1
|
Sanati M, Manavi MA, Noruzi M, Behmadi H, Akbari T, Jalali S, Sharifzadeh M, Khoobi M. Carbohydrates and neurotrophic factors: A promising partnership for spinal cord injury rehabilitation. BIOMATERIALS ADVANCES 2025; 166:214054. [PMID: 39332344 DOI: 10.1016/j.bioadv.2024.214054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
Spinal cord injury (SCI) leaves a temporary or enduring motor, sensory, and autonomic function loss, significantly impacting the patient's quality of life. Given their biocompatibility, bioactivity, and tunable attributes, three-dimensional scaffolds frequently employ carbohydrates to facilitate spinal cord regeneration. These scaffolds have also been engineered to be novel local delivery platforms that present distinct advantages in the targeted transportation of drug candidates to the damaged spinal cord, ensuring the right dosage and duration of administration. Neurotrophic factors have emerged as promising therapeutic candidates, preserved neuron survival and encouraged severed axons repair, although their local and continuous delivery is believed to produce considerable spinal cord rehabilitation. This study aims to discuss breakthroughs in scaffold engineering, exploiting carbohydrates as an essential part of their structure, and highlight their impact on spinal cord regeneration and sustained neurotrophic factors delivery to treat SCI.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Amin Manavi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Noruzi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
| | - Homayoon Behmadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Sara Jalali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, 1417614411 Tehran, Iran.
| |
Collapse
|
2
|
Yuan Y, Li D, Yu F, Kang X, Xu H, Zhang P. Effects of Akt/mTOR/p70S6K Signaling Pathway Regulation on Neuron Remodeling Caused by Translocation Repair. Front Neurosci 2020; 14:565870. [PMID: 33132828 PMCID: PMC7550644 DOI: 10.3389/fnins.2020.565870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/25/2020] [Indexed: 12/23/2022] Open
Abstract
Peripheral nerve injury repair has been considered a difficult problem in the field of trauma for a long time. Conventional surgical methods are not applicable in some special types of nerve injury, prompting scholars to seek to develop more effective nerve translocation repair technologies. The purpose of this study was to explore the functional state of neurons in injured lower limbs after translocation repair, with a view to preliminarily clarify the molecular mechanisms underlying this process. Eighteen Sprague–Dawley rats were divided into the normal, tibial nerve in situ repair, and common peroneal nerve transposition repair tibial nerve groups. Nerve function assessment and immunohistochemical staining of neurofilament 200 (NF-200), protein kinase B (Akt), mammalian target of rapamycin (mTOR), and ribosomal protein S6 kinase (p70S6K) in the dorsal root ganglia were performed at 12 weeks after surgery. Tibial nerve function and neuroelectrophysiological analysis, osmic acid staining, muscle strength testing, and muscle fiber staining showed that the nerve translocation repair could restore the function of the recipient nerve to a certain extent; however, the repair was not as efficient as the in situ repair. Immunohistochemical staining showed that the translocation repair resulted in changes in the microstructure of neuronal cell bodies, and the expressions of Akt, mTOR, and p70S6K in the three dorsal root ganglia groups were significantly different (p < 0.05). This study demonstrates that the nerve translocation repair technology sets up a new reflex loop, with the corresponding neuroskeletal adjustments, in which, donor neurons dominate the recipient nerves. This indicates that nerve translocation repair technology can lead to neuronal remodeling and is important as a supplementary treatment for a peripheral nerve injury. Furthermore, the Akt/mTOR/p70S6K signaling pathway may be involved in the formation of the new neural reflex loop created as a result of the translocation repair.
Collapse
Affiliation(s)
- Yusong Yuan
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| | - Dongdong Li
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,Department of Orthopedics, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Fei Yu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| | - Xuejing Kang
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| | - Hailin Xu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China.,Diabetic Foot Treatment Center, Peking University People's Hospital, Peking University, Beijing, China
| | - Peixun Zhang
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| |
Collapse
|
3
|
Baumann HJ, Betonio P, Abeywickrama CS, Shriver LP, Leipzig ND. Metabolomic and Signaling Programs Induced by Immobilized versus Soluble IFN γ in Neural Stem Cells. Bioconjug Chem 2020; 31:2125-2135. [PMID: 32820900 DOI: 10.1021/acs.bioconjchem.0c00338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neural stem cells (NSCs) provide a strategy to replace damaged neurons following traumatic central nervous system injuries. A major hurdle to translation of this therapy is that direct application of NSCs to CNS injury does not support sufficient neurogenesis due to lack of proper cues. To provide prolonged spatial cues to NSCs IFN-γ was immobilized to biomimetic hydrogel substrate to supply physical and biochemical signals to instruct the encapsulated NSCs to be neurogenic. However, the immobilization of factors, including IFN-γ, versus soluble delivery of the same factor, has been incompletely characterized especially with respect to activation of signaling and metabolism in cells over longer time points. In this study, protein and metabolite changes in NSCs induced by immobilized versus soluble IFN-γ at 7 days were evaluated. Soluble IFN-γ, refreshed daily over 7 days, elicited stronger responses in NSCs compared to immobilized IFN-γ, indicating that immobilization may not sustain signaling or has altered ligand/receptor interaction and integrity. However, both IFN-γ delivery types supported increased βIII tubulin expression in parallel with canonical and noncanonical receptor-signaling compared to no IFN-γ. Global metabolomics and pathway analysis revealed that soluble and immobilized IFN-γ altered metabolic pathway activities including energy, lipid, and amino acid synthesis, with soluble IFN-γ having the greatest metabolic impact overall. Finally, soluble and immobilized IFN-γ support mitochondrial voltage-dependent anion channel (VDAC) expression that correlates to differentiated NSCs. This work utilizes new methods to evaluate cell responses to protein delivery and provides insight into mode of action that can be harnessed to improve regenerative medicine-based strategies.
Collapse
Affiliation(s)
- Hannah J Baumann
- Department of Chemistry, The University of Akron, Akron, Ohio 44325, United States
| | - Patricia Betonio
- School of Nursing, The University of Akron, Akron, Ohio 44325, United States
| | | | - Leah P Shriver
- Department of Chemistry, The University of Akron, Akron, Ohio 44325, United States
| | - Nic D Leipzig
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
4
|
Wang J, Li X, He S, Hu L, Guo J, Huang X, Hu J, Qi Y, Chen B, Shang D, Wen Y. Regulation of the kynurenine metabolism pathway by Xiaoyao San and the underlying effect in the hippocampus of the depressed rat. JOURNAL OF ETHNOPHARMACOLOGY 2018; 214:13-21. [PMID: 29217494 DOI: 10.1016/j.jep.2017.11.037] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 10/31/2017] [Accepted: 11/30/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoyao San (XYS) is a classic Chinese herbal formula for treatment of depression. The present study aimed to investigate the antidepressant effects of XYS in a rat model of chronic unpredictable mild stress (CUMS) and the underlying mechanisms. MATERIALS AND METHODS A CUMS rat model of depression was established via 4 weeks of unpredictable stimulation. Then the rats were orally administered paroxetine and XYS for 2 weeks with continued stress. Behavioral assessments, including an open field test (OFT), sucrose preference test (SPT) and forced swim test (FST), were conducted to evaluate the antidepressant effects of XYS. The concentrations in rat plasma of tryptophan (Trp) and its metabolic products, including kynurenine (Kyn) and quinolinic acid (QUIN), were determined using high performance liquid chromatography tandem mass spectrometry with electrochemical detection (HPLC-MS/MS). The mRNA and protein levels in rat hippocampus of depression-related brain derived neurotrophic factor (BDNF), cyclic AMP response element binding protein (CREB) and nerve cell adhesion molecule (NCAM) were determined by real-time qPCR and Western blot, respectively. Enzyme Linked Immunosorbent Assay (ELISA) was used to detect the activities of indoleamine 2,3-dioxygenase (IDO) and kynurenine-3-monooxygenase (KMO) in rat plasma. RESULTS The results showed that a successful CUMS rat model was established through 4 weeks of continuous unpredictable stimulation, as indicated by the significant decrease in locomotor activity and increase in immobility time in the OFT, reduction in body weight and food intake etc. Compared with the normal group, the concentrations of Kyn and QUIN had significantly (p < 0.05) decreased at day 28 in the control group, but then improved after drug treatment with paroxetine and XYS. There were no obvious changes in the activities of IDO and KMO. Compared with the normal group, the mRNA of NCAM, CREB and BDNF were significantly down-regulated (p < 0.001) in the control group, BDNF gene was up-regulated by paroxetine or XYS treatment, NCAM and CREB gene did not change in XYS group, protein expressions of BDNF and CREB were significantly increased, and NCAM was significantly reduced (p < 0.05). CONCLUSIONS XYS reversed the abnormalities of the tryptophan-kynurenine metabolic pathways in depressed rats and achieved an excellent antidepressant effect. Its direct impact may be observed as changes in biological indicators in rat hippocampus tissue.
Collapse
Affiliation(s)
- Jiajia Wang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Guangzhou 510370, PR China; Guangzhou University of Traditional Chinese Medicine, 16 Airport Road, Guangzhou 510405, PR China
| | - Xiaofang Li
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Guangzhou 510370, PR China
| | - Shugui He
- Guangzhou University of Traditional Chinese Medicine, 16 Airport Road, Guangzhou 510405, PR China
| | - Lijun Hu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Guangzhou 510370, PR China
| | - Jiewen Guo
- Guangzhou Hospital of Traditional Chinese Medicine, 16 Zhuji Road, Guangzhou 510130, PR China
| | - Xiangning Huang
- Guangzhou Xinhai Hospital, Guangzhou, 167 Xingang West Road, Guangzhou 510300, PR China
| | - Jinqing Hu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Guangzhou 510370, PR China
| | - Yaoqun Qi
- Guangzhou University of Traditional Chinese Medicine, 16 Airport Road, Guangzhou 510405, PR China
| | - Bin Chen
- Guangzhou University of Traditional Chinese Medicine, 16 Airport Road, Guangzhou 510405, PR China
| | - Dewei Shang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Guangzhou 510370, PR China.
| | - Yuguan Wen
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Guangzhou 510370, PR China.
| |
Collapse
|
5
|
Gong L, Jiang C, Liu L, Wan S, Tan W, Ma S, Jia X, Wang M, Hu A, Shi Y, Zhang Y, Shen Y, Wang F, Chen Y. Transfection of neurotrophin-3 into neural stem cells using ultrasound with microbubbles to treat denervated muscle atrophy. Exp Ther Med 2017; 15:620-626. [PMID: 29403547 PMCID: PMC5780738 DOI: 10.3892/etm.2017.5439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022] Open
Abstract
Neurotrophin-3 (NT-3) has potential as a therapeutic agent for the treatment of patients with denervated muscle atrophy. However, the endogenous secretion of NT-3 is low and exogenous NT-3 lacks sufficient time to accumulate due to its short half-life. The transfection of NT-3 has been demonstrated to have a beneficial effect on denervated muscle and motor endplates. Neural stem cells (NSCs) differentiate into neurons and form motor endplate nerve-muscle connections. It has been previously demonstrated that local and noninvasive transfection can be performed using ultrasound with microbubbles (MBs). In the current study, hematoxylin and eosin, acetylcholinesterase and gold chloride staining, as well as transmission electron microscopy, were performed to verify the effects of this treatment strategy. The results demonstrated that using ultrasound with MBs for the transfection of NT-3 into NSCs, and their subsequent transplantation in vivo, attenuated the atrophy of denervated muscle and reduced motor endplate degeneration. This noninvasive, efficient and targeted treatment strategy may therefore be a potential treatment for patients with denervated muscle atrophy.
Collapse
Affiliation(s)
- Lin Gong
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Changqing Jiang
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Li Liu
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Shengxiang Wan
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Wen Tan
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Sushuang Ma
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xiaojian Jia
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Biomedical Research Institute, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, P.R. China.,Shenzhen Kangning Hospital & Shenzhen Mental Health Center, Shenzhen, Guangdong 518020, P.R. China
| | - Meiwei Wang
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Azhen Hu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Biomedical Research Institute, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - Yu Shi
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Yu Zhang
- Department of Ultrasound, The Third People's Hospital of Shenzhen, Shenzhen, Guangdong 518055, P.R. China
| | - Yuanyuan Shen
- Department of Biomedical Engineering, National Regional Key Technology Engineering Laboratory for Medical Ultrasound, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Feng Wang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Biomedical Research Institute, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, P.R. China.,Shenzhen Kangning Hospital & Shenzhen Mental Health Center, Shenzhen, Guangdong 518020, P.R. China.,Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, Henan 453002, P.R. China
| | - Yun Chen
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China.,Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Biomedical Research Institute, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
6
|
Rajan TS, Scionti D, Diomede F, Piattelli A, Bramanti P, Mazzon E, Trubiani O. Prolonged Expansion Induces Spontaneous Neural Progenitor Differentiation from Human Gingiva-Derived Mesenchymal Stem Cells. Cell Reprogram 2017; 19:389-401. [PMID: 29058474 DOI: 10.1089/cell.2017.0012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Neural crest-derived mesenchymal stem cells (MSCs) obtained from dental tissues received considerable interest in regenerative medicine, particularly in nerve regeneration owing to their embryonic origin and ease of harvest. Proliferation efficacy and differentiation capacity into diverse cell lineages propose dental MSCs as an in vitro tool for disease modeling. In this study, we investigated the spontaneous differentiation efficiency of dental MSCs obtained from human gingiva tissue (hGMSCs) into neural progenitor cells after extended passaging. At passage 41, the morphology of hGMSCs changed from typical fibroblast-like shape into sphere-shaped cells with extending processes. Next-generation transcriptomics sequencing showed increased expression of neural progenitor markers such as NES, MEIS2, and MEST. In addition, de novo expression of neural precursor genes, such as NRN1, PHOX2B, VANGL2, and NTRK3, was noticed in passage 41. Immunocytochemistry results showed suppression of neurogenesis repressors TP53 and p21, whereas Western blot results revealed the expression of neurotrophic factors BDNF and NT3 at passage 41. Our results showed the spontaneous efficacy of hGMSCs to differentiate into neural precursor cells over prolonged passages and that these cells may assist in producing novel in vitro disease models that are associated with neural development.
Collapse
Affiliation(s)
| | - Domenico Scionti
- 1 Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo," Messina , Italy
| | - Francesca Diomede
- 2 Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio," Chieti-Pescara, Chieti, Italy
| | - Adriano Piattelli
- 2 Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio," Chieti-Pescara, Chieti, Italy
| | - Placido Bramanti
- 1 Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo," Messina , Italy
| | - Emanuela Mazzon
- 1 Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo," Messina , Italy
| | - Oriana Trubiani
- 2 Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio," Chieti-Pescara, Chieti, Italy
| |
Collapse
|
7
|
Jiao Q, Li X, An J, Zhang Z, Chen X, Tan J, Zhang P, Lu H, Liu Y. Cell-Cell Connection Enhances Proliferation and Neuronal Differentiation of Rat Embryonic Neural Stem/Progenitor Cells. Front Cell Neurosci 2017; 11:200. [PMID: 28785204 PMCID: PMC5519523 DOI: 10.3389/fncel.2017.00200] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/26/2017] [Indexed: 01/22/2023] Open
Abstract
Cell-cell interaction as one of the niche signals plays an important role in the balance of stem cell quiescence and proliferation or differentiation. In order to address the effect and the possible mechanisms of cell-cell connection on neural stem/progenitor cells (NSCs/NPCs) proliferation and differentiation, upon passaging, NSCs/NPCs were either dissociated into single cell as usual (named Group I) or mechanically triturated into a mixture of single cell and small cell clusters containing direct cell-cell connections (named Group II). Then the biological behaviors including proliferation and differentiation of NSCs/NPCs were observed. Moreover, the expression of gap junction channel, neurotrophic factors and the phosphorylation status of MAPK signals were compared to investigate the possible mechanisms. Our results showed that, in comparison to the counterparts in Group I, NSCs/NPCs in Group II survived well with preferable neuronal differentiation. In coincidence with this, the expression of connexin 45 (Cx45), as well as brain derived neurotrophic factor (BDNF) and neurotrophin 3 (NT-3) in Group II were significantly higher than those in Group I. Phosphorylation of ERK1/2 and JNK2 were significantly upregulated in Group II too, while no change was found about p38. Furthermore, the differences of NSCs/NPCs biological behaviors between Group I and II completely disappeared when ERK and JNK phosphorylation were inhibited. These results indicated that cell-cell connection in Group II enhanced NSCs/NPCs survival, proliferation and neuronal differentiation through upregulating the expression of gap junction and neurotrophic factors. MAPK signals- ERK and JNK might contribute to the enhancement. Efforts for maintaining the direct cell-cell connection are worth making to provide more favorable niches for NSCs/NPCs survival, proliferation and neuronal differentiation.
Collapse
Affiliation(s)
- Qian Jiao
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong UniversityBeijing, China.,Department of Physiology, Medical College of Qingdao UniversityQingdao, China
| | - Xingxing Li
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong UniversityBeijing, China
| | - Jing An
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong UniversityBeijing, China
| | - Zhichao Zhang
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong UniversityBeijing, China
| | - Xinlin Chen
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong UniversityBeijing, China
| | - Jing Tan
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Department of Anesthesiology, The First Affiliated Hospital, Xi'an Jiaotong University Health Science CenterXi'an, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital, Health Science Center, Xi'an Jiaotong UniversityXi'an, China
| | - Haixia Lu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong UniversityBeijing, China
| | - Yong Liu
- Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science CenterXi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong UniversityBeijing, China
| |
Collapse
|
8
|
Gugliandolo A, Rajan TS, Scionti D, Diomede F, Bramanti P, Mazzon E, Trubiani O. Reprogramming of Oncogene Expression in Gingival Mesenchymal Stem Cells Following Long-Term Culture In Vitro. Cell Reprogram 2017; 19:159-170. [DOI: 10.1089/cell.2016.0056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
| | | | | | - Francesca Diomede
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| | | | | | - Oriana Trubiani
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| |
Collapse
|
9
|
Selvaraj P, Xiao L, Lee C, Murthy SRK, Cawley NX, Lane M, Merchenthaler I, Ahn S, Loh YP. Neurotrophic Factor-α1: A Key Wnt-β-Catenin Dependent Anti-Proliferation Factor and ERK-Sox9 Activated Inducer of Embryonic Neural Stem Cell Differentiation to Astrocytes in Neurodevelopment. Stem Cells 2016; 35:557-571. [PMID: 27709799 DOI: 10.1002/stem.2511] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/08/2016] [Accepted: 09/06/2016] [Indexed: 12/31/2022]
Abstract
Embryonic neurodevelopment involves inhibition of proliferation of multipotent neural stem cells (NSCs) followed by differentiation into neurons, astrocytes and oligodendrocytes to form the brain. We have identified a new neurotrophic factor, NF-α1, which inhibits proliferation and promotes differentiation of NSC/progenitors derived from E13.5 mouse cortex. Inhibition of proliferation of these cells was mediated through negatively regulating the Wnt pathway and decreasing β-catenin. NF-α1 induced differentiation of NSCs to astrocytes by enhancing Glial Fibrillary Acidic Protein (GFAP) expression through activating the ERK1/2-Sox9 signaling pathway. Cultured E13.5 cortical stem cells from NF-α1-knockout mice showed decreased astrocyte numbers compared to wild-type mice, which was rescued by treatment with NF-α1. In vivo, immunocytochemistry of brain sections and Western blot analysis of neocortex of mice showed a gradual increase of NF-α1 expression from E14.5 to P1 and a surge of GFAP expression at P1, the time of increase in astrogenesis. Importantly, NF-α1-Knockout mice showed ∼49% fewer GFAP positive astrocytes in the neocortex compared to WT mice at P1. Thus, NF-α1 is critical for regulating antiproliferation and cell fate determination, through differentiating embryonic stem cells to GFAP-positive astrocytes for normal neurodevelopment. Stem Cells 2017;35:557-571.
Collapse
Affiliation(s)
| | - Lan Xiao
- Section on Cellular Neurobiology, Bethesda, Maryland, USA
| | - Cheol Lee
- Unit on Developmental Neurogenetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Niamh X Cawley
- Section on Cellular Neurobiology, Bethesda, Maryland, USA
| | - Malcolm Lane
- Department of Epidemiology and Public Health and Anatomy and Neurobiology, University of Maryland, Baltimore, Maryland, USA
| | - Istvan Merchenthaler
- Department of Epidemiology and Public Health and Anatomy and Neurobiology, University of Maryland, Baltimore, Maryland, USA
| | - Sohyun Ahn
- Unit on Developmental Neurogenetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Y Peng Loh
- Section on Cellular Neurobiology, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Effect of controlled release of brain-derived neurotrophic factor and neurotrophin-3 from collagen gel on neural stem cells. Neuroreport 2016; 27:116-23. [PMID: 26656937 DOI: 10.1097/wnr.0000000000000507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study aimed to examine the effect of controlled release of brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) from collagen gel on rat neural stem cells (NSCs). With three groups of collagen gel, BDNF/collagen gel, and NT-3/collagen gel as controls, BDNF and NT-3 were tested in the BDNF-NT-3/collagen gel group at different time points. The enzyme-linked immunosorbent assay results showed that BDNF and NT-3 were steadily released from collagen gels for 10 days. The cell viability test and the bromodeoxyuridine incorporation assay showed that BDNF-NT-3/collagen gel supported the survival and proliferation of NSCs. The results also showed that the length of processes was markedly longer and differentiation percentage from NSCs into neurons was much higher in the BDNF-NT-3/collagen gel group than those in the collagen gel, BDNF/collagen gel, and NT-3/collagen gel groups. These findings suggest that BDNF-NT-3/collagen gel could significantly improve the ability of NSCs proliferation and differentiation.
Collapse
|
11
|
Lin XY, Lai BQ, Zeng X, Che MT, Ling EA, Wu W, Zeng YS. Cell Transplantation and Neuroengineering Approach for Spinal Cord Injury Treatment: A Summary of Current Laboratory Findings and Review of Literature. Cell Transplant 2016; 25:1425-38. [DOI: 10.3727/096368916x690836] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Spinal cord injury (SCI) can cause severe traumatic injury to the central nervous system (CNS). Current therapeutic effects achieved for SCI in clinical medicine show that there is still a long way to go to reach the desired goal of full or significant functional recovery. In basic medical research, however, cell transplantation, gene therapy, application of cytokines, and biomaterial scaffolds have been widely used and investigated as treatments for SCI. All of these strategies when used separately would help rebuild, to some extent, the neural circuits in the lesion area of the spinal cord. In light of this, it is generally accepted that a combined treatment may be a more effective strategy. This review focuses primarily on our recent series of work on transplantation of Schwann cells and adult stem cells, and transplantation of stem cell-derived neural network scaffolds with functional synapses. Arising from this, an artificial neural network (an exogenous neuronal relay) has been designed and fabricated by us—a biomaterial scaffold implanted with Schwann cells modified by the neurotrophin-3 (NT-3) gene and adult stem cells modified with the TrkC (receptor of NT-3) gene. More importantly, experimental evidence suggests that the novel artificial network can integrate with the host tissue and serve as an exogenous neuronal relay for signal transfer and functional improvement of SCI.
Collapse
Affiliation(s)
- Xin-Yi Lin
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Ming-Tian Che
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wutian Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
- Jinan University–Hong Kong University Joint Laboratory, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, China
- Co-innovation Center of Neuroregeneration, Nantong, Jiangsu, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
12
|
Interferon-β Inhibits Neurotrophin 3 Signalling and Pro-Survival Activity by Upregulating the Expression of Truncated TrkC-T1 Receptor. Mol Neurobiol 2016; 54:1825-1843. [PMID: 26887385 DOI: 10.1007/s12035-016-9789-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/11/2016] [Indexed: 12/17/2022]
Abstract
Although clinically useful for the treatment of various diseases, type I interferons (IFNs) have been implicated as causative factors of a number of neuroinflammatory disorders characterized by neuronal damage and altered CNS functions. As neurotrophin 3 (NT3) plays a critical role in neuroprotection, we examined the effects of IFN-β on the signalling and functional activity of the NT3/TrkC system. We found that prolonged exposure of differentiated human SH-SY5Y neuroblastoma cells to IFN-β impaired the ability of NT3 to induce transphosphorylation of the full-length TrkC receptor (TrkC-FL) and the phosphorylation of downstream signalling molecules, including PLCγ1, Akt, GSK-3β and ERK1/2. NT3 was effective in protecting the cells against apoptosis triggered by serum withdrawal or thapsigargin but not IFN-β. Prolonged exposure to the cytokine had little effects on TrkC-FL levels but markedly enhanced the messenger RNA (mRNA) and protein levels of the truncated isoform TrkC-T1, a dominant-negative receptor that inhibits TrkC-FL activity. Cell depletion of TrkC-T1 by small interfering RNA (siRNA) treatment enhanced NT3 signalling through TrkC-FL and allowed the neurotrophin to counteract IFN-β-induced apoptosis. Furthermore, the upregulation of TrkC-T1 by IFN-β was associated with the inhibition of NT3-induced recruitment of the scaffold protein tamalin to TrkC-T1 and tamalin tyrosine phosphorylation. These data indicate that IFN-β exerts a negative control on NT3 pro-survival signalling through a novel mechanism involving the upregulation of TrkC-T1.
Collapse
|
13
|
Kim KT, Kim HJ, Cho DC, Bae JS, Park SW. Substance P stimulates proliferation of spinal neural stem cells in spinal cord injury via the mitogen-activated protein kinase signaling pathway. Spine J 2015; 15:2055-65. [PMID: 25921821 DOI: 10.1016/j.spinee.2015.04.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/20/2015] [Accepted: 04/20/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Substance P (SP) is a neuropeptide that can influence neural stem/progenitor cell (NSPC) proliferation and neurogenesis in the brain. However, we could not find any experimental study that investigates SP action in the spinal cord. PURPOSE The aims of our study were to investigate the potential of the neuropeptide SP in promoting the proliferation of spinal cord-derived NSPCs (SC-NSPCs) after spinal cord injury (SCI) and to clarify the roles of the mitogen-activated protein (MAP) kinase signaling pathway in the process. STUDY DESIGN This is a randomized animal study. METHODS The SC-NSPCs were suspended in 100 μL of a neurobasal medium containing SP (binds neurokinin-1 receptor [NK1R]) or L-703,606 (NK1R antagonist) and cultured in a 96-well plate for 5 days. A cell proliferation assay was performed using a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. A cord clipping method was used for the SCI model. Substance P and the NK1R antagonist (L-703,606) were infused intrathecally in SCI and sham models. Neural stem/progenitor cell proliferation was evaluated with immunostaining for bromodeoxyuridine (BrdU) and the immature neural marker nestin. An immunoblotting method was used for evaluating the MAP kinase signaling protein that contains extracellular signal-regulated kinases (ERKs and p38) and β-actin as the control group. RESULTS In vitro, SP (0.01-10 μmol/L) increased the proliferation of cultured SC-NSPCs, with a peak increase of 35±2% at the 0.1 μmol/L concentration. Substance P of 0.1 μmol/L continuously increased SC-NSPC proliferation from 6 hours to 5 days, whereas the proliferation decreased from 18% to 98% with L-703,606 (1-10 μM). Intrathecal infusion of SP (1 μmol/L) for 7 days significantly increased the number of proliferating NPSCs (cells positive for both BrdU and nestin) in the spinal cord (by 120±17%, p<.05) in adult rats, but infusion of L-703,606 (10 μmol/L) significantly decreased the post-SCI induction of NPSC proliferation in the spinal cord (by 87±4%). Also, SP stimulates proliferation of SC-NSPCs via the MAP kinase signaling pathway, especially the phosphorylated ERK and phosphorylated p38 proteins. The phosphorylated ERK and phosphorylated p38 protein levels increased with SP (0.1 μmol/L, p<.05). CONCLUSIONS These data indicate that SP can promote proliferation of SC-NSPCs in SCI and normal conditions and have important roles in neuronal regeneration after SCI. Also, ERKs and p38 MAP kinases are important signaling proteins in this process.
Collapse
Affiliation(s)
- Kyoung-Tae Kim
- Department of Neurosurgery, Kyungpook National University Hospital, 50 Samduk-2-ga, Jung-gu, Daegu 700-721, Republic of Korea.
| | - Hye-Jeong Kim
- Department of Neurosurgery, Kyungpook National University Hospital, 50 Samduk-2-ga, Jung-gu, Daegu 700-721, Republic of Korea
| | - Dae-Chul Cho
- Department of Neurosurgery, Kyungpook National University Hospital, 50 Samduk-2-ga, Jung-gu, Daegu 700-721, Republic of Korea
| | - Jae-Sung Bae
- Department of Physiology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 700-842, Republic of Korea
| | - Seung-Won Park
- Department of Neurosurgery, College of Medicine, Chung-Ang University Hospital, 224-1 Heukseok dong, Dongjak-gu, Seoul 156-755, Republic of Korea
| |
Collapse
|
14
|
Nakano R, Edamura K, Nakayama T, Teshima K, Asano K, Narita T, Okabayashi K, Sugiya H. Differentiation of canine bone marrow stromal cells into voltage- and glutamate-responsive neuron-like cells by basic fibroblast growth factor. J Vet Med Sci 2014; 77:27-35. [PMID: 25284120 PMCID: PMC4349535 DOI: 10.1292/jvms.14-0284] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We investigated the in vitro differentiation of canine bone marrow stromal cells (BMSCs) into voltage- and glutamate-responsive neuron-like cells. BMSCs were obtained from the bone marrow of healthy beagle dogs. Canine BMSCs were incubated with the basal medium for neurons containing recombinant human basic fibroblast growth factor (bFGF; 100 ng/ml). The viability of the bFGF-treated cells was assessed by a trypan blue exclusion assay, and the morphology was monitored. Real-time RT-PCR was performed to evaluate mRNA expression of neuronal, neural stem cell and glial markers. Western blotting and immunocytochemical analysis for the neuronal markers were performed to evaluate the protein expression and localization. The Ca(2+) mobilization of the cells was evaluated using the Ca(2+) indicator Fluo3 to monitor Ca(2+) influx. To investigate the mechanism of bFGF-induced neuronal differentiation, the fibroblast growth factor receptor inhibitor, the phosphoinositide 3-kinase inhibitor or the Akt inhibitor was tested. The bFGF treatment resulted in the maintenance of the viability of canine BMSCs for 10 days, in the expression of neuronal marker mRNAs and proteins and in the manifestation of neuron-like morphology. Furthermore, in the bFGF-treated BMSCs, a high concentration of KCl and L-glutamate induced an increase in intracellular Ca(2+) levels. Each inhibitor significantly attenuated the bFGF-induced increase in neuronal marker mRNA expression. These results suggest that bFGF contributes to the differentiation of canine BMSCs into voltage- and glutamate-responsive neuron-like cells and may lead to the development of new cell-based treatments for neuronal diseases.
Collapse
Affiliation(s)
- Rei Nakano
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, 1866 Kameino, Fujisawa, Kanagawa 252-0880, Japan
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Tang S, Liao X, Shi B, Qu Y, Huang Z, Lin Q, Guo X, Pei F. The effects of controlled release of neurotrophin-3 from PCLA scaffolds on the survival and neuronal differentiation of transplanted neural stem cells in a rat spinal cord injury model. PLoS One 2014; 9:e107517. [PMID: 25215612 PMCID: PMC4162607 DOI: 10.1371/journal.pone.0107517] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 08/18/2014] [Indexed: 02/05/2023] Open
Abstract
Neural stem cells (NSCs) have emerged as a potential source for cell replacement therapy following spinal cord injury (SCI). However, poor survival and low neuronal differentiation remain major obstacles to the use of NSCs. Biomaterials with neurotrophic factors are promising strategies for promoting the proliferation and differentiation of NSCs. Silk fibroin (SF) matrices were demonstrated to successfully deliver growth factors and preserve their potency. In this study, by incorporating NT-3 into a SF coating, we successfully developed NT-3-immobilized scaffolds (membranes and conduits). Sustained release of bioactive NT-3 from the conduits for up to 8 weeks was achieved. Cell viability was confirmed using live/dead staining after 14 days in culture. The efficacy of the immobilized NT-3 was confirmed by assessing NSC neuronal differentiation in vitro. NSC neuronal differentiation was 55.2 ± 4.1% on the NT-3-immobilized membranes, which was significantly higher than that on the NT-3 free membrane. Furthermore, 8 weeks after the NSCs were seeded into conduits and implanted in rats with a transected SCI, the conduit+NT-3+NSCs group achieved higher NSC survival (75.8 ± 15.1%) and neuronal differentiation (21.5 ± 5.2%) compared with the conduit+NSCs group. The animals that received the conduit+NT-3+NSCs treatment also showed improved functional outcomes, as well as increased axonal regeneration. These results indicate the feasibility of fabricating NT-3-immobilized scaffolds using the adsorption of NT-3/SF coating method, as well as the potential of these scaffolds to induce SCI repair by promoting survival and neuronal differentiation of transplanted NSCs.
Collapse
Affiliation(s)
- Shuo Tang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Liao
- Department of Pain Medicine, Shenzhen Nanshan Hospital, Shenzhen, China
| | - Bo Shi
- Department of Orthopaedics, Mianyang Center Hospital, Mianyang, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeyu Huang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Lin
- Department of Orthopaedics, Guangdong hospital of traditional Chinese medicine, Guangzhou, China
- * E-mail: (QL); (XDG); (FXP)
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (QL); (XDG); (FXP)
| | - Fuxing Pei
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
- * E-mail: (QL); (XDG); (FXP)
| |
Collapse
|
16
|
Shaheen BS, Bakir M, Jain S. Corneal nerves in health and disease. Surv Ophthalmol 2014; 59:263-85. [PMID: 24461367 PMCID: PMC4004679 DOI: 10.1016/j.survophthal.2013.09.002] [Citation(s) in RCA: 338] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 08/23/2013] [Accepted: 09/03/2013] [Indexed: 12/14/2022]
Abstract
Corneal nerves are responsible for the sensations of touch, pain, and temperature and play an important role in the blink reflex, wound healing, and tear production and secretion. Corneal nerve dysfunction is a frequent feature of diseases that cause opacities and result in corneal blindness. Corneal opacities rank as the second most frequent cause of blindness. Technological advances in in vivo corneal nerve imaging, such as optical coherence tomography and confocal scanning, have generated new knowledge regarding the phenomenological events that occur during reinnervation of the cornea following disease, injury, or surgery. The recent availability of transgenic neurofluorescent murine models has stimulated the search for molecular modulators of corneal nerve regeneration. New evidence suggests that neuroregenerative and inflammatory pathways in the cornea are intertwined. Evidence-based treatment of neurotrophic corneal diseases includes using neuroregenerative (blood component-based and neurotrophic factors), neuroprotective, and ensconcing (bandage contact lens and amniotic membrane) strategies and avoiding anti-inflammatory therapies, such as cyclosporine and corticosteroids.
Collapse
Affiliation(s)
- Brittany Simmons Shaheen
- Corneal Neurobiology Laboratory, Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - May Bakir
- Corneal Neurobiology Laboratory, Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Sandeep Jain
- Corneal Neurobiology Laboratory, Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
17
|
Ghoroghi FM, Hejazian LB, Esmaielzade B, Dodel M, Roudbari M, Nobakht M. Evaluation of the Effect of NT-3 and Biodegradable Poly-L-lactic Acid Nanofiber Scaffolds on Differentiation of Rat Hair Follicle Stem Cells into Neural Cells In Vitro. J Mol Neurosci 2013; 51:318-327. [PMID: 23959422 DOI: 10.1007/s12031-013-0073-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 07/10/2013] [Indexed: 10/26/2022]
Abstract
Recent improvement in neuroscience has led to new strategies in neural repair. Hair follicle stem cells are high promising source of accessible, active, and pluripotent adult stem cells. They have high affinity to differentiate to neurons. Aside from using cell-scaffold combinations for implantation, scaffolds can provide a suitable microenvironment for cell proliferation, migration, and differentiation. NT-3 is the most interesting neurotrophic factors being an important regulator of neural survival and differentiation. Since treatment duration in neural repair is very important, this study aims to evaluate the effect of NT-3 and poly-L-lactic acid (PLLA) on differentiation time of bulge stem cells of rat hair follicle to neural-like cells. HFSCs of rat whisker was isolated and cultured on PLLA and differentiated with 10 ng/mL NT-3. Biological features of cultured cells were evaluated with immunocytochemistry and flowcytometry methods by using CD34, nestin, and βІІІ-tubulin markers. For cell viability and morphological assessment, MTT assay and SEM were performed. Our results showed that bulge stem cells of hair follicle can express CD34 and Nestin before differentiation. By using NT-3 during differentiation process, the cells showed positive reaction to βІІІ-tubulin antibody. MTT results demonstrated that PLLA significantly increased cell viability. Finally, HFSCs adhesion was confirmed by SEM results. The results indicate that 10 ng/mL NT-3 and PLLA have significant effect on differentiation time of rat HFSCs to neural cells even in 10 days.
Collapse
|
18
|
Oliveira SLB, Pillat MM, Cheffer A, Lameu C, Schwindt TT, Ulrich H. Functions of neurotrophins and growth factors in neurogenesis and brain repair. Cytometry A 2012; 83:76-89. [PMID: 23044513 DOI: 10.1002/cyto.a.22161] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Revised: 07/23/2012] [Accepted: 07/31/2012] [Indexed: 12/21/2022]
Abstract
The identification and isolation of multipotent neural stem and progenitor cells in the brain, giving rise to neurons, astrocytes, and oligodendrocytes initiated many studies in order to understand basic mechanisms of endogenous neurogenesis and repair mechanisms of the nervous system and to develop novel therapeutic strategies for cellular regeneration therapies in brain disease. A previous review (Trujillo et al., Cytometry A 2009;75:38-53) focused on the importance of extrinsic factors, especially neurotransmitters, for directing migration and neurogenesis in the developing and adult brain. Here, we extend our review discussing the effects of the principal growth and neurotrophic factors as well as their intracellular signal transduction on neurogenesis, fate determination and neuroprotective mechanisms. Many of these mechanisms have been elucidated by in vitro studies for which neural stem cells were isolated, grown as neurospheres, induced to neural differentiation under desired experimental conditions, and analyzed for embryonic, progenitor, and neural marker expression by flow and imaging cytometry techniques. The better understanding of neural stem cells proliferation and differentiation is crucial for any therapeutic intervention aiming at neural stem cell transplantation and recruitment of endogenous repair mechanisms.
Collapse
Affiliation(s)
- Sophia L B Oliveira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
19
|
Vaysse L, Labie C, Canolle B, Jozan S, Béduer A, Arnauduc F, Vieu C, Sol JC, Loubinoux I. Adult human progenitor cells from the temporal lobe: another source of neuronal cells. Brain Inj 2012; 26:1636-45. [PMID: 22823462 DOI: 10.3109/02699052.2012.700084] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES In the adult human brain, neurogenesis occurs in the SVZ and the dentate gyrus of the hippocampus, but it is still unclear whether persistent neural progenitor/stem cells are also present in other brain areas. The present work studies the possibility of obtaining neural progenitor/stem cells from the temporal lobe and investigates their potential to differentiate into neuronal cells. METHODS Human biopsies from the temporal lobe of epileptic patients were used to isolate potential neural progenitors. Differentiation was induced in the presence of different agents (NGF, NT3, RA) and immunocytochemistry was then performed for quantitative analysis. RESULTS It was shown that a significant number of cells in the temporal lobe are also capable of expansion and multi-potency. These cells can be amplified as neurospheres and have the potential to differentiate naturally in vitro into neurons, astrocytes and oligodendrocytes. Quantitative analyses show that the progenitor cells of the temporal lobe exhibit a better rate of neuronal differentiation in vitro than the cells from the SVZ, particularly in the presence of NGF. CONCLUSION This study indicates that neural progenitors are also present in the human temporal lobe. Studying them could be of great interest for cell therapy in neurological disorders.
Collapse
Affiliation(s)
- L Vaysse
- Inserm, Imagerie Cérébrale et Handicaps Neurologiques UMR 825, Toulouse, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kim J, Wong PKY. Targeting p38 mitogen-activated protein kinase signaling restores subventricular zone neural stem cells and corrects neuromotor deficits in Atm knockout mouse. Stem Cells Transl Med 2012. [PMID: 23197859 DOI: 10.5966/sctm.2011-0063] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Ataxia-telangiectasia (A-T) is a progressive degenerative disorder that results in major neurological disability. In A-T patients, necropsy has revealed atrophy of cerebellar cortical layers along with Purkinje and granular cell loss. We have previously identified an oxidative stress-mediated increase in phospho-p38 mitogen-activated protein kinase (MAPK) and the resultant downregulation of Bmi-1 and upregulation of p21 as key components of the mechanism causing defective proliferation of neural stem cells (NSCs) isolated from the subventricular zone (SVZ) of Atm(-/-) mice. However, the in vivo aspect of alteration in SVZ tissue and the functional significance of p38MAPK activation in NSCs for neuropathogenesis of ATM deficiency remain unknown. Here we show that the NSC population was abnormally decreased in the SVZ of 3-month-old Atm(-/-) mice; this decrease was accompanied by p38MAPK activation. However, after a 2-month treatment with the p38MAPK inhibitor SB203580, starting at 1 month old, Atm(-/-) mice showed restoration of normal levels of Bmi-1 and p21 with the rescue of NSC population in the SVZ. In addition, treated Atm(-/-) mice exhibited more Purkinje cells in the cerebellum. Most importantly, motor coordination of Atm(-/-) mice was significantly improved in the treatment group. Our results show for the first time in vivo evidence of depleted NSCs in the SVZ of Atm(-/-) mice and also demonstrate that pharmacologic inhibition of p38MAPK signaling has the potential to treat neurological defects of A-T. This study provides a promising approach targeting the oxidative stress-dependent p38 signaling pathway not only for A-T but also for other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jeesun Kim
- Department of Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957, USA
| | | |
Collapse
|
21
|
Yang SR, Rahman I, Trosko JE, Kang KS. Oxidative stress-induced biomarkers for stem cell-based chemical screening. Prev Med 2012; 54 Suppl:S42-9. [PMID: 22197760 DOI: 10.1016/j.ypmed.2011.11.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 11/30/2011] [Indexed: 12/23/2022]
Abstract
Stem cells have been considered for their potential in pharmaceutical research, as well as for stem cell-based therapy for many diseases. Despite the potential for their use, the challenge remains to examine the safety and efficacy of stem cells for their use in therapies. Recently, oxidative stress has been strongly implicated in the functional regulation of cell behavior of stem cells. Therefore, development of rapid and sensitive biomarkers, related to oxidative stress is of growing importance in stem cell-based therapies for treating various diseases. Since stem cells have been implicated as targets for carcinogenesis and might be the origin of "cancer stem cells", understanding of how oxidative stress-induced signaling, known to be involved in the carcinogenic process could lead to potential screening of cancer chemopreventive and chemotherapeutic agents. An evaluation of antioxidant states reducing equivalents like GSH and superoxide dismutase (SOD), as well as reactive oxygen species (ROS) and nitric oxide (NO) generation, can be effective markers in stem cell-based therapies. In addition, oxidative adducts, such as 4-hydroxynonenal, can be reliable markers to detect cellular changes during self-renewal and differentiation of stem cells. This review highlights the biomarker development to monitor oxidative stress response for stem cell-based chemical screening.
Collapse
Affiliation(s)
- Se-Ran Yang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
22
|
Niapour A, Karamali F, Nemati S, Taghipour Z, Mardani M, Nasr-Esfahani MH, Baharvand H. Cotransplantation of Human Embryonic Stem Cell-Derived Neural Progenitors and Schwann Cells in a Rat Spinal Cord Contusion Injury Model Elicits a Distinct Neurogenesis and Functional Recovery. Cell Transplant 2012; 21:827-843. [DOI: 10.3727/096368911x593163] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cotransplantation of neural progenitors (NPs) with Schwann cells (SCs) might be a way to overcome low rate of neuronal differentiation of NPs following transplantation in spinal cord injury (SCI) and the improvement of locomotor recovery. In this study, we initially generated NPs from human embryonic stem cells (hESCs) and investigated their potential for neuronal differentiation and functional recovery when cocultured with SCs in vitro and cotransplanted in a rat acute model of contused SCI. Cocultivation results revealed that the presence of SCs provided a consistent status for hESC-NPs and recharged their neural differentiation toward a predominantly neuronal fate. Following transplantation, a significant functional recovery was observed in all engrafted groups (NPs, SCs, NPs + SCs) relative to the vehicle and control groups. We also observed that animals receiving cotransplants established a better state as assessed with the BBB functional test. Immunohistofluorescence evaluation 5 weeks after transplantation showed invigorated neuronal differentiation and limited proliferation in the cotransplanted group when compared to the individual hESC-NP-grafted group. These findings have demonstrated that the cotransplantation of SCs with hESC-NPs could offer a synergistic effect, promoting neuronal differentiation and functional recovery.
Collapse
Affiliation(s)
- Ali Niapour
- Department of Cell and Molecular Biology, Cell Science Research Center, Royan Institute for Animal Biotechnology, ACECR, Isfahan, Iran
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
- Department of Anatomical Sciences, Ardebil University of Medical Science, Ardebil, Iran
| | - Fereshteh Karamali
- Department of Cell and Molecular Biology, Cell Science Research Center, Royan Institute for Animal Biotechnology, ACECR, Isfahan, Iran
| | - Shiva Nemati
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Taghipour
- Department of Cell and Molecular Biology, Cell Science Research Center, Royan Institute for Animal Biotechnology, ACECR, Isfahan, Iran
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Mohammad Mardani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cell and Molecular Biology, Cell Science Research Center, Royan Institute for Animal Biotechnology, ACECR, Isfahan, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| |
Collapse
|
23
|
Curtis KM, Gomez LA, Schiller PC. Rac1b regulates NT3-stimulated Mek-Erk signaling, directing marrow-isolated adult multilineage inducible (MIAMI) cells toward an early neuronal phenotype. Mol Cell Neurosci 2012; 49:138-48. [DOI: 10.1016/j.mcn.2011.10.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 10/18/2011] [Accepted: 10/19/2011] [Indexed: 12/25/2022] Open
|
24
|
TAKANO N, INOKUCHI Y, KURACHI M. Effects of Ethanol Extracts of Herbal Medicines on Dermatitis in an Atopic Dermatitis Mouse Model. YAKUGAKU ZASSHI 2011; 131:581-6. [DOI: 10.1248/yakushi.131.581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Norikazu TAKANO
- Materials Research & Development, R&D Laboratories, Self Medication Business, Taisho Pharmaceutical Co., Ltd
| | - Yuki INOKUCHI
- Materials Research & Development, R&D Laboratories, Self Medication Business, Taisho Pharmaceutical Co., Ltd
| | - Michio KURACHI
- Kampo & Herval, R&D Headquarters, Self Medication Business, Taisho Pharmaceutical Co., Ltd
| |
Collapse
|
25
|
Kang KS, Trosko JE. Stem cells in toxicology: fundamental biology and practical considerations. Toxicol Sci 2010; 120 Suppl 1:S269-89. [PMID: 21163910 DOI: 10.1093/toxsci/kfq370] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This "Commentary" has examined the use of human stem cells for detection of toxicities of physical, chemical, and biological toxins/toxicants in response to the challenge posed by the NRC Report, "Toxicity Testing in the 21st Century: A vision and Strategy." Before widespread application of the use of human embryonic, pluripotent, "iPS," or adult stem cells be considered, the basic characterization of stem cell biology should be undertaken. Because no in vitro system can mimic all factors that influence cells in vivo (individual genetic, gender, developmental, immunological and diurnal states; niche conditions; complex intercellular interactions between stem, progenitor, terminal differentiated cells, and the signaling from extracellular matrices, oxygen tensions, etc.), attempts should be made to use both embryonic and adult stem cells, grown in three dimension under "niche-like" conditions. Because many toxins and toxicants work by "epigenetic" mechanisms and that epigenetic mechanisms play important roles in regulating gene expression and in the pathogenesis of many human diseases, epigenetic toxicity must be incorporated in toxicity testing. Because modulation of gap junctional intercellular communication by epigenetic agents plays a major role in homeostatic regulation of both stem and progenitor cells in normal tissues, the modulation of this biological process by both endogenous and endogenous chemicals should be incorporated as an end point to monitor for potential toxicities or chemo-preventive attributes. In addition, modulation of quantity, as well as the quality, of stem cells should be considered as potential source of a chemical's toxic potential in affecting any stem cell-based pathology, such as cancer.
Collapse
Affiliation(s)
- Kyung-Sun Kang
- Adult Stem Cell Research Center, Laboratory for Stem Cell and Tumor Biology, Department of Veterinary Public Health, College of Veterinary Medicine, Seoul National University, Sillim-Dong, Seoul 151-742, Korea
| | | |
Collapse
|
26
|
Oh J, McCloskey MA, Blong CC, Bendickson L, Nilsen-Hamilton M, Sakaguchi DS. Astrocyte-derived interleukin-6 promotes specific neuronal differentiation of neural progenitor cells from adult hippocampus. J Neurosci Res 2010; 88:2798-809. [PMID: 20568291 DOI: 10.1002/jnr.22447] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The purpose of this study was to investigate the ability of astrocyte-derived factors to influence neural progenitor cell differentiation. We previously demonstrated that rat adult hippocampal progenitor cells (AHPCs) immunoreactive for the neuronal marker class III beta-tubulin (TUJ1) were significantly increased in the presence of astrocyte-derived soluble factors under noncontact coculture conditions. Using whole-cell patch-clamp analysis, we observed that the cocultured AHPCs displayed two prominent voltage-gated conductances, tetraethyl ammonium (TEA)-sensitive outward currents and fast transient inward currents. The outward and inward current densities of the cocultured AHPCs were approximately 2.5-fold and 1.7-fold greater, respectively, than those of cells cultured alone. These results suggest that astrocyte-derived soluble factors induce neuronal commitment of AHPCs. To investigate further the activity of a candidate neurogenic factor on AHPC differentiation, we cultured AHPCs in the presence or absence of purified rat recombinant interleukin-6 (IL-6). We also confirmed that the astrocytes used in this study produced IL-6 by ELISA and RT-qPCR. When AHPCs were cultured with IL-6 for 6-7 days, the TUJ1-immunoreactive AHPCs and the average length of TUJ1-immunoreactive neurites were significantly increased compared with the cells cultured without IL-6. Moreover, IL-6 increased the inward current density to an extent comparable to that of coculture with astrocytes, with no significant differences in the outward current density, apparent resting potential, or cell capacitance. These results suggest that astrocyte-derived IL-6 may facilitate AHPC neuronal differentiation. Our findings have important implications for understanding injury-induced neurogenesis and developing cell-based therapeutic strategies using neural progenitors.
Collapse
Affiliation(s)
- Jisun Oh
- Neuroscience Program, Iowa State University, Ames, Iowa 50011, USA
| | | | | | | | | | | |
Collapse
|
27
|
Rafalski VA, Brunet A. Energy metabolism in adult neural stem cell fate. Prog Neurobiol 2010; 93:182-203. [PMID: 21056618 DOI: 10.1016/j.pneurobio.2010.10.007] [Citation(s) in RCA: 222] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 10/20/2010] [Accepted: 10/28/2010] [Indexed: 12/26/2022]
Abstract
The adult mammalian brain contains a population of neural stem cells that can give rise to neurons, astrocytes, and oligodendrocytes and are thought to be involved in certain forms of memory, behavior, and brain injury repair. Neural stem cell properties, such as self-renewal and multipotency, are modulated by both cell-intrinsic and cell-extrinsic factors. Emerging evidence suggests that energy metabolism is an important regulator of neural stem cell function. Molecules and signaling pathways that sense and influence energy metabolism, including insulin/insulin-like growth factor I (IGF-1)-FoxO and insulin/IGF-1-mTOR signaling, AMP-activated protein kinase (AMPK), SIRT1, and hypoxia-inducible factors, are now implicated in neural stem cell biology. Furthermore, these signaling modules are likely to cooperate with other pathways involved in stem cell maintenance and differentiation. This review summarizes the current understanding of how cellular and systemic energy metabolism regulate neural stem cell fate. The known consequences of dietary restriction, exercise, aging, and pathologies with deregulated energy metabolism for neural stem cells and their differentiated progeny will also be discussed. A better understanding of how neural stem cells are influenced by changes in energy availability will help unravel the complex nature of neural stem cell biology in both the normal and diseased state.
Collapse
|
28
|
Zahir T, Chen YF, MacDonald JF, Leipzig N, Tator CH, Shoichet MS. Neural stem/progenitor cells differentiate in vitro to neurons by the combined action of dibutyryl cAMP and interferon-gamma. Stem Cells Dev 2010; 18:1423-32. [PMID: 19355840 DOI: 10.1089/scd.2008.0412] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transplantation of neural stem/progenitor cells (NSPCs) is a promising strategy for repair of the diseased/injured central nervous system (CNS); however, controlling their differentiation remains a significant hurdle. This study is aimed at controlling differentiation and specifically at screening exogenous factors to direct NSPC differentiation into neurons in vitro. In this study, adult rat SVZ-derived NSPCs were treated with several factors and screened individually and in combination for changes in cellular morphology, neuronal marker expression, quantitative real-time qRT-PCR, and electrophysiological properties. These in vitro screens showed that of all the different treatments, dibutyryl cyclic AMP (dbcAMP) and interferon-gamma (IFN-gamma) enhanced neuronal differentiation most significantly compared to the 1% fetal bovine serum (FBS) controls. Importantly, the combined treatment of NSPCs with dbcAMP and IFN-gamma promoted greater neuronal differentiation as reflected by an increase in beta-III tubulin expression and morphological differentiation. Interestingly, the neurons that were generated from the NSPCs in vitro in the presence of dbcAMP and IFN-gamma, alone or in combination, responded to exogenous glutamate (Glu), but not gamma-aminobutyric acid (GABA), indicating that these neurons express glutamate receptors. These NSPC-derived neurons may be promising for neural regenerative strategies in the CNS.
Collapse
Affiliation(s)
- Tasneem Zahir
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | | | | | | | | | | |
Collapse
|
29
|
Kim J, Wong PKY. Loss of ATM impairs proliferation of neural stem cells through oxidative stress-mediated p38 MAPK signaling. Stem Cells 2010; 27:1987-98. [PMID: 19544430 DOI: 10.1002/stem.125] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ataxia-telangiectasia (A-T) is a genetic disorder caused by a mutation of the Atm gene, which controls DNA repair, cell cycling, and redox homeostasis. Even though oxidative stress has been implicated in the neurological anomalies in A-T, the effects of ATM loss on neural stem cell (NSC) survival has remained elusive. In this study, we investigated the effects of oxidative stress on NSC proliferation in an animal model for A-T neurodegeneration. We found that cultured subventricular zone neurosphere cells from Atm(-/-) mice show impaired proliferation, as well as intrinsic elevation of reactive oxygen species (ROS) levels, compared with those from Atm(+/+) mice. We also show that increasing the levels of ROS by H(2)O(2) treatment significantly reduces Atm(+/+) neurosphere formation and proliferation. In Atm(-/-) neurosphere cells, the Akt and Erk1/2 pathways are disrupted, together with enhanced activity of the p38 mitogen-activated protein kinase (MAPK). Treatment of these cells with the antioxidant N-acetyl-L-cysteine (NAC) or with a p38 MAPK inhibitor restores normal proliferation and reduced expression of p21(cip1) and p27(kip1) in the Atm(-/-) NSCs. These observations indicate that ATM plays a crucial role in NSC proliferation, by activating Akt and Erk1/2 pathways and by suppressing ROS-p38 MAPK signaling. Together, our results suggest that p38 MAPK signaling acts as a negative regulator of NSC proliferation in response to oxidative stress. These findings suggest a potential mechanism for neuronal cell loss as a result of oxidative stress in NSCs in progressive neurodegenerative diseases such as A-T.
Collapse
Affiliation(s)
- Jeesun Kim
- Department of Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Smithville, Texas 78957, USA
| | | |
Collapse
|
30
|
Li X, Yang Z, Zhang A. The effect of neurotrophin-3/chitosan carriers on the proliferation and differentiation of neural stem cells. Biomaterials 2009; 30:4978-85. [PMID: 19539985 DOI: 10.1016/j.biomaterials.2009.05.047] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 05/21/2009] [Indexed: 01/12/2023]
Abstract
In this study, the behavior of neural stem cells from the newborn rat spinal cord was compared at neurosphere level after the addition of neurotrophin-3 (NT-3) once or daily, blank chitosan carriers, or NT-3-chitosan carriers respectively. We found that NT-3 enhanced the viability and differentiation of neural stem cells, but as NT-3 has an extremely short half-life at 37 degrees C, in order to maintain the NT-3-mediated proliferation and differentiation effects on neural stem cells, NT-3 needed to be added to the medium every 24 h. However, NT-3-chitosan carriers dramatically increase the differentiation percentage of neural stem cells into neurons, which includes GABAergic and as cholinergic neurons. Although blank chitosan carriers also showed good biocompatibility to the neural stem cells, they induced the differentiation of these cells into neurons at a much lower percentage than the daily addition of NT-3 or the NT-3-chitosan carriers. Our results suggest that NT-3-chitosan carriers may not only maintain the viability of neural stem cells and increase their differentiation percentage into neurons, but also reduce the amount of NT-3 required for the survival and differentiation of these cells. These results may provide an experimental basis for the maximum replacement of dead neurons by neural stem cell transplant after spinal cord injury (SCI).
Collapse
Affiliation(s)
- Xiaoguang Li
- Beijing Institute for Neuroscience, Capital Medical University, Beijing 100069, China.
| | | | | |
Collapse
|
31
|
Willerth SM, Sakiyama-Elbert SE. Kinetic analysis of neurotrophin-3-mediated differentiation of embryonic stem cells into neurons. Tissue Eng Part A 2009; 15:307-18. [PMID: 18800878 PMCID: PMC2750852 DOI: 10.1089/ten.tea.2008.0071] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 07/15/2008] [Indexed: 02/03/2023] Open
Abstract
The goal of this study was to develop a kinetic analysis that could predict the behavior of embryonic stem cell-derived neural progenitor cells (ESNPCs) in response to treatment with neurotrophin-3 (NT-3). Previous studies have shown that NT-3 activates the mitogen-activated protein (MAP) kinase cascade in embryonic stem cells and promotes differentiation of ESNPCs into neurons. MAP kinase activation after NT-3 stimulation was confirmed experimentally, and a kinetic analysis was developed using rate constants obtained from the literature. Concentrations of select signaling components were estimated for ESNPCs using real-time reverse transcription polymerase chain reaction by comparing mRNA levels to those of cell types with known protein concentrations. This assumption was validated using Western blots, and incorporated into the analysis. This analysis was used to predict the minimum NT-3 concentration necessary to promote neuronal differentiation of ESNPCs based on the activation of MAP kinase. These predictions were then tested experimentally to confirm the validity of the analysis. Finally, expression of the transcription factor mammalian achate schute homolog 1 and beta-tubulin III (an early neuronal marker) was examined in response to the different NT-3 doses to confirm the link between MAP kinase activation and neuronal differentiation. Overall, this study provides insight into the kinetics of the intracellular processes that promote ESNPC differentiation to neurons.
Collapse
Affiliation(s)
| | - Shelly E. Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
- Center for Materials Innovation, Washington University, St. Louis, Missouri
| |
Collapse
|
32
|
Misumi S, Kim TS, Jung CG, Masuda T, Urakawa S, Isobe Y, Furuyama F, Nishino H, Hida H. Enhanced neurogenesis from neural progenitor cells with G1/S-phase cell cycle arrest is mediated by transforming growth factor beta1. Eur J Neurosci 2008; 28:1049-59. [PMID: 18783370 DOI: 10.1111/j.1460-9568.2008.06420.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have previously demonstrated that a G1/S-phase cell cycle blocker, deferoxamine (DFO), increased the number of new neurons from rat neurosphere cultures, which correlated with prolonged expression of cyclin-dependent kinase (cdk) inhibitor p27(kip1) [H. J. Kim et al. (2006)Brain Research, 1092, 1-15]. The present study focuses on neuronal differentiation mechanisms following treatment of neural stem/progenitor cells (NPCs) with a G1/S-phase cell cycle blocker. The addition of DFO (0.5 mm) or aphidicolin (Aph) (1.5 microm) to neurospheres for 8 h, followed by 3 days of differentiation, resulted in an increased number of neurons and neurite outgrowth. DFO induced enhanced expression of transforming growth factor (TGF)-beta1 and cdk5 at 24 h after differentiation, whereas Aph only increased TGF-beta1 expression. DFO-induced neurogenesis and neurite outgrowth were attenuated by administration of a cdk5 inhibitor, roscovitine, suggesting that the neurogenic mechanisms differ between DFO and Aph. TGF-beta1 (10 ng/mL) did not increase neurite outgrowth but rather the number of beta-tubulin III-positive cells, which was accompanied by enhanced p27(kip1) mRNA expression. In addition, TGF-beta receptor type II expression was observed in nestin-positive NPCs. Results indicated that DFO-induced TGF-beta1 signaling activated smad3 translocation from the cytoplasm to the nucleus. In contrast, TGF-beta1 signaling inhibition, via a TGF-beta receptor type I inhibitor (SB-505124), resulted in decreased DFO-induced neurogenesis, in conjunction with decreased p27(kip1) protein expression and smad3 translocation to the nucleus. These results suggest that cell cycle arrest during G1/S-phase induces TGF-beta1 expression. This, in turn, prompts enhanced neuronal differentiation via smad3 translocation to the nucleus and subsequent p27(kip1) activation in NPCs.
Collapse
Affiliation(s)
- Sachiyo Misumi
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya 467-8601, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lu H, Li M, Song T, Qian Y, Xiao X, Chen X, Zhang P, Feng X, Parker T, Liu Y. Retrovirus delivered neurotrophin-3 promotes survival, proliferation and neuronal differentiation of human fetal neural stem cells in vitro. Brain Res Bull 2008; 77:158-64. [PMID: 19875351 DOI: 10.1016/j.brainresbull.2008.02.037] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Revised: 02/10/2008] [Accepted: 02/18/2008] [Indexed: 12/21/2022]
Abstract
Poor survival and insufficient neuronal differentiation are the main obstacles to neural stem cell (NSC) transplantation therapy. Genetic modification of NSCs with neurotrophins is considered a promising approach to overcome these difficulties. In this study, the effects on survival, proliferation and neuronal differentiation of human fetal NSCs (hfNSCs) were observed after infection by a neurotrophin-3 (NT-3) recombinant retrovirus. The hfNSCs, from 12-week human fetal brains formed neurospheres, expressed the stem cell marker nestin and differentiated into the three main cell types of the nervous system. NT-3 recombinant retrovirus (Retro-NT-3) infected hfNSCs efficiently expressed NT-3 gene for at least 8 weeks, presented an accelerated proliferation, and therefore produced an increased number of neurospheres and after differentiation in vitro, contained a higher percentage of neuronal cells. Eight weeks after infection, 37.9+/-4.2% of hfNSCs in the Retro-NT-3 infection group expressed the neuronal marker, this was significantly higher than the control and mock infection groups. NT-3 transduced hfNSCs also displayed longer protruding neurites compared with other groups. Combined these results demonstrate that NT-3 modification promote the survival/proliferation, neuronal differentiation and growth of neurites of hfNSCs in vitro. This study proposes recombinant retrovirus mediated NT-3 modification may provide a promising means to resolve the poor survival and insufficient neuronal differentiation of NSCs.
Collapse
Affiliation(s)
- Haixia Lu
- Institute of Neurobiology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kim SJ, Son TG, Park HR, Park M, Kim MS, Kim HS, Chung HY, Mattson MP, Lee J. Curcumin stimulates proliferation of embryonic neural progenitor cells and neurogenesis in the adult hippocampus. J Biol Chem 2008; 283:14497-505. [PMID: 18362141 DOI: 10.1074/jbc.m708373200] [Citation(s) in RCA: 269] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Curcumin is a natural phenolic component of yellow curry spice, which is used in some cultures for the treatment of diseases associated with oxidative stress and inflammation. Curcumin has been reported to be capable of preventing the death of neurons in animal models of neurodegenerative disorders, but its possible effects on developmental and adult neuroplasticity are unknown. In the present study, we investigated the effects of curcumin on mouse multi-potent neural progenitor cells (NPC) and adult hippocampal neurogenesis. Curcumin exerted biphasic effects on cultured NPC; low concentrations stimulated cell proliferation, whereas high concentrations were cytotoxic. Curcumin activated extracellular signal-regulated kinases (ERKs) and p38 kinases, cellular signal transduction pathways known to be involved in the regulation of neuronal plasticity and stress responses. Inhibitors of ERKs and p38 kinases effectively blocked the mitogenic effect of curcumin in NPC. Administration of curcumin to adult mice resulted in a significant increase in the number of newly generated cells in the dentate gyrus of hippocampus, indicating that curcumin enhances adult hippocampal neurogenesis. Our findings suggest that curcumin can stimulate developmental and adult hippocampal neurogenesis, and a biological activity that may enhance neural plasticity and repair.
Collapse
Affiliation(s)
- So Jung Kim
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug Development, Longevity Life Science and Technology Institutes, Pusan National University, Geumjeong-Gu, Busan, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|