapse
Affiliation(s)
- Anatoly Korotkov
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Diede W M Broekaart
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Leyla Banchaewa
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Ben Pustjens
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jackelien van Scheppingen
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Jasper J Anink
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Johannes C Baayen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Sander Idema
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| |
Collapse
|
897
|
Human GNPTAB stuttering mutations engineered into mice cause vocalization deficits and astrocyte pathology in the corpus callosum. Proc Natl Acad Sci U S A 2019; 116:17515-17524. [PMID: 31405983 DOI: 10.1073/pnas.1901480116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Stuttering is a common neurodevelopmental disorder that has been associated with mutations in genes involved in intracellular trafficking. However, the cellular mechanisms leading to stuttering remain unknown. Engineering a mutation in N-acetylglucosamine-1-phosphate transferase subunits α and β (GNPTAB) found in humans who stutter into the mouse Gnptab gene resulted in deficits in the flow of ultrasonic vocalizations similar to speech deficits of humans who stutter. Here we show that other human stuttering mutations introduced into this mouse gene, Gnptab Ser321Gly and Ala455Ser, produce the same vocalization deficit in 8-day-old pup isolation calls and do not affect other nonvocal behaviors. Immunohistochemistry showed a marked decrease in staining of astrocytes, particularly in the corpus callosum of the Gnptab Ser321Gly homozygote mice compared to wild-type littermates, while the staining of cerebellar Purkinje cells, oligodendrocytes, microglial cells, and dopaminergic neurons was not significantly different. Diffusion tensor imaging also detected deficits in the corpus callosum of the Gnptab Ser321Gly mice. Using a range of cell type-specific Cre-drivers and a Gnptab conditional knockout line, we found that only astrocyte-specific Gnptab-deficient mice displayed a similar vocalization deficit. These data suggest that vocalization defects in mice carrying human stuttering mutations in Gnptab derive from abnormalities in astrocytes, particularly in the corpus callosum, and provide support for hypotheses that focus on deficits in interhemispheric communication in stuttering.
Collapse
|
898
|
Xu AK, Gong Z, He YZ, Xia KS, Tao HM. Comprehensive therapeutics targeting the corticospinal tract following spinal cord injury. J Zhejiang Univ Sci B 2019; 20:205-218. [PMID: 30829009 DOI: 10.1631/jzus.b1800280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Spinal cord injury (SCI), which is much in the public eye, is still a refractory disease compromising the well-being of both patients and society. In spite of there being many methods dealing with the lesion, there is still a deficiency in comprehensive strategies covering all facets of this damage. Further, we should also mention the structure called the corticospinal tract (CST) which plays a crucial role in the motor responses of organisms, and it will be the focal point of our attention. In this review, we discuss a variety of strategies targeting different dimensions following SCI and some treatments that are especially efficacious to the CST are emphasized. Over recent decades, researchers have developed many effective tactics involving five approaches: (1) tackle more extensive regions; (2) provide a regenerative microenvironment; (3) provide a glial microenvironment; (4) transplantation; and (5) other auxiliary methods, for instance, rehabilitation training and electrical stimulation. We review the basic knowledge on this disease and correlative treatments. In addition, some well-formulated perspectives and hypotheses have been delineated. We emphasize that such a multifaceted problem needs combinatorial approaches, and we analyze some discrepancies in past studies. Finally, for the future, we present numerous brand-new latent tactics which have great promise for curbing SCI.
Collapse
Affiliation(s)
- An-Kai Xu
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, China
| | - Zhe Gong
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, China
| | - Yu-Zhe He
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, China
| | - Kai-Shun Xia
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, China
| | - Hui-Min Tao
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
899
|
Fawcett JW. The Struggle to Make CNS Axons Regenerate: Why Has It Been so Difficult? Neurochem Res 2019; 45:144-158. [PMID: 31388931 PMCID: PMC6942574 DOI: 10.1007/s11064-019-02844-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/09/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
Axon regeneration in the CNS is inhibited by many extrinsic and intrinsic factors. Because these act in parallel, no single intervention has been sufficient to enable full regeneration of damaged axons in the adult mammalian CNS. In the external environment, NogoA and CSPGs are strongly inhibitory to the regeneration of adult axons. CNS neurons lose intrinsic regenerative ability as they mature: embryonic but not mature neurons can grow axons for long distances when transplanted into the adult CNS, and regeneration fails with maturity in in vitro axotomy models. The causes of this loss of regeneration include partitioning of neurons into axonal and dendritic fields with many growth-related molecules directed specifically to dendrites and excluded from axons, changes in axonal signalling due to changes in expression and localization of receptors and their ligands, changes in local translation of proteins in axons, and changes in cytoskeletal dynamics after injury. Also with neuronal maturation come epigenetic changes in neurons, with many of the transcription factor binding sites that drive axon growth-related genes becoming inaccessible. The overall aim for successful regeneration is to ensure that the right molecules are expressed after axotomy and to arrange for them to be transported to the right place in the neuron, including the damaged axon tip.
Collapse
Affiliation(s)
- James W Fawcett
- John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
- Centre of Reconstructive Neuroscience, Institute for Experimental Medicine ASCR, Prague, Czech Republic.
| |
Collapse
|
900
|
Gu Y, Cheng X, Huang X, Yuan Y, Qin S, Tan Z, Wang D, Hu X, He C, Su Z. Conditional ablation of reactive astrocytes to dissect their roles in spinal cord injury and repair. Brain Behav Immun 2019; 80:394-405. [PMID: 30959174 DOI: 10.1016/j.bbi.2019.04.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 12/22/2022] Open
Abstract
Astrocytes become reactive in response to spinal cord injury (SCI) and ultimately form a histologically apparent glial scar at the lesion site. It is controversial whether astrocytic scar is detrimental or beneficial to the axonal regeneration and SCI repair. Therefore, much effort has focused on understanding the functions of reactive astrocytes. Here, we used a lentivirus-mediated herpes simplex thymidine kinase/ganciclovir (HSVtk/GCV) system to selectively kill scar-forming reactive proliferating astrocytes. The suicide gene expression was regulated by human glial fibrillary acidic protein (hGFAP) promoter, which is active primarily in astrocytes. Conditional ablation of reactive astrocytes in a mouse SCI model with crush injury impeded glial scar formation and resulted in widespread infiltration of inflammatory cells, increased neuronal loss, and severe tissue degeneration, which ultimately led to the failure of spontaneous functional recovery. These results suggest that reactive proliferating astrocytes play key roles in the healing process after SCI, shedding light on the potential benefit for the repair after central nervous system (CNS) injury.
Collapse
Affiliation(s)
- Yakun Gu
- Center for Brain Disorders Research, Capital Medical University, Center of NeuralInjury and Repair, Beijing Institute for Brain Disorders, Beijing, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China
| | - Xueyan Cheng
- Center for Brain Disorders Research, Capital Medical University, Center of NeuralInjury and Repair, Beijing Institute for Brain Disorders, Beijing, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China
| | - Xiao Huang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China
| | - Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China
| | - Zijian Tan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China
| | - Dan Wang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China
| | - Xin Hu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China; Department of Neurological Surgery, Xixi Hospital of Hangzhou, Hangzhou, China
| | - Cheng He
- Center for Brain Disorders Research, Capital Medical University, Center of NeuralInjury and Repair, Beijing Institute for Brain Disorders, Beijing, China; Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, China.
| |
Collapse
|