1
|
He J, Liu Q, Guo J, Wu D, Guo Y. Circulatory factors in stroke protection and recovery. Brain Res 2025; 1855:149594. [PMID: 40122323 DOI: 10.1016/j.brainres.2025.149594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Over the past decade, the management of acute ischemic stroke has undergone a paradigm shift, especially a longer time-window and a wider indication for endovascular treatments. However, many patients still have long-term dysfunction despite the best medical care at present. Based on findings from innovative proteomic and transcriptomic technologies, researchers have identified an array of novel or previously underappreciated circulatory factors that play pivotal roles in mediating post-injuries brain communication. Thus, the previous concept of the brain as a privileged compartment isolated from the rest of the body has been replaced by the novel consensus that brain bidirectionally interacts with the other organs after brain diseases. In this review, we make a summary of several axes that connect the brain with the rest of the body after stroke. More importantly, we summarize several circulatory factors that play pivotal roles in fostering post-stroke functional recovery in the chronic stage. Special attention is given to the instrumental role of circulatory signals, positing them as significant contributors to the complex process of brain function recovery and as translational therapeutic targets for ischemic stroke in future studies.
Collapse
Affiliation(s)
- Jiachen He
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin Medical University, Harbin 150081 Heilongjiang, China
| | - Qi Liu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Jiaqi Guo
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China
| | - Di Wu
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 100053, China; Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 10053, China.
| | - Yansu Guo
- Beijing Geriatric Healthcare Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
2
|
Kumari S, Dhapola R, Sharma P, Nagar P, Medhi B, HariKrishnaReddy D. The impact of cytokines in neuroinflammation-mediated stroke. Cytokine Growth Factor Rev 2024; 78:105-119. [PMID: 39004599 DOI: 10.1016/j.cytogfr.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024]
Abstract
Cerebral stroke is ranked as the third most common contributor to global mortality and disability. The involvement of inflammatory mechanisms, both peripherally and within the CNS, holds significance in the pathophysiological cascades following the initiation of stroke. After the onset of acute stroke, predominantly ischemic, a subsequent phase of neuroinflammation ensues. It is a dual-effect process that not only exacerbates injury, leading to cell death, but paradoxically, it also serves a shielding role in facilitating recovery. Cytokines serve as pivotal mediators within the inflammatory cascade, actively contributing to the progression of ischemic damage. Stroke is followed by increased expression of pro-inflammatory cytokines including TNF-α, IL-1β, IL-6, etc. leading to the recruitment and stimulation of glial cells and peripheral leukocytes at the site of injury, promoting neuroinflammation. Cytokines can directly induce neuronal injury and death through various mechanisms, including excitotoxicity, oxidative stress, HPA-axis activation, secretion of matrix metalloproteinase and apoptosis. They can also amplify the inflammatory response, leading to further neuronal damage. Therapeutic strategies aimed at modulating cytokine release, immune response and cytokine signalling or activity are being explored as potential interventions to mitigate neuroinflammation and its detrimental effects in stroke. In this review, we have given a concise summary of our current knowledge of the function of various cytokines, brain inflammation and various signalling and molecular pathways including JAK/STAT3, TGF-β/Smad, MAPK, HMGB1/TLR and NF-κB modulated cytokines regulation in stroke. Therapeutic agents such as MCC950, genistein, edaravone, minocycline, etc. targeting various cytokines-associated signalling pathways have shown efficacy in preclinical and clinical trials reducing the pathophysiology of the illness were also addressed in this study.
Collapse
Affiliation(s)
- Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Pushank Nagar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| |
Collapse
|
3
|
Li D, Liu C, Wang H, Li Y, Wang Y, An S, Sun S. The Role of Neuromodulation and Potential Mechanism in Regulating Heterotopic Ossification. Neurochem Res 2024; 49:1628-1642. [PMID: 38416374 DOI: 10.1007/s11064-024-04118-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/17/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
Heterotopic ossification (HO) is a pathological process characterized by the aberrant formation of bone in muscles and soft tissues. It is commonly triggered by traumatic brain injury, spinal cord injury, and burns. Despite a wide range of evidence underscoring the significance of neurogenic signals in proper bone remodeling, a clear understanding of HO induced by nerve injury remains rudimentary. Recent studies suggest that injury to the nervous system can activate various signaling pathways, such as TGF-β, leading to neurogenic HO through the release of neurotrophins. These pathophysiological changes lay a robust groundwork for the prevention and treatment of HO. In this review, we collected evidence to elucidate the mechanisms underlying the pathogenesis of HO related to nerve injury, aiming to enhance our understanding of how neurological repair processes can culminate in HO.
Collapse
Affiliation(s)
- Dengju Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong First Medical University, Jinan, Shandong, China
| | - Changxing Liu
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Haojue Wang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yunfeng Li
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yaqi Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Senbo An
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
| | - Shui Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
4
|
Bedolla A, Wegman E, Weed M, Stevens MK, Ware K, Paranjpe A, Alkhimovitch A, Ifergan I, Taranov A, Peter JD, Gonzalez RMS, Robinson JE, McClain L, Roskin KM, Greig NH, Luo Y. Adult microglial TGFβ1 is required for microglia homeostasis via an autocrine mechanism to maintain cognitive function in mice. Nat Commun 2024; 15:5306. [PMID: 38906887 PMCID: PMC11192737 DOI: 10.1038/s41467-024-49596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
While TGF-β signaling is essential for microglial function, the cellular source of TGF-β1 ligand and its spatial regulation remains unclear in the adult CNS. Our data supports that microglia but not astrocytes or neurons are the primary producers of TGF-β1 ligands needed for microglial homeostasis. Microglia-Tgfb1 KO leads to the activation of microglia featuring a dyshomeostatic transcriptome that resembles disease-associated, injury-associated, and aged microglia, suggesting microglial self-produced TGF-β1 ligands are important in the adult CNS. Astrocytes in MG-Tgfb1 inducible (i)KO mice show a transcriptome profile that is closely aligned with an LPS-associated astrocyte profile. Additionally, using sparse mosaic single-cell microglia KO of TGF-β1 ligand we established an autocrine mechanism for signaling. Here we show that MG-Tgfb1 iKO mice present cognitive deficits, supporting that precise spatial regulation of TGF-β1 ligand derived from microglia is required for the maintenance of brain homeostasis and normal cognitive function in the adult brain.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Max Weed
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | | | - Kierra Ware
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Information Services for Research, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Anastasia Alkhimovitch
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Igal Ifergan
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Aleksandr Taranov
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Joshua D Peter
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Rosa Maria Salazar Gonzalez
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
| | - J Elliott Robinson
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
| | - Lucas McClain
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA
| | - Krishna M Roskin
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, US
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH, USA.
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH, USA.
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
5
|
Chiu YS, Wu KJ, Yu SJ, Wu KL, Hsieh CY, Chou YS, Chen KY, Wang YS, Bae EK, Hung TW, Lin SH, Lin CH, Hsu SC, Wang Y, Chen YH. Transplantation of Exosomes Derived From Human Wharton's Jelly Mesenchymal Stromal Cells Enhances Functional Improvement in Stroke Rats. Cell Transplant 2024; 33:9636897241296366. [PMID: 39624898 PMCID: PMC11613244 DOI: 10.1177/09636897241296366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/28/2024] [Accepted: 10/15/2024] [Indexed: 12/06/2024] Open
Abstract
Cerebral ischemic stroke is a major cerebrovascular disease and the leading cause of adult disability. We and others previously demonstrated that transplantation of human Wharton's jelly mesenchymal stromal cells (WJ-MSCs) attenuated neuronal damage and promoted functional improvement in stroke animals. This study aimed to investigate the protective effects of human WJ-MSC exosome (Exo) transplant in cellular and rat models of cerebral stroke. Administration of Exo significantly antagonized glutamate-mediated neuronal loss and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-X nick end labeling (TUNEL) in rat primary cortical neuronal cultures. Adult male rats underwent a 60-min middle cerebral artery occlusion (MCAo); Exo or vehicle was injected through the tail vein 5-10 min after the MCAo. Two days later, the rats underwent a series of behavioral tests. Stroke rats receiving Exo developed a significant improvement in locomotor function and forelimb strength while reductions in body asymmetry and Bederson's neurological score. After the behavioral test, brain tissues were harvested for histological and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) analyses. Animals receiving Exo had less infarction volume, measured by 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Transplantation of Exo increased the expression of protective neurotrophic factors (BMP7, GDNF) and anti-apoptotic factors (Bcl2, Bcl-xL) in the ischemic brain. These findings suggest that early post-treatment with WJ-MSC Exo, given non-invasively through the vein, improved functional recovery and reduced brain damage in the stroke brain.
Collapse
Affiliation(s)
- Yu-Sung Chiu
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
| | - Kuo-Jen Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kun-Lieh Wu
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
- Department of Electrical Engineering, I-Shou University, Kaohsiung, Taiwan
| | | | | | - Kuan-Yu Chen
- YJ Biotechnology Co., Ltd., New Taipei City, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Tsai-Wei Hung
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Hsun Lin
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Hsueh Lin
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Shu-Ching Hsu
- Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- PhD Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung, Taiwan
- Immunology Research and Development Center, China Medical University, Taichung City, Taiwan
- Department of Life Sciences, Tzu Chi University, Hualien, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yun-Hsiang Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
6
|
Bedolla A, Wegman E, Weed M, Paranjpe A, Alkhimovitch A, Ifergan I, McClain L, Luo Y. Microglia-derived TGF-β1 ligand maintains microglia homeostasis via autocrine mechanism and is critical for normal cognitive function in adult mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547814. [PMID: 37461569 PMCID: PMC10349967 DOI: 10.1101/2023.07.05.547814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
While TGF-β signaling is essential for microglial function, the cellular source of TGF-β ligand and its spatial regulation remains unclear in the adult CNS. Our data support that microglia, not astrocytes or neurons, are the primary producers of TGF-β1 ligands needed for microglial homeostasis. Microglia (MG)-Tgfb1 inducible knockout (iKO) leads to the activation of microglia featuring a dyshomeostatic transcriptomic profile that resembles disease-associated microglia (DAMs), injury-associated microglia, and aged microglia, suggesting that microglial self-produced TGF-β1 ligands are important in the adult CNS. Interestingly, astrocytes in MG-Tgfb1 iKO mice show a transcriptome profile that closely aligns with A1-like astrocytes. Additionally, using sparse mosaic single-cell microglia iKO of TGF-β1 ligand, we established an autocrine mechanism for TGF-β signaling. Importantly MG-Tgfb1 iKO mice show cognitive deficits, supporting that precise spatial regulation of TGF-β1 ligand derived from microglia is critical for the maintenance of brain homeostasis and normal cognitive function in the adult brain.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Max Weed
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Aditi Paranjpe
- Information Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anastasia Alkhimovitch
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Igal Ifergan
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Lucas McClain
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
7
|
Zhang Z, Sun GY, Ding S. Glial Cell Line-Derived Neurotrophic Factor and Focal Ischemic Stroke. Neurochem Res 2021; 46:2638-2650. [PMID: 33591443 PMCID: PMC8364922 DOI: 10.1007/s11064-021-03266-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 11/29/2022]
Abstract
Focal ischemic stroke (FIS) is a leading cause of human debilitation and death. Following the onset of a FIS, the brain experiences a series of spatiotemporal changes which are exemplified in different pathological processes. One prominent feature of FIS is the development of reactive astrogliosis and glial scar formation in the peri-infarct region (PIR). During the subacute phase, astrocytes in PIR are activated, referred to as reactive astrocytes (RAs), exhibit changes in morphology (hypotrophy), show an increased proliferation capacity, and altered gene expression profile, a phenomenon known as reactive astrogliosis. Subsequently, the morphology of RAs remains stable, and proliferation starts to decline together with the formation of glial scars. Reactive astrogliosis and glial scar formation eventually cause substantial tissue remodeling and changes in permanent structure around the PIR. Glial cell line-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line and regarded as a potent survival neurotrophic factor. Under normal conditions, GDNF is expressed in neurons but is upregulated in RAs after FIS. This review briefly describes properties of GDNF, its receptor-mediated signaling pathways, as well as recent studies regarding the role of RAs-derived GDNF in neuronal protection and brain recovery. These results provide evidence suggesting an important role of RA-derived GDNF in intrinsic brain repair and recovery after FIS, and thus targeting GDNF in RAs may be effective for stroke therapy.
Collapse
Affiliation(s)
- Zhe Zhang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri-Columbia, Columbia, MO, 65211, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA.
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, Columbia, MO, 65211, USA.
- Dalton Cardiovascular Research Center, Department of Biomedical, Biological and Chemical Engineering, University of Missouri-Columbia, 134 Research Park Drive, Columbia, MO, 65211, USA.
| |
Collapse
|
8
|
Vogel DDS, Ortiz-Villatoro NN, Araújo NS, Marques MJG, Aimbire F, Scorza FA, Scorza CA, Albertini R. Transcranial low-level laser therapy in an in vivo model of stroke: Relevance to the brain infarct, microglia activation and neuroinflammation. JOURNAL OF BIOPHOTONICS 2021; 14:e202000500. [PMID: 33580734 DOI: 10.1002/jbio.202000500] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 06/12/2023]
Abstract
Stroke is the main cause of death and functional disability. The available therapy affects only 5% of patients, and new therapeutic approaches have been constantly tested. Transcranial photobiomodulation (PBM) is promising for its neuroprotective effect on brain injuries. Thus, the present study investigated the PBM effects in an in vivo model of ischemic stroke induced by photothrombosis (PT). Five different groups of Wistar rats were submitted or not to a daily dose of fish oil or/and laser sessions for 2 months. The ischemia volume was evaluated by stereology; GFAP, Iba and NeuN by immunohistochemistry; TNF-α, IL-1β, IL-6, IL-10 and TGF-β by ELISA assay. PBM influenced both the lesion volume and the GFAP. Furthermore, PBM and Ω-3 or both reduced Iba RNAm. PBM reduced TNF-α, IL-1β, IL-6, brain damage, neuroinflammation and microglial activation, and it increased astroglial activity in peri-lesioned region after stroke.
Collapse
Affiliation(s)
- Débora D S Vogel
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Nancy N Ortiz-Villatoro
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Noemi S Araújo
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Márcia Jonathas Guimarães Marques
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Flavio Aimbire
- Programa de Pós-graduação em Medicina Translacional, Departamento de Ciência e Tecnologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Fúlvio A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Carla A Scorza
- Disciplina de Neurociência, Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Regiane Albertini
- Programa de Pós-graduação em Ciência do Movimento Humano e Reabilitação, Departamento de Ciência e Tecnologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
9
|
Sotiropoulos MG, Chitnis T. Opposing and potentially antagonistic effects of BMP and TGF-β in multiple sclerosis: The "Yin and Yang" of neuro-immune Signaling. J Neuroimmunol 2020; 347:577358. [PMID: 32795734 DOI: 10.1016/j.jneuroim.2020.577358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/31/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Bone Morphogenetic Proteins (BMP) and Transforming Growth Factor-beta (TGF-β) are cytokines with similar receptors and messengers. They are important for immune cell function, with BMPs exerting mainly proinflammatory but also anti-inflammatory effects, and TGF-β suppressing inflammation. Patients with Multiple Sclerosis exhibit BMP overactivity and suppressed TGF-β signaling. This dysregulated signaling participates in the crosstalk between infiltrating immune cells and glia, where BMP inhibits remyelination. Reciprocal antagonism between the two pathways takes place via a variety of mechanisms. Although this antagonism has not been studied in the setting of Multiple Sclerosis, it could inform further research and treatment discovery.
Collapse
Affiliation(s)
- Marinos G Sotiropoulos
- Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA.
| | - Tanuja Chitnis
- Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Ceci M, Mariano V, Romano N. Zebrafish as a translational regeneration model to study the activation of neural stem cells and role of their environment. Rev Neurosci 2019; 30:45-66. [PMID: 30067512 DOI: 10.1515/revneuro-2018-0020] [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: 03/04/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
The review is an overview of the current knowledge of neuronal regeneration properties in mammals and fish. The ability to regenerate the damaged parts of the nervous tissue has been demonstrated in all vertebrates. Notably, fish and amphibians have the highest capacity for neurogenesis, whereas reptiles and birds are able to only regenerate specific regions of the brain, while mammals have reduced capacity for neurogenesis. Zebrafish (Danio rerio) is a promising model of study because lesions in the brain or complete cross-section of the spinal cord are followed by an effective neuro-regeneration that successfully restores the motor function. In the brain and the spinal cord of zebrafish, stem cell activity is always able to re-activate the molecular programs required for central nervous system regeneration. In mammals, traumatic brain injuries are followed by reduced neurogenesis and poor axonal regeneration, often insufficient to functionally restore the nervous tissue, while spinal injuries are not repaired at all. The environment that surrounds the stem cell niche constituted by connective tissue and stimulating factors, including pro-inflammation molecules, seems to be a determinant in triggering stem cell proliferation and/or the trans-differentiation of connective elements (mainly fibroblasts). Investigating and comparing the neuronal regeneration in zebrafish and mammals may lead to a better understanding of the mechanisms behind neurogenesis, and the failure of the regenerative response in mammals, first of all, the role of inflammation, considered the main inhibitor of the neuronal regeneration.
Collapse
Affiliation(s)
- Marcello Ceci
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicla Romano
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| |
Collapse
|
11
|
Guo M, Ma X, Feng Y, Han S, Dong Q, Cui M, Zhao Y. In chronic hypoxia, glucose availability and hypoxic severity dictate the balance between HIF-1 and HIF-2 in astrocytes. FASEB J 2019; 33:11123-11136. [PMID: 31298941 DOI: 10.1096/fj.201900402rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Astrocyte function is an important contributor to cellular viability during brain hypoxia and ischemia. Levels of the hypoxia-inducible transcription factors (HIFs) HIF-1 and HIF-2 are increased in hypoxic conditions and impact the neuroprotective properties of astrocytes. For example, HIF-2 induces levels of erythropoietin (EPO), a neuroprotectant, by astrocytes. In contrast, HIF-1 activity in astrocytes diminishes the viability of neurons in cocultures during hypoxia. Thus, HIF-1 and HIF-2 may have opposing effects on astrocytes. In this study, we explore the balance of HIF-1 and HIF-2 signaling in astrocytes during chronic (1-7 d) hypoxia while altering the degree of hypoxia and glucose availability. In addition, we investigate the effects of these conditions on neuron apoptosis. During exposure to chronic moderate hypoxia (2% O2) and plentiful glucose (10 mM), HIF-2 and EPO abundance increases from d 1 to 7. Similarly, pretreatment with moderate hypoxia markedly increases the abundance of HIF-2 and EPO when astrocytes are subsequently exposed to severe hypoxia (0.5% O2; 24 h) in 10 mM glucose, which inhibits neuron apoptosis in coculture. Although HIF-1 targets the expression increase during the 7 d in chronic moderate hypoxia (2% O2) and limited glucose (2 mM), further exposure to severe hypoxia (0.5% O2; 24 h) induces a decrease of most HIF-1 targets in astrocytes. Notably, in astrocyte exposure to 2% O2 prior to 0.5% O2, the expression of iNOS, an HIF-1-regulated protein, keeps increasing when glucose is limited, whereas EPO and VEGF abundance is suppressed, inducing increased apoptosis of neurons in coculture under limited glucose (2 mM). Thus, both hypoxic severity and glucose abundance regulate the balance of HIF-1 and HIF-2 activity in astrocytes, leading to diverse effects on neurons. These results could have important implications on the adaptive or pathologic role of astrocytes during chronic hypoxia and ischemia.-Guo, M., Ma, X., Feng, Y., Han, S., Dong, Q., Cui, M., Zhao, Y. In chronic hypoxia, glucose availability and hypoxic severity dictate the balance between HIF-1 and HIF-2 in astrocytes.
Collapse
Affiliation(s)
- Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoye Ma
- Department of Neurology, The 10th People's Hospital, Tongji University, Shanghai, China
| | - Yiwei Feng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Sida Han
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, State Key Laboratory of Medical Neurobiology, Ministry of Education (MOE) Frontiers Center for Brain Science, Huashan Hospital, Fudan University, Shanghai, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanxin Zhao
- Department of Neurology, The 10th People's Hospital, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Barua S, Kim JY, Lee JE. Role of Agmatine on Neuroglia in Central Nervous System Injury. BRAIN & NEUROREHABILITATION 2019. [DOI: 10.12786/bn.2019.12.e2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Sumit Barua
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Ito M, Aswendt M, Lee AG, Ishizaka S, Cao Z, Wang EH, Levy SL, Smerin DL, McNab JA, Zeineh M, Leuze C, Goubran M, Cheng MY, Steinberg GK. RNA-Sequencing Analysis Revealed a Distinct Motor Cortex Transcriptome in Spontaneously Recovered Mice After Stroke. Stroke 2018; 49:2191-2199. [PMID: 30354987 PMCID: PMC6205731 DOI: 10.1161/strokeaha.118.021508] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/28/2018] [Indexed: 12/14/2022]
Abstract
Background and Purpose- Many restorative therapies have been used to study brain repair after stroke. These therapeutic-induced changes have revealed important insights on brain repair and recovery mechanisms; however, the intrinsic changes that occur in spontaneously recovery after stroke is less clear. The goal of this study is to elucidate the intrinsic changes in spontaneous recovery after stroke, by directly investigating the transcriptome of primary motor cortex in mice that naturally recovered after stroke. Methods- Male C57BL/6J mice were subjected to transient middle cerebral artery occlusion. Functional recovery was evaluated using the horizontal rotating beam test. A novel in-depth lesion mapping analysis was used to evaluate infarct size and locations. Ipsilesional and contralesional primary motor cortices (iM1 and cM1) were processed for RNA-sequencing transcriptome analysis. Results- Cluster analysis of the stroke mice behavior performance revealed 2 distinct recovery groups: a spontaneously recovered and a nonrecovered group. Both groups showed similar lesion profile, despite their differential recovery outcome. RNA-sequencing transcriptome analysis revealed distinct biological pathways in the spontaneously recovered stroke mice, in both iM1 and cM1. Correlation analysis revealed that 38 genes in the iM1 were significantly correlated with improved recovery, whereas 74 genes were correlated in the cM1. In particular, ingenuity pathway analysis highlighted the involvement of cAMP signaling in the cM1, with selective reduction of Adora2a (adenosine receptor A2A), Drd2 (dopamine receptor D2), and Pde10a (phosphodiesterase 10A) expression in recovered mice. Interestingly, the expressions of these genes in cM1 were negatively correlated with behavioral recovery. Conclusions- Our RNA-sequencing data revealed a panel of recovery-related genes in the motor cortex of spontaneously recovered stroke mice and highlighted the involvement of contralesional cortex in spontaneous recovery, particularly Adora2a, Drd2, and Pde10a-mediated cAMP signaling pathway. Developing drugs targeting these candidates after stroke may provide beneficial recovery outcome.
Collapse
MESH Headings
- Animals
- Cluster Analysis
- Cyclic AMP/metabolism
- Gene Expression Profiling
- Infarction, Middle Cerebral Artery/diagnostic imaging
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Magnetic Resonance Imaging
- Mice
- Motor Cortex/diagnostic imaging
- Motor Cortex/metabolism
- Motor Cortex/pathology
- Motor Cortex/physiopathology
- Phosphoric Diester Hydrolases/genetics
- RNA, Messenger/metabolism
- Receptor, Adenosine A2A/genetics
- Receptors, Dopamine D2/genetics
- Receptors, Prostaglandin E, EP4 Subtype/genetics
- Recovery of Function/genetics
- Remission, Spontaneous
- Sequence Analysis, RNA
- Signal Transduction
- Stroke/diagnostic imaging
- Stroke/genetics
- Stroke/pathology
- Stroke/physiopathology
Collapse
Affiliation(s)
- Masaki Ito
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | - Markus Aswendt
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | | | - Shunsuke Ishizaka
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | - Zhijuan Cao
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | - Eric H Wang
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | - Sabrina L Levy
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | - Daniel L Smerin
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | - Jennifer A McNab
- Department of Radiology (J.A.M., M.Z., C.L., M.G.), Stanford University School of Medicine, CA
| | - Michael Zeineh
- Department of Radiology (J.A.M., M.Z., C.L., M.G.), Stanford University School of Medicine, CA
| | - Christoph Leuze
- Department of Radiology (J.A.M., M.Z., C.L., M.G.), Stanford University School of Medicine, CA
| | - Maged Goubran
- Department of Radiology (J.A.M., M.Z., C.L., M.G.), Stanford University School of Medicine, CA
| | - Michelle Y Cheng
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| | - Gary K Steinberg
- From the Department of Neurosurgery (M.I., M.A., S.I., Z.C., E.H.W., S.L.L., D.L.S., M.Y.C., G.K.S.)
| |
Collapse
|
14
|
Navarro Quiroz E, Navarro Quiroz R, Ahmad M, Gomez Escorcia L, Villarreal JL, Fernandez Ponce C, Aroca Martinez G. Cell Signaling in Neuronal Stem Cells. Cells 2018; 7:E75. [PMID: 30011912 PMCID: PMC6070865 DOI: 10.3390/cells7070075] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/30/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023] Open
Abstract
The defining characteristic of neural stem cells (NSCs) is their ability to multiply through symmetric divisions and proliferation, and differentiation by asymmetric divisions, thus giving rise to different types of cells of the central nervous system (CNS). A strict temporal space control of the NSC differentiation is necessary, because its alterations are associated with neurological dysfunctions and, in some cases, death. This work reviews the current state of the molecular mechanisms that regulate the transcription in NSCs, organized according to whether the origin of the stimulus that triggers the molecular cascade in the CNS is internal (intrinsic factors) or whether it is the result of the microenvironment that surrounds the CNS (extrinsic factors).
Collapse
Affiliation(s)
- Elkin Navarro Quiroz
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
- School of Medicine, Universidad Rafael Nuñez, Cartagena 130001, Colombia.
| | - Roberto Navarro Quiroz
- Centro de Investigación en Salud para el Trópico, Universidad Cooperativa de Colombia, Santa Marta 470002, Colombia.
| | - Mostapha Ahmad
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
| | - Lorena Gomez Escorcia
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
| | | | | | - Gustavo Aroca Martinez
- Faculty of basic sciences and biomedical; Universidad Simón Bolívar, Barranquilla 080002, Colombia.
- Clinica de la Costa, Barranquilla 080002, Colombia.
| |
Collapse
|
15
|
Zammit V, Baron B, Ayers D. MiRNA Influences in Neuroblast Modulation: An Introspective Analysis. Genes (Basel) 2018; 9:genes9010026. [PMID: 29315268 PMCID: PMC5793179 DOI: 10.3390/genes9010026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB) is the most common occurring solid paediatric cancer in children under the age of five years. Whether of familial or sporadic origin, chromosome abnormalities contribute to the development of NB and cause dysregulation of microRNAs (miRNAs). MiRNAs are small non-coding, single stranded RNAs that target messenger RNAs at the post-transcriptional levels by repressing translation within all facets of human physiology. Such gene 'silencing' activities by miRNAs allows the development of regulatory feedback loops affecting multiple functions within the cell, including the possible differentiation of neural stem cell (NSC) lineage selection. Neurogenesis includes stages of self-renewal and fate specification of NSCs, migration and maturation of young neurones, and functional integration of new neurones into the neural circuitry, all of which are regulated by miRNAs. The role of miRNAs and their interaction in cellular processes are recognised aspects of cancer genetics, and miRNAs are currently employed as biomarkers for prognosis and tumour characterisation in multiple cancer models. Consequently, thorough understanding of the mechanisms of how these miRNAs interplay at the transcriptomic level will definitely lead to the development of novel, bespoke and efficient therapeutic measures, with this review focusing on the influences of miRNAs on neuroblast modulations leading to neuroblastoma.
Collapse
Affiliation(s)
- Vanessa Zammit
- National Blood Transfusion Service, St. Luke's Hospital, PTA1010 G'Mangia, Malta.
- School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LY, UK.
| | - Byron Baron
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
| | - Duncan Ayers
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
16
|
Bokara KK, Kim JH, Kim JY, Lee JE. Transfection of arginine decarboxylase gene increases the neuronal differentiation of neural progenitor cells. Stem Cell Res 2016; 17:256-265. [PMID: 27591482 DOI: 10.1016/j.scr.2016.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/26/2016] [Accepted: 08/16/2016] [Indexed: 10/21/2022] Open
Abstract
Growing evidence suggests that the clinical use of neural progenitor cells (NPCs) is hampered by heterogeneity, poor neuronal yield and low survival rate. Recently, we reported that retrovirus-delivered human arginine decarboxylase (hADC) genes improve cell survival against oxidative insult in murine NPCs in vitro. This study investigates whether the induced expression of hADC gene in mNPCs induces any significant change in the cell fate commitment. The evaluation of induced hADC gene function was assessed by knockdown of hADC gene using specific siRNA. The hADC gene delivery triggered higher expression of N-CAM, cell adhesion molecule and MAP-2, neuronal marker. However, the hADC gene knockdown showed downregulation of N-CAM and MAP-2 expression suggesting that hADC gene delivery favors cell fate commitment of mNPCs towards neuronal lineage. Neurite outgrowth was significantly longer in the hADC infected cells. The neurotrophic signal, BDNF aided in the neuronal commitment, differentiation, and maturation of hADC-mNPCs through PI3K and ERK1/2 activation. The induction of neuron-like differentiation is believed to be regulated by the expression of GSK-3β and Wnt/β-catenin signaling pathways. Our findings suggest that hADC gene delivery favors cell fate commitment of mNPCs towards neuronal lineage, bring new advances in the field of neurogenesis and cell therapy.
Collapse
Affiliation(s)
- Kiran Kumar Bokara
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; CSIR-Centre for Cellular and Molecular Biology, Medical Biotechnology Complex, ANNEXE II, Uppal Road, Uppal, Hyderabad 500007, India.
| | - Jae Hwan Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea; Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Jae Young Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; BK 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
17
|
Wang X, Xu J, Wang Y, Yang L, Li Z. Protective effects of BMP‐7 against tumor necrosis factor α‐induced oligodendrocyte apoptosis. Int J Dev Neurosci 2016; 53:10-17. [DOI: 10.1016/j.ijdevneu.2016.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/19/2016] [Accepted: 04/20/2016] [Indexed: 12/25/2022] Open
Affiliation(s)
- Xin Wang
- Department of AnesthesiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Anesthesia Research InstituteCentral South UniversityChangshaHunanChina
| | - Jun‐Mei Xu
- Department of AnesthesiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Anesthesia Research InstituteCentral South UniversityChangshaHunanChina
| | - Ya‐Ping Wang
- Department of AnesthesiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Anesthesia Research InstituteCentral South UniversityChangshaHunanChina
| | - Lin Yang
- Department of AnesthesiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Anesthesia Research InstituteCentral South UniversityChangshaHunanChina
| | - Zhi‐Jian Li
- Department of AnesthesiologyThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Anesthesia Research InstituteCentral South UniversityChangshaHunanChina
| |
Collapse
|
18
|
Glial influences on BBB functions and molecular players in immune cell trafficking. Biochim Biophys Acta Mol Basis Dis 2015; 1862:472-82. [PMID: 26454208 DOI: 10.1016/j.bbadis.2015.10.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) constitutes an elaborate structure formed by specialized capillary endothelial cells, which together with pericytes and perivascular glial cells regulates the exchanges between the central nervous system (CNS) and the periphery. Intricate interactions between the different cellular constituents of the BBB are crucial in establishing a functional BBB and maintaining the delicate homeostasis of the CNS microenvironment. In this review, we discuss the role of astrocytes and microglia in inducing and maintaining barrier properties under physiological conditions as well as their involvement during neuroinflammatory pathologies. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
19
|
Wong JK, Chen L, Huang Y, Sehba FA, Friedel RH, Zou H. Attenuation of Cerebral Ischemic Injury in Smad1 Deficient Mice. PLoS One 2015; 10:e0136967. [PMID: 26317208 PMCID: PMC4552810 DOI: 10.1371/journal.pone.0136967] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 08/11/2015] [Indexed: 12/03/2022] Open
Abstract
Stroke results in brain tissue damage from ischemia and oxidative stress. Molecular regulators of the protective versus deleterious cellular responses after cerebral ischemia remain to be identified. Here, we show that deletion of Smad1, a conserved transcription factor that mediates canonical bone morphogenetic protein (BMP) signaling, results in neuroprotection in an ischemia-reperfusion (I/R) stroke model. Uninjured mice with conditional deletion of Smad1 in the CNS (Smad1 cKO) displayed upregulation of the reactive astrocyte marker GFAP and hypertrophic morphological changes in astrocytes compared to littermate controls. Additionally, cultured Smad1-/- astrocytes exhibited an enhanced antioxidant capacity. When subjected to I/R injury by transient middle cerebral artery occlusion (tMCAO), Smad1 cKO mice showed enhanced neuronal survival and improved neurological recovery at 7 days post-stroke. This neuroprotective phenotype is associated with attenuated reactive astrocytosis and neuroinflammation, along with reductions in oxidative stress, p53 induction, and apoptosis. Our data suggest that Smad1-mediated signaling pathway is involved in stroke pathophysiology and may present a new potential target for stroke therapy.
Collapse
Affiliation(s)
- Jamie K Wong
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Lei Chen
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Yong Huang
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Fatima A Sehba
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Roland H Friedel
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America; Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| | - Hongyan Zou
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America; Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States of America
| |
Collapse
|
20
|
Yang Z, Wang KKW. Glial fibrillary acidic protein: from intermediate filament assembly and gliosis to neurobiomarker. Trends Neurosci 2015; 38:364-74. [PMID: 25975510 PMCID: PMC4559283 DOI: 10.1016/j.tins.2015.04.003] [Citation(s) in RCA: 643] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 04/03/2015] [Accepted: 04/07/2015] [Indexed: 12/20/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is an intermediate filament (IF) III protein uniquely found in astrocytes in the central nervous system (CNS), non-myelinating Schwann cells in the peripheral nervous system (PNS), and enteric glial cells. GFAP mRNA expression is regulated by several nuclear-receptor hormones, growth factors, and lipopolysaccharides (LPSs). GFAP is also subject to numerous post-translational modifications (PTMs), while GFAP mutations result in protein deposits known as Rosenthal fibers in Alexander disease. GFAP gene activation and protein induction appear to play a critical role in astroglial cell activation (astrogliosis) following CNS injuries and neurodegeneration. Emerging evidence also suggests that, following traumatic brain and spinal cord injuries and stroke, GFAP and its breakdown products are rapidly released into biofluids, making them strong candidate biomarkers for such neurological disorders.
Collapse
Affiliation(s)
- Zhihui Yang
- Program for Neurotrauma, Neuroproteomics, and Biomarkers Research, Departments of Psychiatry and Neuroscience, McKnight Brain Institute, L4-100, University of Florida, 1149 South Newell Drive, Gainesville, FL 32611, USA
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics, and Biomarkers Research, Departments of Psychiatry and Neuroscience, McKnight Brain Institute, L4-100, University of Florida, 1149 South Newell Drive, Gainesville, FL 32611, USA.
| |
Collapse
|
21
|
Naseer MI, Faheem M, Chaudhary AG, Kumosani TA, Al-Quaiti MM, Jan MM, Saleh Jamal H, Al-Qahtani MH. Genome wide analysis of novel copy number variations duplications/deletions of different epileptic patients in Saudi Arabia. BMC Genomics 2015; 16 Suppl 1:S10. [PMID: 25923336 PMCID: PMC4315149 DOI: 10.1186/1471-2164-16-s1-s10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Epilepsy is genetically complex neurological disorder affecting millions of people of different age groups varying in its type and severity. Copy number variants (CNVs) are key players in the genetic etiology of numerous neurodevelopmental disorders and prior findings also revealed that chromosomal aberrations are more susceptible against the pathogenesis of epilepsy. Novel technologies, such as array comparative genomic hybridization (array-CGH), may help to uncover the pathogenic CNVs in patients with epilepsy. Results This study was carried out by high density whole genome array-CGH analysis with blood DNA samples from a cohort of 22 epilepsy patients to search for CNVs associated with epilepsy. Pathogenic rearrangements which include 6p12.1 microduplications in 5 patients covering a total region of 99.9kb and 7q32.3 microdeletions in 3 patients covering a total region of 63.9kb were detected. Two genes BMP5 and PODXL were located in the predicted duplicated and deleted regions respectively. Furthermore, these CNV findings were confirmed by qPCR. Conclusion We have described, for the first time, several novel CNVs/genes implicated in epilepsy in the Saudi population. These findings enable us to better describe the genetic variations in epilepsy, and could provide a foundation for understanding the critical regions of the genome which might be involved in the development of epilepsy.
Collapse
|
22
|
Nevo I, Woolard K, Cam M, Li A, Webster JD, Kotliarov Y, Kim HS, Ahn S, Walling J, Kotliarova S, Belova G, Song H, Bailey R, Zhang W, Fine HA. Identification of molecular pathways facilitating glioma cell invasion in situ. PLoS One 2014; 9:e111783. [PMID: 25365423 PMCID: PMC4218815 DOI: 10.1371/journal.pone.0111783] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/30/2014] [Indexed: 12/22/2022] Open
Abstract
Gliomas are mostly incurable secondary to their diffuse infiltrative nature. Thus, specific therapeutic targeting of invasive glioma cells is an attractive concept. As cells exit the tumor mass and infiltrate brain parenchyma, they closely interact with a changing micro-environmental landscape that sustains tumor cell invasion. In this study, we used a unique microarray profiling approach on a human glioma stem cell (GSC) xenograft model to explore gene expression changes in situ in Invading Glioma Cells (IGCs) compared to tumor core, as well as changes in host cells residing within the infiltrated microenvironment relative to the unaffected cortex. IGCs were found to have reduced expression of genes within the extracellular matrix compartment, and genes involved in cell adhesion, cell polarity and epithelial to mesenchymal transition (EMT) processes. The infiltrated microenvironment showed activation of wound repair and tissue remodeling networks. We confirmed by protein analysis the downregulation of EMT and polarity related genes such as CD44 and PARD3 in IGCs, and EFNB3, a tissue-remodeling agent enriched at the infiltrated microenvironment. OLIG2, a proliferation regulator and glioma progenitor cell marker upregulated in IGCs was found to function in enhancing migration and stemness of GSCs. Overall, our results unveiled a more comprehensive picture of the complex and dynamic cell autonomous and tumor-host interactive pathways of glioma invasion than has been previously demonstrated. This suggests targeting of multiple pathways at the junction of invading tumor and microenvironment as a viable option for glioma therapy.
Collapse
Affiliation(s)
- Ido Nevo
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kevin Woolard
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maggie Cam
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Aiguo Li
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joshua D. Webster
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yuri Kotliarov
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hong Sug Kim
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Susie Ahn
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jennifer Walling
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Svetlana Kotliarova
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Galina Belova
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hua Song
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rolanda Bailey
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wei Zhang
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Howard A. Fine
- Neuro-Oncology Branch, National Cancer Institute, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
23
|
Baek SE, Kim JY, Song WT, Lee SH, Hong JH, Lee CK, Kang SG. Neuroprotective effect of rice bran extract supplemented with ferulic acid in the rat model of ischemic brain injury. Anim Cells Syst (Seoul) 2014. [DOI: 10.1080/19768354.2014.904249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
24
|
Varendi K, Airavaara M, Anttila J, Vose S, Planken A, Saarma M, Mitchell JR, Andressoo JO. Short-term preoperative dietary restriction is neuroprotective in a rat focal stroke model. PLoS One 2014; 9:e93911. [PMID: 24705386 PMCID: PMC3976327 DOI: 10.1371/journal.pone.0093911] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/07/2014] [Indexed: 01/13/2023] Open
Abstract
Stroke is a major complication of cardiovascular surgery, resulting in over 100,000 deaths and over a million postoperative encephalopathies annually in the US and Europe. While mitigating damage from stroke after it occurs has proven elusive, opportunities to reduce the incidence and/or severity of stroke prior to surgery in at-risk individuals remain largely unexplored. We tested the potential of short-term preoperative dietary restriction to provide neuroprotection in rat models of focal stroke. Rats were preconditioned with either three days of water-only fasting or six days of a protein free diet prior to induction of transient middle cerebral artery occlusion using two different methods, resulting in either a severe focal stroke to forebrain and midbrain, or a mild focal stroke localized to cortex only. Infarct volume, functional recovery and molecular markers of damage and protection were assessed up to two weeks after reperfusion. Preoperative fasting for 3 days reduced infarct volume after severe focal stroke. Neuroprotection was associated with modulation of innate immunity, including elevation of circulating neutrophil chemoattractant C-X-C motif ligand 1 prior to ischemia and suppression of striatal pro-inflammatory markers including tumor necrosis factor α, its receptor and downstream effector intercellular adhesion molecule-1 after reperfusion. Similarly, preoperative dietary protein restriction for 6 days reduced ischemic injury and improved functional recovery in a milder cortical infarction model. Our results suggest that short-term dietary restriction regimens may provide simple and translatable approaches to reduce perioperative stroke severity in high-risk elective vascular surgery.
Collapse
Affiliation(s)
- Kärt Varendi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jenni Anttila
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Sarah Vose
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Anu Planken
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
- * E-mail: (JRM); (JOA)
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- * E-mail: (JRM); (JOA)
| |
Collapse
|
25
|
Bone morphogenetic protein-7 ameliorates cerebral ischemia and reperfusion injury via inhibiting oxidative stress and neuronal apoptosis. Int J Mol Sci 2013; 14:23441-53. [PMID: 24287916 PMCID: PMC3876055 DOI: 10.3390/ijms141223441] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 11/12/2013] [Accepted: 11/19/2013] [Indexed: 01/21/2023] Open
Abstract
Previous studies have indicated that bone morphogenetic protein-7 (BMP-7) is neuroprotective against cerebral ischemia/reperfusion (IR) injury. The present study was undertaken to determine the molecular mechanisms involved in this effect. Adult male Wistar rats were subjected to 2 h of transient middle cerebral artery occlusion (MCAO), followed by 24 h of reperfusion. BMP-7 (10−4 g/kg) or vehicle was infused into rats at the onset of reperfusion via the tail vein. Neurological deficits, infarct volume, histopathological changes, oxidative stress-related biochemical parameters, neuronal apoptosis, and apoptosis-related proteins were assessed. BMP-7 significantly improved neurological and histological deficits, reduced the infarct volume, and decreased apoptotic cells after cerebral ischemia. BMP-7 also markedly enhanced the activities of antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX), and reduced the level of malondialdehyde (MDA) in IR rats. In addition, Western blot analysis indicated that BMP-7 prevented cytochrome c release, inhibited activation of caspase-3, caspase-9 and caspase-8. Our data suggested that BMP-7 has protective effects against cerebral IR injury in rats, and the neuroprotective effects may be attributed to attenuating oxidative stress and inhibiting neuronal apoptosis.
Collapse
|
26
|
Bone morphogenetic protein-7 (BMP-7) mediates ischemic preconditioning-induced ischemic tolerance via attenuating apoptosis in rat brain. Biochem Biophys Res Commun 2013; 441:560-6. [DOI: 10.1016/j.bbrc.2013.10.121] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 10/22/2013] [Indexed: 12/14/2022]
|
27
|
Xu JH, Zhang TZ, Zhao YY, Wang JK, Yuan ZG. Protective effects of recombinant human bone morphogenetic protein-7 on focal cerebral ischemia-reperfusion injury. Int J Neurosci 2013; 123:375-84. [PMID: 23327662 DOI: 10.3109/00207454.2012.761614] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study was to investigate the protective effect of recombinant human bone morphogenetic protein-7 (rhBMP-7) on focal cerebral ischemia-reperfusion (IR) injuries and their underlying mechanisms. An intraluminal suture method was used to generate a middle cerebral artery occlusion model in rats, which was followed by reperfusion. A sham operation (SO) group underwent the procedure without occlusion, whereas an IR group and rhBMP-7 treated group (RT) underwent occlusion in the absence and presence of rhBMP-7 (250 μg/kg) administered via a femoral vein injection 30 minutes prior to reperfusion. Twenty-four hours after reperfusion, neurological function, brain water content and morphological alterations were examined. Apoptosis was detected using terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assays, and immunohistochemical staining and Western blot assays were used to detect nuclear nuclear factor-kappa B (NF-κB) p65 expression. Compared with the SO group, IR rats showed a decrease in neurological function, an increase in brain water content, and pathological and morphological damage (p < 0.05). Higher levels of apoptosis were also detected in the infarct region area. In contrast, RT rats had reduced injury after IR. In addition, while immunohistochemical staining and western blot assays consistently detected increased expression of nuclear NF-κB after IR, these levels were reduced in the RT group. Administration of rhBMP-7 prior to reperfusion effectively inhibited the extent of IR injury by attenuating cerebral edema and ameliorating ultrastructural damage. The underlying mechanisms responsible for these observations potentially involve the inhibition of apoptosis induced by IR by rhBMP-7 via an NF-κB-related signaling cascade.
Collapse
Affiliation(s)
- Ji-Hong Xu
- Department of Anesthesiology, General Hospital of Shenyang Military Region, Shenyang, China
| | | | | | | | | |
Collapse
|
28
|
Park YM, Lee WT, Bokara KK, Seo SK, Park SH, Kim JH, Yenari MA, Park KA, Lee JE. The multifaceted effects of agmatine on functional recovery after spinal cord injury through Modulations of BMP-2/4/7 expressions in neurons and glial cells. PLoS One 2013; 8:e53911. [PMID: 23349763 PMCID: PMC3549976 DOI: 10.1371/journal.pone.0053911] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 12/04/2012] [Indexed: 11/29/2022] Open
Abstract
Presently, few treatments for spinal cord injury (SCI) are available and none have facilitated neural regeneration and/or significant functional improvement. Agmatine (Agm), a guanidinium compound formed from decarboxylation of L-arginine by arginine decarboxylase, is a neurotransmitter/neuromodulator and been reported to exert neuroprotective effects in central nervous system injury models including SCI. The purpose of this study was to demonstrate the multifaceted effects of Agm on functional recovery and remyelinating events following SCI. Compression SCI in mice was produced by placing a 15 g/mm(2) weight for 1 min at thoracic vertebra (Th) 9 segment. Mice that received an intraperitoneal (i.p.) injection of Agm (100 mg/kg/day) within 1 hour after SCI until 35 days showed improvement in locomotor recovery and bladder function. Emphasis was made on the analysis of remyelination events, neuronal cell preservation and ablation of glial scar area following SCI. Agm treatment significantly inhibited the demyelination events, neuronal loss and glial scar around the lesion site. In light of recent findings that expressions of bone morphogenetic proteins (BMPs) are modulated in the neuronal and glial cell population after SCI, we hypothesized whether Agm could modulate BMP- 2/4/7 expressions in neurons, astrocytes, oligodendrocytes and play key role in promoting the neuronal and glial cell survival in the injured spinal cord. The results from computer assisted stereological toolbox analysis (CAST) demonstrate that Agm treatment dramatically increased BMP- 2/7 expressions in neurons and oligodendrocytes. On the other hand, BMP- 4 expressions were significantly decreased in astrocytes and oligodendrocytes around the lesion site. Together, our results reveal that Agm treatment improved neurological and histological outcomes, induced oligodendrogenesis, protected neurons, and decreased glial scar formation through modulating the BMP- 2/4/7 expressions following SCI.
Collapse
Affiliation(s)
- Yu Mi Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- BK 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Won Taek Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kiran Kumar Bokara
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su Kyoung Seo
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- BK 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Hwa Park
- Department of Anatomy, Konkuk University College of Medicine, Seoul, Republic of Korea
| | - Jae Hwan Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Midori A. Yenari
- Department of Neurology, University of California San Francisco and Veterans Affairs Medical Center, San Francisco, California, United States of America
| | - Kyung Ah Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
- BK 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
29
|
Promoting engraftment of transplanted neural stem cells/progenitors using biofunctionalised electrospun scaffolds. Biomaterials 2012; 33:9188-97. [PMID: 23022345 DOI: 10.1016/j.biomaterials.2012.09.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 09/09/2012] [Indexed: 02/02/2023]
Abstract
With the brain's limited capacity for repair, new and innovative approaches are required to promote regeneration. While neural transplantation for a number of neural disease/injuries have been demonstrated, major limitations in the field include poor cell survival and integration. This, in part, is due to the non-conducive environment of the adult brain, failing to provide adequate chemical and physical support for new neurons. Here we examine the capacity of fibrous poly ε-caprolactone (PCL) scaffolds, biofunctionalised with immobilised glial cell-derived neurotrophic factor (GDNF), to influence primary cortical neural stem cells/progenitors in vitro and enhance integration of these cells following transplantation into the brain parenchyma. Immobilisation of GDNF was confirmed prior to in vitro culturing and at 28 days after implantation into the brain, demonstrating long-term delivery of the protein. In vitro, we demonstrate that PCL with immobilised GDNF (iGDNF) significantly enhances cell viability and neural stem cell/progenitor proliferation compared to conventional 2-dimensional cultureware. Upon implantation, PCL scaffolds including iGDNF enhanced the survival, proliferation, migration, and neurite growth of transplanted cortical cells, whilst suppressing inflammatory reactive astroglia.
Collapse
|
30
|
Faigle R, Song H. Signaling mechanisms regulating adult neural stem cells and neurogenesis. Biochim Biophys Acta Gen Subj 2012; 1830:2435-48. [PMID: 22982587 DOI: 10.1016/j.bbagen.2012.09.002] [Citation(s) in RCA: 251] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2012] [Revised: 08/11/2012] [Accepted: 09/05/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Adult neurogenesis occurs throughout life in discrete regions of the mammalian brain and is tightly regulated via both extrinsic environmental influences and intrinsic genetic factors. In recent years, several crucial signaling pathways have been identified in regulating self-renewal, proliferation, and differentiation of neural stem cells, as well as migration and functional integration of developing neurons in the adult brain. SCOPE OF REVIEW Here we review our current understanding of signaling mechanisms, including Wnt, notch, sonic hedgehog, growth and neurotrophic factors, bone morphogenetic proteins, neurotransmitters, transcription factors, and epigenetic modulators, and crosstalk between these signaling pathways in the regulation of adult neurogenesis. We also highlight emerging principles in the vastly growing field of adult neural stem cell biology and neural plasticity. MAJOR CONCLUSIONS Recent methodological advances have enabled the field to identify signaling mechanisms that fine-tune and coordinate neurogenesis in the adult brain, leading to a better characterization of both cell-intrinsic and environmental cues defining the neurogenic niche. Significant questions related to niche cell identity and underlying regulatory mechanisms remain to be fully addressed and will be the focus of future studies. GENERAL SIGNIFICANCE A full understanding of the role and function of individual signaling pathways in regulating neural stem cells and generation and integration of newborn neurons in the adult brain may lead to targeted new therapies for neurological diseases in humans. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Roland Faigle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | |
Collapse
|
31
|
Low gene expression of bone morphogenetic protein 7 in brainstem astrocytes in major depression. Int J Neuropsychopharmacol 2012; 15:855-68. [PMID: 21896235 DOI: 10.1017/s1461145711001350] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The noradrenergic locus coeruleus (LC) is the principal source of brain norepinephrine, a neurotransmitter thought to play a major role in the pathology of major depressive disorder (MDD) and in the therapeutic action of many antidepressant drugs. The goal of this study was to identify potential mediators of brain noradrenergic dysfunction in MDD. Bone morphogenetic protein 7 (BMP7), a member of the transforming growth factor-β superfamily, is a critical mediator of noradrenergic neuron differentiation during development and has neurotrophic and neuroprotective effects on mature catecholaminergic neurons. Real-time PCR of reversed transcribed RNA isolated from homogenates of LC tissue from 12 matched pairs of MDD subjects and psychiatrically normal control subjects revealed low levels of BMP7 gene expression in MDD. No differences in gene expression levels of other members of the BMP family were observed in the LC, and BMP7 gene expression was normal in the prefrontal cortex and amygdala in MDD subjects. Laser capture microdissection of noradrenergic neurons, astrocytes, and oligodendrocytes from the LC revealed that BMP7 gene expression was highest in LC astrocytes relative to the other cell types, and that the MDD-associated reduction in BMP7 gene expression was limited to astrocytes. Rats exposed to chronic social defeat exhibited a similar reduction in BMP7 gene expression in the LC. BMP7 has unique developmental and trophic actions on catecholamine neurons and these findings suggest that reduced astrocyte support for pontine LC neurons may contribute to pathology of brain noradrenergic neurons in MDD.
Collapse
|
32
|
Shin JA, Kang JL, Lee KE, Park EM. Different temporal patterns in the expressions of bone morphogenetic proteins and noggin during astroglial scar formation after ischemic stroke. Cell Mol Neurobiol 2012; 32:587-97. [PMID: 22297545 PMCID: PMC11498437 DOI: 10.1007/s10571-012-9806-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 01/17/2012] [Indexed: 01/15/2023]
Abstract
Bone morphogenetic proteins (BMPs) and their antagonists have roles in scar formation and regeneration after central nervous system injuries. However, temporal changes in their expression during astroglial scar formation in the ischemic brain are unknown. Here, we examined protein levels of BMP2, BMP7, and their antagonist noggin in the ischemic brain up to 4 weeks after experimental stroke in mice. BMP2 and BMP7 levels were increased from 1 to 4 weeks in the ischemic brain, and their expression was associated with astrogliosis. BMP7 expression was more intense and co-localized in reactive astrocytes in the ischemic subcortex at 1 week. Noggin expression began to increase after 2 weeks and was further increased at 4 weeks only in the ischemic subcortex, but the intensity was weak compared to the intensity of BMPs. Noggin was co-localized mainly in activated microglia. These findings show that expression of BMPs and noggin differed over time, in intensity and in types of cell, and suggest that BMPs and noggin have different roles in the processes of glial scar formation and neurorestoration in the ischemic brain.
Collapse
Affiliation(s)
- Jin A. Shin
- Department of Pharmacology, Ewha Medical Research Institute, School of Medicine, Ewha Womans University, 911-1 Mok6dong Yangcheon-gu, Seoul, 158-710 Republic of Korea
| | - Jihee Lee Kang
- Department of Physiology, Tissue Injury Defense Research Center, Ewha Medical Research Institute, School of Medicine, Ewha Womans University, 911-1 Mok6dong Yangcheon-gu, Seoul, 158-710 Republic of Korea
| | - Kyung-Eun Lee
- Department of Pharmacology, Ewha Medical Research Institute, School of Medicine, Ewha Womans University, 911-1 Mok6dong Yangcheon-gu, Seoul, 158-710 Republic of Korea
| | - Eun-Mi Park
- Department of Pharmacology, Ewha Medical Research Institute, School of Medicine, Ewha Womans University, 911-1 Mok6dong Yangcheon-gu, Seoul, 158-710 Republic of Korea
- Department of Brain & Cognitive Sciences, Brain Disease Research Institute, Ewha Womans University, 52, Ewhayeodae-gil, Seodaemun-gu, Seoul, 120-750 Republic of Korea
| |
Collapse
|
33
|
Zhang L, Qu Y, Tang B, Zhao F, Xiong T, Ferriero D, Mu D. Integrin β8 Signaling in Neonatal Hypoxic–Ischemic Brain Injury. Neurotox Res 2012; 22:280-91. [DOI: 10.1007/s12640-012-9312-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 01/10/2012] [Accepted: 01/16/2012] [Indexed: 11/24/2022]
|
34
|
Kim JH, Lee YW, Park YM, Park KA, Park SH, Lee WT, Lee JE. Agmatine-reduced collagen scar area accompanied with surface righting reflex recovery after complete transection spinal cord injury. Spine (Phila Pa 1976) 2011; 36:2130-2138. [PMID: 21325984 DOI: 10.1097/brs.0b013e318205e3f7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Intended to investigate whether agmatine treatment reduces collagen scar area in mice subjected to spinal cord injury (SCI). OBJECTIVE The purpose of the present study is to demonstrate the protective effect of agmatine on complete transection SCI mice. SUMMARY OF BACK GROUND DATA: The deposition of collagen that occurs at the lesion site, during the SCI, was well known. Agmatine has been reported to exert neuroprotective effect in various stress models including central nervous system injuries. In the present investigation, we hypothesized that agmatine treatment could rescue the mice subjected to SCI. METHODS Complete SCI was made at the T9 level. Agmatine was dissolved in normal saline (100 mg/kg, Sigma, St. Louis, MO) and given intraperitoneally 5 minutes after complete transection daily for 4 weeks (agmatine-treated mice, n = 30). Controls received normal saline in the same manner (experimental control, n = 30). Surface righting reflex test, expression of bone morphogenetic protein-7 (BMP-7), TGFβ-2 (transforming growth factor β-2), and collagen scar area were measured and the results were compared with Mann-Whitney U test using SAS. RESULTS Agmatine treatment improved the surface righting reflex of mice at 4 weeks after SCI (P = 0.030). The collagen scar, physical barrier to axon regeneration, was noticeably diminished by agmatine treatment at 4 weeks after SCI (P = 0.001). The expression of BMP-7, which is considered both neuroprotective and neuroregenerative, was increased in agmatine treatment group compared with experimental control group in the early stages after SCI (P = 0.015 at 1 day after SCI; P = 0.010 at 3 days; P = 0.035 at 1 week; P = 0.826 at 2 weeks). The expression of TGFβ-2 correlated with the deposition of the collagen matrix at the lesion site was decreased with agmatine treatment at 1 and 2 weeks after SCI (P = 0.001 at 1 week; P = 0.002 at 2 weeks). Survival rate was found to be higher in agmatine treatment group than in the experimental control group for 4 weeks after SCI (P = 0.076). CONCLUSION These results suggest that agmatine treatment could support neuroregeneration by reducing the collagen scar area through decreasing the expression of TGFβ-2 and increasing the expression of BMP-7 after SCI. Especially, the improved surface righting reflex of agmatine-treated group proposes that agmatine treatment have the potency to facilitate functional recovery after SCI. However, the drug (agmatine) warrants further investigation in higher mammals.
Collapse
Affiliation(s)
- Jae Hwan Kim
- Department of Anatomy, Yonsei University College of Medicine, Seodaemun-gu, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
Khundakar A, Morris C, Slade J, Thomas AJ. Examination of glucose transporter-1, transforming growth factor-β and neuroglobin immunoreactivity in the orbitofrontal cortex in late-life depression. Psychiatry Clin Neurosci 2011; 65:158-64. [PMID: 21232076 DOI: 10.1111/j.1440-1819.2010.02176.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIMS This study immunohistochemically examined the orbitofrontal cortex for three possible candidates in hypoxic/ischemic signaling: the cytokine transforming growth factor-β, the glucose transporter-1 and the neuron-specific oxygen-binding protein neuroglobin. METHODS Post-mortem tissue from 20 depressed and 20 non-depressed individuals was obtained and the expression of the three proteins was analyzed using image analysis software. RESULTS No significant changes were found in transforming growth factor-β or neuroglobin in the orbitofrontal cortex between depressed and non-depressed individuals. There was, however, a trend towards a reduction in glucose transporter-1 in the depressed group. CONCLUSIONS This study does not clearly support the hypothesis that hypoxic/ischemic processes are behind the pathological deficits in the frontal-subcortical circuitry associated with depression and therefore does not provide evidence to support the 'vascular depression' hypothesis.
Collapse
Affiliation(s)
- Ahmad Khundakar
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK.
| | | | | | | |
Collapse
|
36
|
Kam KY, Yu SJ, Jeong N, Hong JH, Anthony Jalin AMA, Lee S, Choi YW, Lee CK, Kang SG. p-Hydroxybenzyl alcohol prevents brain injury and behavioral impairment by activating Nrf2, PDI, and neurotrophic factor genes in a rat model of brain ischemia. Mol Cells 2011; 31:209-15. [PMID: 21347705 PMCID: PMC3932695 DOI: 10.1007/s10059-011-0028-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 11/03/2010] [Accepted: 12/08/2010] [Indexed: 01/21/2023] Open
Abstract
The therapeutic goal in treating cerebral ischemia is to reduce the extent of brain injury and thus minimize neurological impairment. We examined the effects of p-hydroxybenzyl alcohol (HBA), an active component of Gastrodia elata Blume, on transient focal cerebral ischemia-induced brain injury with respect to the involvement of protein disulphide isomerase (PDI), nuclear factor-E2-related factor 2 (Nrf2), and neurotrophic factors. All animals were ovariectomized 14 days before ischemic injury. Ischemic injury was induced for 1 h by middle cerebral artery occlusion (MCAO) followed by 24-h reperfusion. Three days before MCAO, the vehicle-treated and the HBA-treated groups received intramuscular sesame oil and HBA (25 mg/kg BW), respectively. 2,3,5-Triphenyltetrazolium chloride (TTC) staining showed decreased infarct volume in the ischemic lesion of HBA-treated animals. HBA pretreatment also promoted functional recovery, as measured by the modified neurological severity score (mNSS; p < 0.05). Moreover, expression of PDI, Nrf2, BDNF, GDNF, and MBP genes increased by HBA treatment. In vitro, H(2)O(2)-induced PC12 cell death was prevented by 24 h HBA treatment, but bacitracin, a PDI inhibitor, attenuated this cytoprotective effect in a dose-dependent manner. HBA treatment for 2 h also induced nuclear translocation of Nrf2, possibly activating the intracellular antioxidative system. These results suggest that HBA protects against brain damage by modulating cytoprotective genes, such as Nrf2 and PDI, and neurotrophic factors.
Collapse
Affiliation(s)
- Kyung-Yoon Kam
- Department of Occupational Therapy, Inje University, Gimhae 621-749, Korea
- FIRST Research Group, Inje University, Gimhae 621-749, Korea
- Institute of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Seong Jin Yu
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Nahee Jeong
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Jeong Hwa Hong
- School of Food and Life Science , Inje University, Gimhae 621-749, Koreaof Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Angela M. A. Anthony Jalin
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| | - Sungja Lee
- Department of Occupational Therapy, Inje University, Gimhae 621-749, Korea
| | - Yong Won Choi
- Department of Occupational Therapy, Inje University, Gimhae 621-749, Korea
| | - Chae Kwan Lee
- Institute of Environmental and Occupational Medicine, Department of Occu-pational and Environmental Medicine, Busan Paik Hospital, Inje University, Busan 614-735, Korea
| | - Sung Goo Kang
- FIRST Research Group, Inje University, Gimhae 621-749, Korea
- School of Biological Sciences, Inje University, Gimhae 621-749, Koreate of Aged Life Redesign, Inje University, Gimhae 621-749, Korea
| |
Collapse
|
37
|
Rosell A, Vilalta A, García-Berrocoso T, Fernández-Cadenas I, Domingues-Montanari S, Cuadrado E, Delgado P, Ribó M, Martínez-Sáez E, Ortega-Aznar A, Montaner J. Brain perihematoma genomic profile following spontaneous human intracerebral hemorrhage. PLoS One 2011; 6:e16750. [PMID: 21311749 PMCID: PMC3032742 DOI: 10.1371/journal.pone.0016750] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 01/10/2011] [Indexed: 01/09/2023] Open
Abstract
Background Spontaneous intracerebral hemorrhage (ICH) represents about 15% of all strokes and is associated with high mortality rates. Our aim was to identify the gene expression changes and biological pathways altered in the brain following ICH. Methodology/Principal Findings Twelve brain samples were obtained from four deceased patients who suffered an ICH including perihematomal tissue (PH) and the corresponding contralateral white (CW) and grey (CG) matter. Affymetrix GeneChip platform for analysis of over 47,000 transcripts was conducted. Microarray Analysis Suite 5.0 was used to process array images and the Ingenuity Pathway Analysis System was used to analyze biological mechanisms and functions of the genes. We identified 468 genes in the PH areas displaying a different expression pattern with a fold change between −3.74 and +5.16 when compared to the contralateral areas (291 overexpressed and 177 underexpressed). The top genes which appeared most significantly overexpressed in the PH areas codify for cytokines, chemokines, coagulation factors, cell growth and proliferation factors while the underexpressed codify for proteins involved in cell cycle or neurotrophins. Validation and replication studies at gene and protein level in brain samples confirmed microarray results. Conclusions The genomic responses identified in this study provide valuable information about potential biomarkers and target molecules altered in the perihematomal regions.
Collapse
Affiliation(s)
- Anna Rosell
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | - Anna Vilalta
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | - Teresa García-Berrocoso
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | - Israel Fernández-Cadenas
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | - Sophie Domingues-Montanari
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | - Eloy Cuadrado
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | - Pilar Delgado
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | - Marc Ribó
- Stroke Unit and Department of Neurology, Universitat Autònoma de Barcelona, Hospital Vall d'Hebron, Barcelona, Spain
| | - Elena Martínez-Sáez
- Neuropathology Unit, Department of Pathology, Universitat Autònoma de Barcelona, Hospital Vall d'Hebron, Barcelona, Spain
| | - Arantxa Ortega-Aznar
- Neuropathology Unit, Department of Pathology, Universitat Autònoma de Barcelona, Hospital Vall d'Hebron, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory and Department of Neurology, Universitat Autònoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
- Stroke Unit and Department of Neurology, Universitat Autònoma de Barcelona, Hospital Vall d'Hebron, Barcelona, Spain
- * E-mail:
| |
Collapse
|
38
|
Schindowski K, von Bohlen und Halbach O, Strelau J, Ridder DA, Herrmann O, Schober A, Schwaninger M, Unsicker K. Regulation of GDF-15, a distant TGF-β superfamily member, in a mouse model of cerebral ischemia. Cell Tissue Res 2010; 343:399-409. [PMID: 21128084 PMCID: PMC3032194 DOI: 10.1007/s00441-010-1090-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/04/2010] [Indexed: 11/20/2022]
Abstract
GDF-15 is a novel distant member of the TGF-β superfamily and is widely distributed in the brain and peripheral nervous system. We have previously reported that GDF-15 is a potent neurotrophic factor for lesioned dopaminergic neurons in the substantia nigra, and that GDF-15-deficient mice show progressive postnatal losses of motor and sensory neurons. We have now investigated the regulation of GDF-15 mRNA and immunoreactivity in the murine hippocampal formation and selected cortical areas following an ischemic lesion by occlusion of the middle cerebral artery (MCAO). MCAO prominently upregulates GDF-15 mRNA in the hippocampus and parietal cortex at 3 h and 24 h after lesion. GDF-15 immunoreactivity, which is hardly detectable in the unlesioned brain, is drastically upregulated in neurons identified by double-staining with NeuN. NeuN staining reveals that most, if not all, neurons in the granular layer of the dentate gyrus and pyramidal layers of the cornu ammonis become GDF-15-immunoreactive. Moderate induction of GDF-15 immunoreactivity has been observed in a small number of microglial cells identified by labeling with tomato lectin, whereas astroglial cells remain GDF-15-negative after MCAO. Comparative analysis of the size of the infarcted area after MCAO in GDF-15 wild-type and knockout mice has failed to reveal significant differences. Together, our data substantiate the notion that GDF-15 is prominently upregulated in the lesioned brain and might be involved in orchestrating post-lesional responses other than the trophic support of neurons.
Collapse
Affiliation(s)
- Katharina Schindowski
- Institute for Pharmaceutical Biotechnology, University of Applied Science Biberach, Biberach/Riss, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Effects of mouse recombinant bone morphogenetic protein-7 transfection on cell apoptosis, NF-kappaB, and downstream genes in cultured primary cardiomyocytes after simulated ischemia and reperfusion injury. J Cardiovasc Pharmacol 2010; 56:69-77. [PMID: 20631551 DOI: 10.1097/fjc.0b013e3181e0badc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIM To evaluate the acting mechanism of bone morphogenetic protein-7 in anti-ischemic protective effect, we investigate the effects of BMP-7 transfection on cell apoptosis, NF-kappaB activity, and downstream genes in neonatal rat cardiomyocytes during simulated ischemia-reperfusion. METHOD In vitro cultured neonatal rat cardiomyocytes were divided into four groups: normal control group (Group C), simulated ischemia-reperfusion group (Group IR: cultured cardiomyocytes were subjected to 2 hours hypoxia followed by 4 hours reoxygenation), transfected group (Group BT: after transfection with pcDNA3.1-BMP-7 plasmid, cardiomyocytes were subjected to 2 hours hypoxia/4 hours reoxygenation), and empty vector control group (Group ET: same as group BT except that cells were transfected with empty pcDNA3.1 vector). Malondialdehyde content, superoxide dismutase activity, and Ca(2+) concentration were measured. Terminal deoxynucleotide transferase-mediated dUTP nick-end labeling staining and fluorescence activated cell sorting assay were applied to determine the apoptotic rate of cardiomyocytes and immunocytochemistry and Western blot were used to detect nuclear expression of NF-kappaB in cardiomyocytes. Reverse transcription-polymerase chain reaction was used to detect mRNA expression of monocyte chemoattractant protein-1 and intercellular cell adhesion molecule-1. RESULTS Compared with Group IR, malondialdehyde content of Group BT significantly decreased, whereas superoxide dismutase activity significantly elevated. In addition, intracellular Ca(2+) concentration of Group BT significantly reduced and apoptotic cells were significantly less. Decreased optical density ratio of NF-kappaB in the nucleus and reduced monocyte chemoattractant protein-1 and intercellular cell adhesion molecule-1 mRNA expression were also detected. CONCLUSION These results suggest that rat recombinant BMP-7 transfection provides sustained support for the repair of myocardium from ischemia injury through reducing cell apoptosis, inhibiting lipid peroxidation, antagonizing intracellular Ca(2+) overload, and preventing the activation of downstream signaling factors in signal transduction pathway mediated by NF-kappaB.
Collapse
|
40
|
Increased BMP6 levels in the brains of Alzheimer's disease patients and APP transgenic mice are accompanied by impaired neurogenesis. J Neurosci 2010; 30:12252-62. [PMID: 20844121 DOI: 10.1523/jneurosci.1305-10.2010] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During aging and in the progression of Alzheimer's disease (AD), synaptic plasticity and neuronal integrity are disturbed. In addition to the alterations in plasticity in mature neurons, the neurodegenerative process in AD has been shown to be accompanied by alterations in neurogenesis. Members of the bone morphogenetic protein (BMP) family of growth factors have been implicated as important regulators of neurogenesis and neuronal cell fate determination during development; however, their role in adult neurogenesis and in AD is less clear. We show here by qRT-PCR analysis that BMP6 mRNA levels were significantly increased in the hippocampus of human patients with AD and in APP transgenic mice compared to controls. Immunoblot and immunohistochemical analyses confirmed that BMP6 protein levels were increased in human AD brains and APP transgenic mouse brains compared to controls and accumulated around hippocampal plaques. The increased levels of BMP6 were accompanied by defects in hippocampal neurogenesis in AD patients and APP transgenic mice. In support of a role for BMP6 in defective neurogenesis in AD, we show in an in vitro model of adult neurogenesis that treatment with amyloid-β(1-42) protein (Aβ) resulted in increased expression of BMP6, and that exposure to recombinant BMP6 resulted in reduced proliferation with no toxic effects. Together, these results suggest that Aβ-associated increases in BMP6 expression in AD may have deleterious effects on neurogenesis in the hippocampus, and therapeutic approaches could focus on normalization of BMP6 levels to protect against AD-related neurogenic deficits.
Collapse
|
41
|
Shen LH, Li Y, Chopp M. Astrocytic endogenous glial cell derived neurotrophic factor production is enhanced by bone marrow stromal cell transplantation in the ischemic boundary zone after stroke in adult rats. Glia 2010; 58:1074-81. [PMID: 20468049 DOI: 10.1002/glia.20988] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bone marrow stromal cells (BMSCs) facilitate functional recovery in rats after focal ischemic attack. Growing evidence suggests that the secretion of various bioactive factors underlies BMSCs' beneficial effects. This study investigates the expression of glial cell derived neurotrophic factor (GDNF) in the ischemic hemisphere with or without BMSC administration. Adult male Wistar rats were subjected to 2 h of middle cerebral artery occlusion followed by an injection of 3 x 10(6) BMSCs (n = 11) or phosphate-buffered saline (n = 10) into the tail vein 24 h later. Animals were sacrificed seven days later. Single and double immunohistochemical staining was performed to measure GDNF, Ki67, doublecortin, and glial fibrillary acidic protein expression as well as the number of apoptotic cells along the ischemic boundary zone (IBZ) and/or in the subventricular zone (SVZ). BMSC treatment significantly increased GDNF expression and decreased the number of apoptotic cells in the IBZ (P < 0.05). GDNF expression was colocalized with GFAP. Meanwhile, BMSCs increased the number of Ki-67 positive cells and the density of DCX positive migrating neuroblasts (P < 0.05). GDNF expression was significantly increased in single astrocytes collected from animals treated with BMSCs, and in astrocytes cocultured with BMSCs after OGD (P < 0.05). Our data suggest that BMSCs increase GDNF levels in the ischemic hemisphere; the major source of GDNF protein is reactive astrocytes. We propose that the increase of GDNF in response to BMSC administration creates a hospitable environment for local cellular repair as well as for migrating neuroblasts from the SVZ, and thus contributes to the functional improvement.
Collapse
Affiliation(s)
- L H Shen
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
42
|
Bone morphogenetic proteins mediate cellular response and, together with Noggin, regulate astrocyte differentiation after spinal cord injury. Exp Neurol 2010; 221:353-66. [DOI: 10.1016/j.expneurol.2009.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 10/31/2009] [Accepted: 12/01/2009] [Indexed: 11/20/2022]
|
43
|
Samanta J, Alden T, Gobeske K, Kan L, Kessler JA. Noggin protects against ischemic brain injury in rodents. Stroke 2009; 41:357-62. [PMID: 20019326 DOI: 10.1161/strokeaha.109.565523] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Bone morphogenetic proteins and their receptors are expressed in adult brains, and their expression levels increase after cerebral ischemia. The brain also expresses an inhibitor of bone morphogenetic protein signaling, noggin, but the role of noggin in ischemic disease outcome has not been studied. METHODS We used transgenic mice overexpressing noggin to assess whether inhibition of bone morphogenetic protein signaling affects ischemic injury responses after permanent middle cerebral artery occlusion. RESULTS Transgenic mice overexpressing noggin mice had significantly smaller infarct volumes and lower motor deficits compared to wild-type mice. CD11b(+) and IBA1(+) microglia along with oligodendroglial progenitors were significantly increased in transgenic mice overexpressing noggin mice at 14 days after permanent middle cerebral artery occlusion. CONCLUSIONS These results provide genetic evidence that overexpression of noggin reduces ischemic brain injury after permanent middle cerebral artery occlusion via enhanced activation of microglia and oligodendrogenesis.
Collapse
Affiliation(s)
- Jayshree Samanta
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Ill, USA
| | | | | | | | | |
Collapse
|
44
|
Sawada N, Liao JK. Targeting eNOS and beyond: emerging heterogeneity of the role of endothelial Rho proteins in stroke protection. Expert Rev Neurother 2009; 9:1171-86. [PMID: 19673606 DOI: 10.1586/ern.09.70] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Currently available modalities for the treatment of acute ischemic stroke are aimed at preserving or augmenting cerebral blood flow. Experimental evidence suggests that statins, which show 25-30% reduction of stroke incidence in clinical trials, confer stroke protection by upregulation of eNOS and increasing cerebral blood flow. The upregulation of eNOS by statins is mediated by inhibition of small GTP-binding protein RhoA. Our recent study uncovered a unique role for a Rho-family member Rac1 in stroke protection. Rac1 in endothelium does not affect cerebral blood flow. Instead, inhibition of endothelial Rac1 leads to broad upregulation of the genes relevant to neurovascular protection. Intriguingly, inhibition of endothelial Rac1 enhances neuronal cell survival through endothelium-derived neurotrophic factors, including artemin. This review discusses the emerging therapeutic opportunities to target neurovascular signaling beyond the BBB, with special emphasis on the novel role of endothelial Rac1 in stroke protection.
Collapse
Affiliation(s)
- Naoki Sawada
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Center for Life Sciences, Boston, MA 02115, USA.
| | | |
Collapse
|
45
|
Lim ST, Airavaara M, Harvey BK. Viral vectors for neurotrophic factor delivery: a gene therapy approach for neurodegenerative diseases of the CNS. Pharmacol Res 2009; 61:14-26. [PMID: 19840853 DOI: 10.1016/j.phrs.2009.10.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 10/11/2009] [Accepted: 10/11/2009] [Indexed: 01/11/2023]
Abstract
The clinical manifestation of most diseases of the central nervous system results from neuronal dysfunction or loss. Diseases such as stroke, epilepsy and neurodegeneration (e.g. Alzheimer's disease and Parkinson's disease) share common cellular and molecular mechanisms (e.g. oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction) that contribute to the loss of neuronal function. Neurotrophic factors (NTFs) are secreted proteins that regulate multiple aspects of neuronal development including neuronal maintenance, survival, axonal growth and synaptic plasticity. These properties of NTFs make them likely candidates for preventing neurodegeneration and promoting neuroregeneration. One approach to delivering NTFs to diseased cells is through viral vector-mediated gene delivery. Viral vectors are now routinely used as tools for studying gene function as well as developing gene-based therapies for a variety of diseases. Currently, many clinical trials using viral vectors in the nervous system are underway or completed, and seven of these trials involve NTFs for neurodegeneration. In this review, we discuss viral vector-mediated gene transfer of NTFs to treat neurodegenerative diseases of the central nervous system.
Collapse
Affiliation(s)
- Seung T Lim
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, United States
| | | | | |
Collapse
|
46
|
de Rivero Vaccari JP, Marcillo A, Nonner D, Dietrich WD, Keane RW. Neuroprotective effects of bone morphogenetic protein 7 (BMP7) treatment after spinal cord injury. Neurosci Lett 2009; 465:226-9. [PMID: 19765637 DOI: 10.1016/j.neulet.2009.09.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 08/31/2009] [Accepted: 09/11/2009] [Indexed: 11/16/2022]
Abstract
Bone morphogenetic protein 7 (BMP7) has been shown to ameliorate reduced dendritic growth induced by glutamate excitotoxicity in neuronal tissue cultures and/or provide an enhancement of functional recovery in central nervous system (CNS) injury. BMP7 expression is modulated by spinal cord injury (SCI), but the molecular mechanisms involved in neuroprotection have not been clearly defined. Here, we show that BMP7 treatment of rats subjected to mild cervical SCI significantly increased the pro-survival mitogen-activated protein kinase-38 (MAPK-38) pathway and levels of N-methyl-D-aspartate receptor 1 (NMDAR-1) resulting in a significant increase in neuronal sparing in the ventral horn of the spinal cord. Moreover, BMP7 was neuroprotective against glutamate-mediated excitotoxicity in cultured cortical neurons. These studies show that BMP7 administration may be used as a therapeutic strategy to reduce the damaging excitotoxic effects following SCI.
Collapse
Affiliation(s)
- Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | | | | | | | | |
Collapse
|
47
|
Airavaara M, Shen H, Kuo CC, Peränen J, Saarma M, Hoffer B, Wang Y. Mesencephalic astrocyte-derived neurotrophic factor reduces ischemic brain injury and promotes behavioral recovery in rats. J Comp Neurol 2009; 515:116-24. [PMID: 19399876 DOI: 10.1002/cne.22039] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF), also known as arginine-rich, mutated in early stage of tumors (ARMET), is a secreted protein that reduces endoplasmic reticulum (ER) stress. Previous studies have shown that MANF mRNA expression and protein levels are increased in the cerebral cortex after brain ischemia, a condition that induces ER stress. The function of MANF during brain ischemia is still not known. The purpose of this study was to examine the protective effect of MANF after ischemic brain injury. Recombinant human MANF was administrated locally to the cerebral cortex before a 60-min middle cerebral artery occlusion (MCAo) in adult rats. Triphenyltetrazolium chloride (TTC) staining indicated that pretreatment with MANF significantly reduced the volume of infarction at 2 days after MCAo. MANF also attenuated TUNEL labeling, a marker of cell necrosis/apoptosis, in the ischemic cortex. Animals receiving MANF pretreatment demonstrated a decrease in body asymmetry and neurological score as well as an increase in locomotor activity after MCAo. Taken together, these data suggest that MANF has neuroprotective effects against cerebral ischemia, possibly through the inhibition of cell necrosis/apoptosis in cerebral cortex.
Collapse
Affiliation(s)
- Mikko Airavaara
- National Institute on Drug Abuse, I.R.P., Baltimore, Maryland 21224, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Sawada N, Kim HH, Moskowitz MA, Liao JK. Rac1 is a critical mediator of endothelium-derived neurotrophic activity. Sci Signal 2009; 2:ra10. [PMID: 19278959 PMCID: PMC2668716 DOI: 10.1126/scisignal.2000162] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The therapeutic potential of neurotrophic factors has been hampered by their inability to achieve adequate tissue penetration. Brain blood vessels, however, could be an alternative target for neurosalvage therapies by virtue of their close proximity to neurons. Here we show that hemizygous deletion of Rac1 in mouse endothelial cells (ECs) attenuates brain injury and edema after focal cerebral ischemia. Microarray analysis of Rac1(+/-) ECs revealed enrichment of stress response genes, basement membrane components, and neurotrophic factors that could affect neuronal survival. Consistent with these expression profiles, endothelial Rac1 hemizygosity enhanced antioxidative and endothelial barrier capacities and potentiated paracrine neuroprotective activities through the up-regulation of the neurotrophic factor, artemin. Endothelial Rac1, therefore, could be an important therapeutic target for promoting endothelial barrier integrity and neurotrophic activity.
Collapse
Affiliation(s)
- Naoki Sawada
- Vascular Medicine Research, Brigham and Women's Hospital, 65 Landsdowne Street, Room 275, Cambridge, MA 02139, USA
| | - Hyung-Hwan Kim
- Vascular Medicine Research, Brigham and Women's Hospital, 65 Landsdowne Street, Room 275, Cambridge, MA 02139, USA
| | - Michael A. Moskowitz
- Stroke and Neurovascular Regulation Laboratory, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - James K. Liao
- Vascular Medicine Research, Brigham and Women's Hospital, 65 Landsdowne Street, Room 275, Cambridge, MA 02139, USA
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
50
|
Bani-Yaghoub M, Tremblay RG, Ajji A, Nzau M, Gangaraju S, Chitty D, Zurakowski B, Sikorska M. Neuroregenerative strategies in the brain: emerging significance of bone morphogenetic protein 7 (BMP7). Biochem Cell Biol 2008; 86:361-9. [DOI: 10.1139/o08-116] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Every year thousands of people suffer from brain injuries and stroke, and develop motor, sensory, and cognitive problems as a result of neuronal loss in the brain. Unfortunately, the damaged brain has a limited ability to enact repair and current modes of treatment are not sufficient to offset the damage. An extensive list of growth factors, neurotrophic factors, cytokines, and drugs has been explored as potential therapies. However, only a limited number of them may actually have the potential to effectively offset the brain injury or stroke-related problems. One of the treatments considered for future brain repair is bone morphogenetic protein 7 (BMP7), a factor currently used in patients to treat non-neurological diseases. The clinical application of BMP7 is based on its neuroprotective role in stroke animal models. This paper reviews the current approaches considered for brain repair and discusses the novel convergent strategies by which BMP7 potentially can induce neuroregeneration.
Collapse
Affiliation(s)
- Mahmud Bani-Yaghoub
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Roger G. Tremblay
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Abdellah Ajji
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Munyao Nzau
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Sandhya Gangaraju
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - David Chitty
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Bogdan Zurakowski
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| | - Marianna Sikorska
- Neurogenesis and Brain Repair Group, Neurobiology Program, Institute for Biological Sciences, National Research Council Canada, 1200 Montreal Rd., Bldg. M-54, Ottawa, ON K1A 0R6, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Functional Polymer Systems Group, Industrial Materials Institute, National Research Council Canada, Boucherville, QC J4B 6Y4, Canada
- Division of Neurosurgery, Children’s Hospital of Eastern Ontario, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|