1
|
Niaz M, Iftikhar K, Shahid M, Faizi S, Usman Simjee S. Quinic acid contributes to neurogenesis: Targeting Notch pathway a key player in hippocampus. Brain Res 2024; 1846:149291. [PMID: 39442647 DOI: 10.1016/j.brainres.2024.149291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/21/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
Coordinated proliferation and differentiation of neural stem cells (NSCs) results in continuous neurogenesis. The present study provides novel insights into the Notch intracellular signaling in neuronal cell proliferation, maintenance, migration, and differentiation regulated by naturally based Quinic acid (QA) in primary hippocampal cell culture. Further, this study might help in the discovery and development of lead molecules that can overcome the challenges in the treatment of neurodegenerative diseases. The growth supporting effect of QA was studied using Alamar Blue assay. The migratory potential of QA was evaluated using scratch assay. The in vitro H2O2-induced oxidative stress model was used to upregulate neuronal survival after QA treatment. The RT-qPCR and immunocytochemical analysis were performed for selected markers of Notch signaling to determine the proliferation, differentiation, and maintenance of NSCs at gene and molecular levels. The Mash1 and Ngn2 are the upstream proneural genes of the Notch pathway which were included to evaluate the differentiation of NSCs into mature neurons after treatment with QA. Furthermore, regarding the role of QA in maintaining the pool of NPCs, we used Notch1 and Hes1 markers for proliferation analysis. Also, secondary neuronal markers i.e. Pax6, PCNA, and Mcm2 were included in this study and their gene expression analysis was analyzed following treatment with QA. Based on the study's results, we suggest that naturally based QA can promote the growth and differentiation of neonatal NSCs residing in hippocampal regions into neuronal lineage. Therefore, we propose that the neurogenic potential of QA can be employed to prevent and treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Maryam Niaz
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Kanwal Iftikhar
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Maha Shahid
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shaheen Faizi
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shabana Usman Simjee
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
2
|
Polysialylation and disease. Mol Aspects Med 2020; 79:100892. [PMID: 32863045 DOI: 10.1016/j.mam.2020.100892] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/31/2022]
Abstract
Polysialic acid (polySia, PSA) is a unique constituent of the glycocalyx on the surface of bacterial and vertebrate cells. In vertebrates, its biosynthesis is highly regulated, not only in quantity and quality, but also in time and location, which allows polySia to be involved in various important biological phenomena. Therefore, impairments in the expression and structure of polySia sometimes relate to diseases, such as schizophrenia, bipolar disorder, and cancer. Some bacteria express polySia as a tool for protecting themselves from the host immune system during invasion. PolySia is proven to be a biosafe material; polySia, as well as polySia-recognizing molecules, are key therapeutic agents. This review first comprehensive outlines the occurrence, features, biosynthesis, and functions of polySia and subsequently focuses on the related diseases.
Collapse
|
3
|
Zuo Y, Wang J, Enkhjargal B, Doycheva D, Yan X, Zhang JH, Liu F. Neurogenesis changes and the fate of progenitor cells after subarachnoid hemorrhage in rats. Exp Neurol 2019; 311:274-284. [PMID: 30359565 DOI: 10.1016/j.expneurol.2018.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular disease that leads to poor outcomes. Neurogenesis, an essential recovery mechanism after brain injury, has not been fully elucidated after SAH. METHODS A total of 122 SD rats were used in this study. For experiment one, the rats were randomly divided into six groups: sham and SAH with different time points (1,3,5,7,14 days) (n = 12/group). An endovascular perforation method was conducted for SAH model. Rats were injected with 5-Bromo-2'-deoxyuridine (BrdU, 50 mg/kg) 24 h before euthanasia at different time points after SAH. The BrdU labeled cells were detected by immunohistochemistry; Doublecortin (DCX) and glial fibrillary acidic protein (GFAP) were measured by western blot and immunohistochemistry. For experiment two, rats were randomly divided into five groups: sham and SAH with different time points (1, 2, 4, 8 weeks) (n = 6/group). Rats received BrdU (50 mg/kg) once daily for 7 days after the induction of SAH. Double immunofluorescence staining was used to verify proliferation, differentiation and migration of progenitor cells. Rotarod test and water maze used to test the neurobehavioral recovery. RESULTS Our results showed that BrdU positive cells in hippocampus changed overtime after SAH. BrdU positive cells decreased as early as 1 day reaching lowest levels at 3 days after SAH, after which it gradually recovered. Similar change patterns were observed with DCX, which was reversed with GFAP. In addition, BrdU did not co-localize with cleaved caspase-3. The BrdU positive cells mainly differentiated into immature neurons for short-term fate, whereas they differentiated into mature neurons for long-term fate but not astrocytes, which facilitated neurobehavioral recovery after SAH. CONCLUSION Neurogenesis in the hippocampus changes overtime after SAH. The neuronal progenitor cells may play an essential role in the neurobehavioral recovery after brain injury induced by SAH, since short-term progenitors helped with the recovery of immature neurons in the hippocampus, whereas long-term progenitors differentiated into mature neurons.
Collapse
Affiliation(s)
- Yuchun Zuo
- Department of Neurosurgery, The third XiangYa Hospital, Central South University, Changsha 410013, China
| | - Jikai Wang
- Department of Neurosurgery, The third XiangYa Hospital, Central South University, Changsha 410013, China
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, CA 92354, USA
| | - Desislava Doycheva
- Department of Physiology and Pharmacology, Loma Linda University, CA 92354, USA
| | - Xiaoxin Yan
- Department of Anatomy, XiangYa Medical School, Central South University, Changsha 410013, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, CA 92354, USA.
| | - Fei Liu
- Department of Neurosurgery, The third XiangYa Hospital, Central South University, Changsha 410013, China.
| |
Collapse
|
4
|
Adamczak J, Aswendt M, Kreutzer C, Rotheneichner P, Riou A, Selt M, Beyrau A, Uhlenküken U, Diedenhofen M, Nelles M, Aigner L, Couillard-Despres S, Hoehn M. Neurogenesis upregulation on the healthy hemisphere after stroke enhances compensation for age-dependent decrease of basal neurogenesis. Neurobiol Dis 2016; 99:47-57. [PMID: 28007584 DOI: 10.1016/j.nbd.2016.12.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/12/2016] [Accepted: 12/18/2016] [Indexed: 01/27/2023] Open
Abstract
Stroke is a leading cause of death and disability worldwide with no treatment for the chronic phase available. Interestingly, an endogenous repair program comprising inflammation and neurogenesis is known to modulate stroke outcome. Several studies have shown that neurogenesis decreases with age but the therapeutic importance of endogenous neurogenesis for recovery from cerebral diseases has been indicated as its ablation leads to stroke aggravation and worsened outcome. A detailed characterization of the neurogenic response after stroke related to ageing would help to develop novel and targeted therapies. In an innovative approach, we used the DCX-Luc mouse, a transgenic model expressing luciferase in doublecortin-positive neuroblasts, to monitor the neurogenic response following middle cerebral artery occlusion over three weeks in three age groups (2, 6, 12months) by optical imaging while the stroke lesion was monitored by quantitative MRI. The individual longitudinal and noninvasive time profiles provided exclusive insight into age-dependent decrease in basal neurogenesis and neurogenic upregulation in response to stroke which are not accessible by conventional BrdU-based measures of cell proliferation. For cortico-striatal strokes the maximal upregulation occurred at 4days post stroke followed by a continuous decrease to basal levels by three weeks post stroke. Older animals effectively compensated for reduced basal neurogenesis by an enhanced sensitivity to the cerebral lesion, resulting in upregulated neurogenesis levels approaching those measured in young mice. In middle aged and older mice, but not in the youngest ones, additional upregulation of neurogenesis was observed in the contralateral healthy hemisphere. This further substantiates the increased propensity of older brains to respond to lesion situation. Our results clearly support the therapeutic relevance of endogenous neurogenesis for stroke recovery and particularly in older brains.
Collapse
Affiliation(s)
- Joanna Adamczak
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany; Percuros B.V., Drienerlolaan 5-Zuidhorst, 7522 NB Enschede, The Netherlands
| | - Markus Aswendt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Christina Kreutzer
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Peter Rotheneichner
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Adrien Riou
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Marion Selt
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Andreas Beyrau
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Ulla Uhlenküken
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Michael Diedenhofen
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Melanie Nelles
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Austria
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Gleuelerstrasse 50, 50931 Cologne, Germany; Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; Percuros B.V., Drienerlolaan 5-Zuidhorst, 7522 NB Enschede, The Netherlands.
| |
Collapse
|
5
|
Qu H, Zhao M, Zhao S, Xiao T, Song C, Cao Y, Jolkkonen J, Zhao C. Forced limb-use enhanced neurogenesis and behavioral recovery after stroke in the aged rats. Neuroscience 2015; 286:316-24. [DOI: 10.1016/j.neuroscience.2014.11.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/21/2014] [Accepted: 11/10/2014] [Indexed: 11/25/2022]
|
6
|
Mino M, Kamii H, Fujimura M, Kondo T, Takasawa S, Okamoto H, Yoshimoto T. Temporal changes of neurogenesis in the mouse hippocampus after experimental subarachnoid hemorrhage. Neurol Res 2013; 25:839-45. [PMID: 14669527 DOI: 10.1179/016164103771953934] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recent studies indicate the existence of progenitor cells and their potential for neurogenesis in the subventricular zone (SVZ) and the hippocampus dentate gyrus (DG) of normal adult mammalian brain. Increased neurogenesis has been shown following cerebral ischemia and traumatic brain injury; however, the involvement of neurogenesis in subarachnoid hemorrhage (SAH) has not been examined. Adult male CD-1 mice were subjected to SAH by endovascular perforation of the left anterior cerebral artery. Mice received intraperitoneal injections of the cell proliferation-specific marker 5'-bromodeoxyuridine (BrdU) after SAH induction. BrdU incorporation was examined from 1 to 30 days after SAH by immunohistochemistry. The BrdU-positive cells were detected in SVZ and DG of normal control brain, and were significantly decreased in both areas three days after SAH. The number of these cells had recovered to its control level seven days after SAH. Double staining with BrdU and NeuN indicated that the majority of the BrdU-positive cells migrating into the granular cell layer of the DG became NeuN-positive 30 days after SAH. In conclusion, temporal changes of the neurogenesis as shown in the present study suggest that neurogenesis in the hippocampus may affect functional outcome after SAH. The induction of the neurogenesis can provide therapeutic value against SAH.
Collapse
Affiliation(s)
- Masaki Mino
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | | | | | | | | | | | | |
Collapse
|
7
|
Lecanu L. Sex, the Underestimated Potential Determining Factor in Brain Tissue Repair Strategy. Stem Cells Dev 2011; 20:2031-5. [DOI: 10.1089/scd.2011.0188] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Laurent Lecanu
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Puchkov D, Leshchyns'ka I, Nikonenko AG, Schachner M, Sytnyk V. NCAM/spectrin complex disassembly results in PSD perforation and postsynaptic endocytic zone formation. ACTA ACUST UNITED AC 2011; 21:2217-32. [PMID: 21339376 DOI: 10.1093/cercor/bhq283] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Mechanisms inducing perforation of the postsynaptic density (PSD) are poorly understood. We show that neural cell adhesion molecule- deficient (NCAM-/-) hippocampal neurons have an abnormally high percentage of synapses with perforated PSDs. The percentage of synapses with perforated PSDs is also increased in wild-type (NCAM+/+) neurons after the disruption of the NCAM/spectrin complex indicating that the NCAM-assembled spectrin cytoskeleton maintains the structural integrity of PSDs. We demonstrate that PSD perforations contain endocytic zones involved in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) internalization. Induction of long-term potentiation in NCAM+/+ neurons accompanied by insertion of AMPAR into the neuronal cell surface is subsequently followed by formation of perforated synapses and AMPAR endocytosis suggesting that perforation of PSDs is important for membrane homeostasis in activated synapses. In NCAM-/- or NCAM+/+ neurons with dissociated spectrin meshwork, AMPAR endocytosis is enhanced under conditions of basal activity. An abnormally high rate of postsynaptic membrane endocytosis may thus contribute to brain pathologies associated with mutations in NCAM or spectrin.
Collapse
Affiliation(s)
- Dmytro Puchkov
- Zentrum für Molekulare Neurobiologie, Universitätskrankenhaus Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | | | | | | |
Collapse
|
9
|
Enhanced expression of vascular endothelial growth factor receptor-3 in the subventricular zone of stroke-lesioned rats. Neurosci Lett 2010; 469:194-8. [DOI: 10.1016/j.neulet.2009.11.073] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 11/18/2009] [Accepted: 11/27/2009] [Indexed: 11/17/2022]
|
10
|
Chen X, Tian Y, Yao L, Zhang J, Liu Y. Hypoxia stimulates proliferation of rat neural stem cells with influence on the expression of cyclin D1 and c-Jun N-terminal protein kinase signaling pathway in vitro. Neuroscience 2009; 165:705-14. [PMID: 19909792 DOI: 10.1016/j.neuroscience.2009.11.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 10/26/2009] [Accepted: 11/04/2009] [Indexed: 02/01/2023]
Abstract
Ischemia/hypoxia is known to induce the neural stem cells proliferation and neural differentiation in rodent and human brain; however its mechanisms remain largely unknown. In this study we investigated the effect of hypoxia on neural stem cells (NSCs) proliferation with the expression of cyclin D1 and the phosphorylation of mitogen-activated protein kinases (MAPK) signaling molecules. NSCs were cultured from cortex of fetal Sprague-Dawley rats on embryonic day 5.5. The hypoxia was made using a microaerophilic incubation system. The NSCs proliferation was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, diameter measurement of neurospheres, bromodeoxyuridine (BrdU) incorporation assay and cell cycle analysis. The cell death of NSCs was evaluated by terminal dUTP nick-end labeling (TUNEL) assay. The expression of cyclin D1, phosphorylated extracellular signal regulated kinase (ERK), c-Jun N-terminal protein kinase (JNK) and p38 were analyzed by immunoblotting assay. The results showed that hypoxia increased NSCs proliferation in cell amount, diameter of neurospheres, BrdU incorporation and cell division, and the highest proliferation of the NSCs was observed with 12 h hypoxic treatment; hypoxia did not decrease cell death of NSCs; after hypoxic treatment, the expression of cyclin D1 increased, meanwhile P-JNK2 level increased, P-p38 decreased, and no significant change in P-ERK2 level compared to normoxic cultures. JNK inhibitor SP600125 attenuated the increase of cyclin D1 induced by hypoxia. These findings propose that hypoxia increases cyclin D1 expression through activation of JNK in NSCs of rat in vitro, suggesting a novel possible mechanism for hypoxia-induced proliferation of NSCs.
Collapse
Affiliation(s)
- X Chen
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, The State Key Subject for Physiology, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, PR China
| | | | | | | | | |
Collapse
|
11
|
Horie N, So K, Moriya T, Kitagawa N, Tsutsumi K, Nagata I, Shinohara K. Effects of oxygen concentration on the proliferation and differentiation of mouse neural stem cells in vitro. Cell Mol Neurobiol 2008; 28:833-45. [PMID: 18236013 PMCID: PMC11515460 DOI: 10.1007/s10571-007-9237-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2007] [Accepted: 10/31/2007] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral ischemia is known to elicit the activation of neural stem cells (NSCs); however its mechanism is not fully determined. Although oxygen concentration is known to mediate many ischemic actions, there has been little attention given to the role of pathological oxygen changes under cerebral ischemia on the activation of NSCs. We investigated the effects of various oxygen concentrations on mouse neural stem cells in vitro. METHODS NSCs were cultured from the ganglionic eminence of fetal ICR mice on embryonic day 15.5 using a neurosphere method. The effects of oxygen concentrations on proliferation, differentiation, and cell death of NSCs were evaluated by bromodeoxyuridine (BrdU) incorporation, immunocytochemistry, and TUNEL assay, respectively. RESULTS The highest proliferation and the neuronal differentiation of the NSCs were observed in 2% oxygen, which yielded significantly higher proportions of both BrdU-labeled cells and Tuj1-positive cells when compared with 20% and 4% oxygen. On the other hand, the differentiation to the astrocytes was not affected by oxygen concentrations, except in the case of anoxia (0% oxygen). The cell death of the NSCs increased in lower oxygen conditions and peaked at anoxia. Furthermore, the switching of the neuronal subtype differentiation from GABA-positive to glutamate-positive neurons was observed in lower oxygen conditions. CONCLUSIONS These findings raise the possibility that reduced oxygen levels occurring with cerebral ischemia enhance NSC proliferation and neural differentiation, and that mild hypoxia (2% oxygen), which is known to occur in the ischemic penumbra, is suitable for abundant neuronal differentiation.
Collapse
Affiliation(s)
- Nobutaka Horie
- Department of Neurobiology and Behavior, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
- Department of Neurosurgery, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| | - Kenji So
- Department of Neurobiology and Behavior, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Takahiro Moriya
- Department of Neurobiology and Behavior, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Naoki Kitagawa
- Department of Neurosurgery, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| | - Keisuke Tsutsumi
- Department of Neurosurgery, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| | - Izumi Nagata
- Department of Neurosurgery, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| | - Kazuyuki Shinohara
- Department of Neurobiology and Behavior, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| |
Collapse
|
12
|
Kleim JA, Jones TA. Principles of experience-dependent neural plasticity: implications for rehabilitation after brain damage. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2008; 51:S225-S239. [PMID: 18230848 DOI: 10.1044/1092-4388(2008/018)] [Citation(s) in RCA: 1367] [Impact Index Per Article: 80.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
PURPOSE This paper reviews 10 principles of experience-dependent neural plasticity and considerations in applying them to the damaged brain. METHOD Neuroscience research using a variety of models of learning, neurological disease, and trauma are reviewed from the perspective of basic neuroscientists but in a manner intended to be useful for the development of more effective clinical rehabilitation interventions. RESULTS Neural plasticity is believed to be the basis for both learning in the intact brain and relearning in the damaged brain that occurs through physical rehabilitation. Neuroscience research has made significant advances in understanding experience-dependent neural plasticity, and these findings are beginning to be integrated with research on the degenerative and regenerative effects of brain damage. The qualities and constraints of experience-dependent neural plasticity are likely to be of major relevance to rehabilitation efforts in humans with brain damage. However, some research topics need much more attention in order to enhance the translation of this area of neuroscience to clinical research and practice. CONCLUSION The growing understanding of the nature of brain plasticity raises optimism that this knowledge can be capitalized upon to improve rehabilitation efforts and to optimize functional outcome.
Collapse
Affiliation(s)
- Jeffrey A Kleim
- McKnight Brain Institute, University of Florida, Gainesville, and Brain Rehabilitation Research Center (151A), Malcom Randall VA Hospital, 1610 SW Archer Road, Gainesville, FL 32610, USA.
| | | |
Collapse
|
13
|
Chambon C, Paban V, Manrique C, Alescio-Lautier B. Behavioral and immunohistological effects of cholinergic damage in immunolesioned rats: Alteration of c-Fos and polysialylated neural cell adhesion molecule expression. Neuroscience 2007; 147:893-905. [PMID: 17601671 DOI: 10.1016/j.neuroscience.2007.05.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 05/16/2007] [Accepted: 05/21/2007] [Indexed: 11/19/2022]
Abstract
The aim of this study was to determine the brain structures as well as the plasticity events associated with the behavioral effects of cholinergic damage. Rats were submitted to injection of 192 IgG-saporin in the medial septum/diagonal band of Broca complex and the nucleus basalis magnocellularis. The immunohistochemical expression of c-Fos protein and PSA-NCAM (polysialylated neural cell adhesion molecule) and the behavioral performances in the nonmatching-to-position task were assessed at various post-lesion times. Thus, 3 days after injection of the immunotoxin, increased c-Fos labeling was observed in the areas of infusion, indicating these cells were undergoing some plastic changes and/or apoptotic processes. A drastic increase was observed in the number of PSA-NCAM positive cells and in their dendritic arborization in the dentate gyrus. At 7 days post-lesion, no behavioral deficit was observed in immunolesioned rats despite the drastic loss of cholinergic neurons. These neurons showed decreased c-Fos protein expression in the piriform and entorhinal cortex and in the dentate gyrus. In the latter, PSA-NCAM induction was high, suggesting that remodeling occurred, which in turn might contribute to sustaining some mnemonic function in immunolesioned rats. At 1 month, cholinergic neurons totally disappeared and behavioral deficits were drastic. c-Fos expression showed no change. In contrast, the increased PSA-NCAM-labeling observed at short post-lesion times was maintained but the plastic changes due to this molecule could not compensate the behavioral deficit caused by the immunotoxin. Thus, as the post-lesion time increases, a gradual degeneration process should occur that may contribute to mnemonic impairments. This neuronal loss leads to molecular and cellular alterations, which in turn may aggravate cognitive deficits.
Collapse
Affiliation(s)
- C Chambon
- Université d'Aix-Marseille I, Laboratoire de Neurobiologie Intégrative et Adaptative, UMR/CNRS 6149, 3 Place Victor Hugo, 13331 Marseille Cedex 03, France
| | | | | | | |
Collapse
|
14
|
Moriya T, Horie N, Mitome M, Shinohara K. Melatonin influences the proliferative and differentiative activity of neural stem cells. J Pineal Res 2007; 42:411-8. [PMID: 17439558 DOI: 10.1111/j.1600-079x.2007.00435.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Though melatonin has a wide variety of biological functions, its effects on the neural stem cells (NSCs) is still unknown. In this study, we examined the effects of melatonin at either physiological (0.01-10 nm) or pharmacological concentrations (1-100 microM) on the proliferation and neural and astroglial differentiation of NSCs derived from the mouse embryo striatum using an in vitro culture system. We found that melatonin at pharmacological concentrations, but not at physiological concentrations, suppressed epidermal growth factor (EGF)-stimulated NSC proliferation (increment of viable cells, DNA synthesis and neurosphere formation) in a concentration-dependent manner. Furthermore, treatment with melatonin at a pharmacological concentration during the proliferation period facilitated 1% FBS-induced neural differentiation of NSCs without affecting the astroglial differentiation. In contrast, the treatment with melatonin at pharmacological concentrations during the differentiation period decreased the neural differentiation of the NSCs. As with melatonin, MCI-186, an antioxidant, suppressed EGF-stimulated NSC proliferation and facilitated the subsequent neural differentiation of NSCs. These results suggest that melatonin exerts potent modulatory effects on NSC functions including the suppression of the proliferation and facilitation of neuronal differentiation, likely via its antioxidant activity. As neurogenesis is thought to play an important role in ameliorating the deficit in neurodegenerative diseases, melatonin might be beneficially used for the treatment diseases such as cerebral infarction.
Collapse
Affiliation(s)
- Takahiro Moriya
- Department of Translational Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | | | | | | |
Collapse
|
15
|
Duveau V, Arthaud S, Rougier A, Le Gal La Salle G. Polysialylation of NCAM is upregulated by hyperthermia and participates in heat shock preconditioning-induced neuroprotection. Neurobiol Dis 2007; 26:385-95. [PMID: 17336079 DOI: 10.1016/j.nbd.2007.01.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Revised: 01/16/2007] [Accepted: 01/22/2007] [Indexed: 11/23/2022] Open
Abstract
"Brain tolerance"--a phenomenon in which a subtoxic challenge confers resistance to subsequent brain injuries--provides an ideal opportunity for investigating endogenous neuroprotective mechanisms. We investigated the potential role of the polysialylated (PSA) form of neural cell adhesion molecule (NCAM), which is thought to play a key role in plasticity. In a model where prior exposure to heat shock protects against kainate-induced cell damage in the hippocampus, we show that hyperthermia upregulates PSA-NCAM expression for at least 1 week, without affecting neurogenesis. Pharmacological manipulation of heat shock protein (HSP) expression demonstrates a tight positive link between HSP70 and PSA-NCAM. Finally, the presence of PSA was functionally linked to brain tolerance, as protection against kainate-induced cell death by heat shock pre-exposure was abolished in the absence of NCAM polysialylation. The upregulation of PSA-NCAM by hyperthermia may have a significant impact on hippocampal plasticity, permitting induction of the complex molecular cascade responsible for neuroprotection.
Collapse
Affiliation(s)
- V Duveau
- Université Bordeaux 2, CNRS UMR 5227-Mouvement, Adaptation, Cognition, 146, rue Léo Saignat, 33076 Bordeaux Cedex, France
| | | | | | | |
Collapse
|
16
|
Mazzetti S, Ortino B, Inverardi F, Frassoni C, Amadeo A. PSA-NCAM in the developing and mature thalamus. Brain Res Bull 2006; 71:578-86. [PMID: 17292800 DOI: 10.1016/j.brainresbull.2006.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Revised: 11/28/2006] [Accepted: 11/30/2006] [Indexed: 11/28/2022]
Abstract
The polysialylated form of the neural cell adhesion molecule (PSA-NCAM) is involved in several morphogenetic processes of the central nervous system. In the present study the expression of PSA-NCAM has been investigated in the rat thalamus during embryonic and postnatal development using light and electron microscopic immunocytochemical techniques. At all the examined ages, PSA-NCAM staining in the thalamus was mainly observed along neuronal plasmatic membranes and absent in astrocytes identified by labelling with cytoskeletal (vimentin and glial fibrillary acidic protein) and membrane (GABA transporter-3) markers. At embryonic day 14 the immunoreactivity was restricted to the dorsal thalamic mantle and to the region of reticular thalamic migration and subsequently it extended throughout the whole thalamic primordium. PSA-NCAM labelling remained intense and homogeneously distributed along perinatal period, but from P4 it began to decrease selectively, persisting throughout adulthood only in the reticular nucleus, ventral lateral geniculate nucleus and midline and intralaminar nuclei. The expression of this adhesion molecule differed in areas characterized by the presence of neurons containing distinct calcium binding proteins, as PSA-NCAM labelling was intense around calretinin-positive neurons, whereas it decreased in some calbindin-immunoreactive regions. These findings show evidence of a selective neuronal expression of PSA-NCAM in developing thalamus, supporting its suggested role in cell migration and synaptogenesis as it occurs in the cerebral cortex. In adulthood PSA-NCAM could instead be a marker of thalamic nuclei that retain a potential for synaptic plasticity.
Collapse
Affiliation(s)
- Samanta Mazzetti
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Università degli Studi di Milano, Via Celoria 26, 20133 Milano, Italy.
| | | | | | | | | |
Collapse
|
17
|
Bonfanti L. PSA-NCAM in mammalian structural plasticity and neurogenesis. Prog Neurobiol 2006; 80:129-64. [PMID: 17029752 DOI: 10.1016/j.pneurobio.2006.08.003] [Citation(s) in RCA: 347] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 08/04/2006] [Accepted: 08/21/2006] [Indexed: 12/14/2022]
Abstract
Polysialic acid (PSA) is a linear homopolymer of alpha2-8-N acetylneuraminic acid whose major carrier in vertebrates is the neural cell adhesion molecule (NCAM). PSA serves as a potent negative regulator of cell interactions via its unusual biophysical properties. PSA on NCAM is developmentally regulated thus playing a prominent role in different forms of neural plasticity spanning from embryonic to adult nervous system, including axonal growth, outgrowth and fasciculation, cell migration, synaptic plasticity, activity-induced plasticity, neuronal-glial plasticity, embryonic and adult neurogenesis. The cellular distribution, developmental changes and possible function(s) of PSA-NCAM in the central nervous system of mammals here are reviewed, along with recent findings and theories about the relationships between NCAM protein and PSA as well as the role of different polysialyltransferases. Particular attention is focused on postnatal/adult neurogenesis, an issue which has been deeply investigated in the last decade as an example of persisting structural plasticity with potential implications for brain repair strategies. Adult neurogenic sites, although harbouring all subsequent steps of cell differentiation, from stem cell division to cell replacement, do not faithfully recapitulate development. After birth, they undergo morphological and molecular modifications allowing structural plasticity to adapt to the non-permissive environment of the mature nervous tissue, that are paralled by changes in the expression of PSA-NCAM. The use of PSA-NCAM as a marker for exploring differences in structural plasticity and neurogenesis among mammalian species is also discussed.
Collapse
Affiliation(s)
- Luca Bonfanti
- Department of Veterinary Morphophysiology, University of Turin, Via Leonardo da Vinci 44, 10095 Grugliasco, Italy.
| |
Collapse
|
18
|
Abstract
Background and Purpose—
Stroke induced by middle cerebral artery occlusion (MCAO) triggers increased neurogenesis in the damaged striatum and nondamaged hippocampus of young adult rodents. We explored whether stroke influences neurogenesis similarly in the aged brain.
Methods—
Young adult (3 months) and old (15 months) rats were subjected to 1 hour of MCAO, and new cells were labeled by intraperitoneal injection of 5-bromo-2′-deoxyuridine 5′-monophosphate (BrdU), a marker for dividing cells, for 2 weeks thereafter. Animals were euthanized at 7 weeks after the insult, and neurogenesis was assessed immunocytochemically with antibodies against BrdU and neuronal markers with epifluorescence or confocal microscopy.
Results—
Young and old rats exhibited the same increased numbers of new striatal neurons after stroke, despite basal cell proliferation in the subventricular zone being reduced in the aged brain. In contrast, both the number of stroke-generated granule cells and basal neurogenesis in the dentate subgranular zone were lower in old compared with young animals. Also, the ability of newly formed cells to differentiate into neurons was impaired in the aged dentate gyrus.
Conclusions—
Basal neurogenesis is impaired in the subgranular and subventricular zones of aged animals, but both regions react to stroke with increased formation of new neurons. The magnitude of striatal neurogenesis after stroke is similar in young and old animals, indicating that this potential mechanism for self-repair also operates in the aged brain.
Collapse
Affiliation(s)
- Vladimer Darsalia
- Laboratory of Neural Stem Cell Biology, University Hospital, The Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund, Sweden
| | | | | | | |
Collapse
|
19
|
Gribkoff VK, Winquist RJ. Voltage-gated cation channel modulators for the treatment of stroke. Expert Opin Investig Drugs 2005; 14:579-92. [PMID: 15926865 DOI: 10.1517/13543784.14.5.579] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Neuronal voltage-gated cation channels regulate the transmembrane flux of calcium, sodium and potassium. Neuronal ischaemia occurring during acute ischaemic stroke results in the breakdown in the normal function of these ion channels, contributing to a series of pathological events leading to cell death. A dramatic increase in the intracellular concentration of calcium during neuronal ischaemia plays a particularly important role in the neurotoxic cascade resulting in stroke-related acute neurodegeneration. One approach to provide therapeutic benefit following ischaemic stroke has been to target neuronal voltage-gated cation channels, and particularly blockers of calcium and sodium channels, for post-stroke neuroprotection. A recent development has been the identification of openers of large-conductance calcium- and voltage-dependent potassium channels (maxi-K channels), which hyperpolarize ischaemic neurons, reduce excitatory amino acid release, and reduce ischaemic calcium entry. Thus far, targeting these voltage-gated cation channels has not yet yielded significant clinical benefit. The reasons for this may involve the lack of small-molecule blockers of many neuronal members of these ion channel families and the design of preclinical stroke models, which do not adequately emulate the clinical condition and hence lack sufficient rigor to predict efficacy in human stroke. Furthermore, there may be a need for changes in clinical trial designs to optimise the selection of patients and the course of drug treatment to protect neurons during all periods of potential neuronal sensitivity to neuro-protectants. Clinical trials may also have to be powered to detect small effect sizes or be focused on patients more likely to respond to a particular therapy. The development of future solutions to these problems should result in an improved probability of success for the treatment of stroke.
Collapse
Affiliation(s)
- Valentin K Gribkoff
- Department of Biology, Scion Pharmaceuticals, Inc., 200 Boston Avenue, Suite 3600, Medford, MA 02155, USA.
| | | |
Collapse
|
20
|
Tao T, Xing-qun L, Heng W, Jie-kun L, Hua-xian Z, Tuan-lian L. The Property of Nao Yi-an granule in increasing Nestin immunoreactive neural stem cells in rat brains with intracerebral hemorrhage. Chin J Integr Med 2004. [DOI: 10.1007/bf02836431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
21
|
Horie N, Moriya T, Mitome M, Kitagawa N, Nagata I, Shinohara K. Lowered glucose suppressed the proliferation and increased the differentiation of murine neural stem cells in vitro. FEBS Lett 2004; 571:237-42. [PMID: 15280049 DOI: 10.1016/j.febslet.2004.06.085] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2004] [Accepted: 06/26/2004] [Indexed: 11/22/2022]
Abstract
Cerebral ischemia is known to activate endogenous neural stem cells (NSCs), but its mechanisms remain unknown. Since lowered glucose supply seems to mediate ischemic actions, we examined the effect of low glucose on NSC activities in vitro. Low glucose applied during the proliferation period diminished EGF-induced proliferation of NSCs without affecting subsequent differentiation, but low glucose directly exposed during the differentiation period facilitated the differentiation of NSCs into neurons and astrocytes. These findings suggest that low glucose facilitated NSC differentiation, but it diminished NSC proliferation. Moreover, the effect of low glucose may be dependent on the timing of application.
Collapse
Affiliation(s)
- Nobutaka Horie
- Division of Neurobiology and Behavior, Department of Translational Medical Science, Course of Medical and Dental Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | | | | | | | | | | |
Collapse
|
22
|
Jin K, Sun Y, Xie L, Peel A, Mao XO, Batteur S, Greenberg DA. Directed migration of neuronal precursors into the ischemic cerebral cortex and striatum. Mol Cell Neurosci 2004; 24:171-89. [PMID: 14550778 DOI: 10.1016/s1044-7431(03)00159-3] [Citation(s) in RCA: 408] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pathological processes, including cerebral ischemia, can enhance neurogenesis in the adult brain, but the fate of the newborn neurons that are produced and their role in brain repair are obscure. To determine if ischemia-induced neuronal proliferation is associated with migration of nascent neurons toward ischemic lesions, we mapped the migration of cells labeled by cell proliferation markers and antibodies against neuronal marker proteins, for up to 2 weeks after a 90-min episode of focal cerebral ischemia caused by occlusion of the middle cerebral artery. Doublecortin-immunoreactive cells in the rostral subventricular zone, but not the dentate gyrus, migrated into the ischemic penumbra of the adjacent striatum and, via the rostral migratory stream and lateral cortical stream, into the penumbra of ischemic cortex. These results indicate that after cerebral ischemia, new neurons are directed toward sites of brain injury, where they might be in a position to participate in brain repair and functional recovery.
Collapse
Affiliation(s)
- Kunlin Jin
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Each year hundreds of thousands of people must cope with the severe neurological consequences of a stroke. Current therapeutic strategies for stroke focus on acute treatment and neuroprotection. Unfortunately, these practices do little to reduce the long-term morbidity associated with the injury. To develop effective therapies that promote regeneration, we must have an understanding of the cellular and molecular events involved in the recovery from an insult. Neural stem and progenitor cells are likely to be affected during this period. Here we review how the proliferation, migration, and maturation of these precursors are affected by ischemia. Furthermore, we summarize data available on the underlying mechanisms and the therapeutic implications of these studies. The studies that we review provide compelling evidence that neural precursors resident in the brain initiate a compensatory response to stroke that results in the production of new neurons. Moreover, administration of growth factors can enhance this compensatory response. Based on these encouraging results, we may eventually be able to manipulate these precursors to improve recovery of function in individuals afflicted by this devastating injury.
Collapse
Affiliation(s)
- Ryan J Felling
- Department of Neuroscience and Anatomy, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | | |
Collapse
|
24
|
Jin K, Sun Y, Xie L, Batteur S, Mao XO, Smelick C, Logvinova A, Greenberg DA. Neurogenesis and aging: FGF-2 and HB-EGF restore neurogenesis in hippocampus and subventricular zone of aged mice. Aging Cell 2003; 2:175-83. [PMID: 12882410 DOI: 10.1046/j.1474-9728.2003.00046.x] [Citation(s) in RCA: 303] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neurogenesis, which may contribute to the ability of the adult brain to function normally and adapt to disease, nevertheless declines with advancing age. Adult neurogenesis can be enhanced by administration of growth factors, but whether the aged brain remains responsive to these factors is unknown. We compared the effects of intracerebroventricular fibroblast growth factor (FGF)-2 and heparin-binding epidermal growth factor-like growth factor (HB-EGF) on neurogenesis in the hippocampal dentate subgranular zone (SGZ) and the subventricular zone (SVZ) of young adult (3-month) and aged (20-month) mice. Neurogenesis, measured by labelling with bromodeoxyuridine (BrdU) and by expression of doublecortin, was reduced by approximately 90% in SGZ and by approximately 50% in SVZ of aged mice. HB-EGF increased BrdU labelling in SGZ at 3 months by approximately 60% and at 20 months by approximately 450%, which increased the number of BrdU-labelled cells in SGZ of aged mice to approximately 25% of that in young adults. FGF-2 also stimulated BrdU labelling in SGZ, by approximately 25% at 3 months and by approximately 250% at 20 months, increasing the number of newborn neurones in older mice to approximately 20% of that in younger mice. In SVZ, HB-EGF and FGF-2 increased BrdU incorporation by approximately 140% at 3 months and approximately 170% at 20 months, so the number of BrdU-labelled cells was comparable in untreated 3-month-old and growth factor-treated 20-month-old mice. These results demonstrate that the aged brain retains the capacity to respond to exogenous growth factors with increased neurogenesis, which may have implications for the therapeutic potential of neurogenesis enhancement in age-associated neurological disorders.
Collapse
Affiliation(s)
- Kunlin Jin
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Aging in the central nervous system is associated with progressive loss of function which is exacerbated by neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. The two primary cell replacement strategies involve transplantation of exogenous tissue, and activation of proliferation of endogenous cells. Transplanted tissue is used to either directly replace lost tissue, or to implant genetically engineered cells that secrete factors which promote survival and/or proliferation. However, successful application of any cell replacement therapy requires knowledge of the complex relationships between neural stem cells and the more restricted neural and glial progenitor cells. This review focuses on recent advances in the field of stem cell biology of the central nervous system, with an emphasis on cellular and molecular approaches to replacing cells lost in neurodegenerative disorders.
Collapse
Affiliation(s)
- T L Limke
- National Institute on Aging, Baltimore, MD 21224, USA.
| | | |
Collapse
|
26
|
Keyvani K, Schallert T. Plasticity-associated molecular and structural events in the injured brain. J Neuropathol Exp Neurol 2002; 61:831-40. [PMID: 12387449 DOI: 10.1093/jnen/61.10.831] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Injury to the brain appears to create a fertile ground for functional and structural plasticity that is, at least partly, responsible for functional recovery. Increases in dendritic arborization, spine density, and synaptogenesis in both peri-injury and intact cortical areas are the potential morphological strategies that enable the brain to reorganize its neuronal circuits. These injury-initiated alterations are time-dependent and frequently proceed in interaction with behavior-related signals. A complex concert of a variety of genes/proteins is required to tightly control these changes. Two broad categories of molecules appear to be involved. First, regulatory molecules or effector molecules with regulatory function, such as immediate early genes/transcription factors, kinase network proteins, growth factors, and neurotransmitter receptors, and second, structural proteins, such as adhesion molecules and compounds of synapses, growth cones, and cytoskeleton. A better understanding of the processes contributing to postinjury plasticity may be an advantage for developing new and more effective therapeutic approaches. This knowledge might also shed light on other forms of brain plasticity, such as those involved in learning processes or ontogeny.
Collapse
Affiliation(s)
- Kathy Keyvani
- Institute of Neuropathology, University of Muenster, Germany
| | | |
Collapse
|