1
|
Disse P, Aymanns I, Ritter N, Peischard S, Korn L, Wiendl H, Pawlowski M, Kovac S, Meuth SG, Budde T, Strutz-Seebohm N, Wünsch B, Seebohm G. A novel NMDA receptor test model based on hiPSC-derived neural cells. Biol Chem 2023; 404:267-277. [PMID: 36630596 DOI: 10.1515/hsz-2022-0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
N-Methyl-D-aspartate receptors (NMDARs) are central for learning and information processing in the brain. Dysfunction of NMDARs can play a key role in the pathogenesis of neurodegeneration and drug addiction. The development of selective NMDAR modulators represents a promising strategy to target these diseases. Among such modulating compounds are ifenprodil and its 3-benzazepine derivatives. Classically, the effects of these NMDAR modulators have been tested by techniques like two-electrode voltage clamp (TEVC), patch clamp, or fluorescence-based assays. However, testing their functional effects in complex human systems requires more advanced approaches. Here, we established a human induced pluripotent stem cell-derived (hiPSC-derived) neural cell system and proved its eligibility as a test system for investigating NMDAR modulators and pharmaceutical effects on human neurons.
Collapse
Affiliation(s)
- Paul Disse
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
- Chembion, GRK 2515, Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Isabel Aymanns
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Nadine Ritter
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
- Chembion, GRK 2515, Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Stefan Peischard
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Lisanne Korn
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Heinz Wiendl
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Matthias Pawlowski
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Stjepana Kovac
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Sven G Meuth
- Neurologische Klinik, Heinrich-Heine-Universität Düsseldorf, D-40225 Düsseldorf, Germany
| | - Thomas Budde
- Institut für Physiologie I, Westfälische Wilhelms-Universität, D-48149 Münster, Germany
| | - Nathalie Strutz-Seebohm
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische and Medizinische Chemie, Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Guiscard Seebohm
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| |
Collapse
|
2
|
Transition from Animal-Based to Human Induced Pluripotent Stem Cells (iPSCs)-Based Models of Neurodevelopmental Disorders: Opportunities and Challenges. Cells 2023; 12:cells12040538. [PMID: 36831205 PMCID: PMC9954744 DOI: 10.3390/cells12040538] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) arise from the disruption of highly coordinated mechanisms underlying brain development, which results in impaired sensory, motor and/or cognitive functions. Although rodent models have offered very relevant insights to the field, the translation of findings to clinics, particularly regarding therapeutic approaches for these diseases, remains challenging. Part of the explanation for this failure may be the genetic differences-some targets not being conserved between species-and, most importantly, the differences in regulation of gene expression. This prompts the use of human-derived models to study NDDS. The generation of human induced pluripotent stem cells (hIPSCs) added a new suitable alternative to overcome species limitations, allowing for the study of human neuronal development while maintaining the genetic background of the donor patient. Several hIPSC models of NDDs already proved their worth by mimicking several pathological phenotypes found in humans. In this review, we highlight the utility of hIPSCs to pave new paths for NDD research and development of new therapeutic tools, summarize the challenges and advances of hIPSC-culture and neuronal differentiation protocols and discuss the best way to take advantage of these models, illustrating this with examples of success for some NDDs.
Collapse
|
3
|
Zhang SY, Zhao J, Ni JJ, Li H, Quan ZZ, Qing H. Application and prospects of high-throughput screening for in vitro neurogenesis. World J Stem Cells 2022; 14:393-419. [PMID: 35949394 PMCID: PMC9244953 DOI: 10.4252/wjsc.v14.i6.393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/07/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, high-throughput screening (HTS) has made great contributions to new drug discovery. HTS technology is equipped with higher throughput, minimized platforms, more automated and computerized operating systems, more efficient and sensitive detection devices, and rapid data processing systems. At the same time, in vitro neurogenesis is gradually becoming important in establishing models to investigate the mechanisms of neural disease or developmental processes. However, challenges remain in generating more mature and functional neurons with specific subtypes and in establishing robust and standardized three-dimensional (3D) in vitro models with neural cells cultured in 3D matrices or organoids representing specific brain regions. Here, we review the applications of HTS technologies on in vitro neurogenesis, especially aiming at identifying the essential genes, chemical small molecules and adaptive microenvironments that hold great prospects for generating functional neurons or more reproductive and homogeneous 3D organoids. We also discuss the developmental tendency of HTS technology, e.g., so-called next-generation screening, which utilizes 3D organoid-based screening combined with microfluidic devices to narrow the gap between in vitro models and in vivo situations both physiologically and pathologically.
Collapse
Affiliation(s)
- Shu-Yuan Zhang
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Juan Zhao
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Jun-Jun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Zhen-Zhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
4
|
Cho Y, Baek J, Lee E, Im SG. Heparin-mediated electrostatic immobilization of bFGF via functional polymer films for enhanced self-renewal of human neural stem cells. J Mater Chem B 2021; 9:2084-2091. [PMID: 33595038 DOI: 10.1039/d0tb02799e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Preserving the self-renewal capability of undifferentiated human neural stem cells (hNSCs) is one of the crucial prerequisites for efficient hNSC-based regenerative medicine. Considering that basic fibroblast growth factor (bFGF) is one of the key contributing factors in maintaining the self-renewal property of hNSCs, the bioactivity and stability of bFGF in the hNSC culture should be regulated carefully. In this study, we developed a functional polymer film of poly(glycidyl methacrylate (GMA)-co-N,N-dimethylaminoethyl methacrylate (DMAEMA)) (coGD, or p(GMA-co-DMAEMA)) via initiated chemical vapor deposition (iCVD), which facilitated a stable, electrostatic adsorption of heparin and subsequent immobilization of bFGF. The bFGF-immobilized coGD surface substantially enhanced the proliferation rate and neurosphere forming ability of hNSCs compared to tissue culture plate (TCP). The expression of the stemness markers of hNSCs such as NESTIN and SOX-2 was also upregulated prominently on the coGD surface. Also, the hNSCs cultured on the coGD surface showed enhanced neurogenesis upon spontaneous differentiation. The immobilized bFGF on the coGD surface stimulated the expression of bFGF receptors and subsequently activated the mitogen-activated protein kinase (MAPK) pathway, attributed to the increase in self-renewal property of hNSCs. Our results indicate that the coGD surface allowed in situ heparin-mediated bFGF immobilization, which served as a robust platform to generate hNSC neurospheres with enhanced self-renewal and differentiation capabilities and thereby will prompt an advance in the field of therapeutics of neurodegenerative diseases.
Collapse
Affiliation(s)
- Younghak Cho
- Department of Chemical and Biomolecular Engineering & KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Daejeon 34141, Republic of Korea.
| | - Jieung Baek
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Eunjung Lee
- Department of Chemical and Biomolecular Engineering & KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Daejeon 34141, Republic of Korea.
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering & KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Daejeon 34141, Republic of Korea.
| |
Collapse
|
5
|
Gaggi G, Di Credico A, Izzicupo P, Iannetti G, Di Baldassarre A, Ghinassi B. Chemical and Biological Molecules Involved in Differentiation, Maturation, and Survival of Dopaminergic Neurons in Health and Parkinson's Disease: Physiological Aspects and Clinical Implications. Biomedicines 2021; 9:biomedicines9070754. [PMID: 34209807 PMCID: PMC8301385 DOI: 10.3390/biomedicines9070754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disease characterized by a specific and progressive loss of dopaminergic (DA) neurons and dopamine, causing motor dysfunctions and impaired movements. Unfortunately, available therapies can partially treat the motor symptoms, but they have no effect on non-motor features. In addition, the therapeutic effect reduces gradually, and the prolonged use of drugs leads to a significative increase in the number of adverse events. For these reasons, an alternative approach that allows the replacement or the improved survival of DA neurons is very appealing for the treatment of PD patients and recently the first human clinical trials for DA neurons replacement have been set up. Here, we review the role of chemical and biological molecules that are involved in the development, survival and differentiation of DA neurons. In particular, we review the chemical small molecules used to differentiate different type of stem cells into DA neurons with high efficiency; the role of microRNAs and long non-coding RNAs both in DA neurons development/survival as far as in the pathogenesis of PD; and, finally, we dissect the potential role of exosomes carrying biological molecules as treatment of PD.
Collapse
Affiliation(s)
- Giulia Gaggi
- Beth Israel Deaconess Medical Center, Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA;
| | - Andrea Di Credico
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| | - Pascal Izzicupo
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| | | | - Angela Di Baldassarre
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
- Correspondence:
| | - Barbara Ghinassi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| |
Collapse
|
6
|
Dermatan sulfate obtained from the Phallusia nigra marine organism is responsible for antioxidant activity and neuroprotection in the neuroblastoma-2A cell lineage. Int J Biol Macromol 2020; 164:1099-1111. [PMID: 32629049 DOI: 10.1016/j.ijbiomac.2020.06.285] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
Neurodegenerative diseases are characterized by progressive loss of neurons in the central nervous system (CNS). Several molecules play a role in mammalian CNS regeneration, including glycosaminoglycans (GAGs). GAGs are found in abundance in many marine invertebrates, such as ascidians that belong to the phylum Chordata, which show a high CNS regeneration capacity even in adulthood. Here, we investigated the roles of dermatan sulfate, a type of GAG that was obtained from the ascidian Phallusia nigra. We investigated the neuroprotective and antioxidant properties of Phallusia nigra dermatan sulfate (PnDS) after neurotoxic damage induced by the pesticide rotenone using the Neuro-2A cell lineage. Neuroprotection was observed through a mitochondrial activity analysis. A morphometric analysis revealed long unbranched neurites after incubation with PnDS and co-incubation with PnDS and rotenone. Furthermore, PnDS showed antioxidant activity that reduced reactive oxygen species (ROS) even in co-incubation with rotenone. The reduced ROS probably occurred because PnDS increased the activity of the antioxidant enzymes superoxide dismutase and catalase and improved total antioxidant capacity, which protected cells from damage, as observed through decreased levels of lipid peroxidation. These data suggest a neuroprotective and antioxidant role of PnDS even under neurodegenerative conditions caused by rotenone.
Collapse
|
7
|
Development and Differentiation of Midbrain Dopaminergic Neuron: From Bench to Bedside. Cells 2020; 9:cells9061489. [PMID: 32570916 PMCID: PMC7349799 DOI: 10.3390/cells9061489] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/29/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s Disease (PD) is a neurodegenerative disorder affecting the motor system. It is primarily due to substantial loss of midbrain dopamine (mDA) neurons in the substantia nigra pars compacta and to decreased innervation to the striatum. Although existing drug therapy available can relieve the symptoms in early-stage PD patients, it cannot reverse the pathogenic progression of PD. Thus, regenerating functional mDA neurons in PD patients may be a cure to the disease. The proof-of-principle clinical trials showed that human fetal graft-derived mDA neurons could restore the release of dopamine neurotransmitters, could reinnervate the striatum, and could alleviate clinical symptoms in PD patients. The invention of human-induced pluripotent stem cells (hiPSCs), autologous source of neural progenitors with less ethical consideration, and risk of graft rejection can now be generated in vitro. This advancement also prompts extensive research to decipher important developmental signaling in differentiation, which is key to successful in vitro production of functional mDA neurons and the enabler of mass manufacturing of the cells required for clinical applications. In this review, we summarize the biology and signaling involved in the development of mDA neurons and the current progress and methodology in driving efficient mDA neuron differentiation from pluripotent stem cells.
Collapse
|
8
|
Liu Y, Yu M, Jiang D. Downregulation of STAT1 induces the differentiation of neural stem cells through JNK pathway. Tissue Cell 2019; 61:61-66. [PMID: 31759408 DOI: 10.1016/j.tice.2019.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 01/13/2023]
Abstract
Neural stem cells (NSCs) generated neurons and glial cells. Thus, it is a preferable candidate to the cell replacement-based therapy against neural disorders. The signaling pathways that regulate differentiation of NSCs are widely studied. In the current study, we used in vitro culture system to elucidate the role of signal transducer and activator of transcription 1 (STAT1) in NSCs' differentiation. Downregulation of STAT1 inhibited the proliferation of NSCs. Meanwhile, we also found STAT1 regulation could control the differentiation of NSCs. More neurons and glia cells were generated from NSCs with STAT1 silencing. This process was mediated by the JNK/STAT1 signaling. STAT1 inhibitor promoted differentiation of NSCs. After transplantation, we observed more neurons generated from NSCs with shRNA-STAT1 treatment. Collectively, this work showed an efficient way to regulate neuronal differentiation of NSCs through regulating the STAT1 expression. This is likely to provide source and theoretical support to cell replacement based theory.
Collapse
Affiliation(s)
- Yigang Liu
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Min Yu
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Dudu Jiang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| |
Collapse
|
9
|
Patel DK, Lim KT. Biomimetic Polymer-Based Engineered Scaffolds for Improved Stem Cell Function. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2950. [PMID: 31514460 PMCID: PMC6766224 DOI: 10.3390/ma12182950] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022]
Abstract
Scaffolds are considered promising materials for tissue engineering applications due to their unique physiochemical properties. The high porosity and adequate mechanical properties of the scaffolds facilitate greater cell adhesion, proliferation, and differentiation. Stem cells are frequently applied in tissue engineering applications due to their excellent potential. It has been noted that cell functions are profoundly affected by the nature of the extracellular matrix (ECM). Naturally derived ECM contains the bioactive motif that also influences the immune response of the organism. The properties of polymer scaffolds mean they can resemble the native ECM and can regulate cellular responses. Various techniques such as electrospinning and 3D printing, among others, are frequently used to fabricate polymer scaffolds, and their cellular responses are different with each technique. Furthermore, enhanced cell viability, as well as the differentiation ability of stem cells on the surface of scaffolds, opens a fascinating approach to the formation of ECM-like environments for tissue engineering applications.
Collapse
Affiliation(s)
- Dinesh K Patel
- The Institute of Forest Science, Kangwon National University, Chuncheon-24341, Korea.
| | - Ki-Taek Lim
- Department of Biosystems Engineering, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon-24341, Korea.
| |
Collapse
|
10
|
Wu C, Xue LD, Su LW, Xie JL, Jiang H, Yu XJ, Liu HM. Magnesium promotes the viability and induces differentiation of neural stem cells both in vitro and in vivo. Neurol Res 2018; 41:208-215. [PMID: 30596346 DOI: 10.1080/01616412.2018.1544400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Neural stem cells (NSCs) are multipotent stem cells that generating various neural cells, including neurons, astrocytes and oligodendrocytes. This showed that NSCs is an ideal candidate in the application of neural disease treatment. In the current study, we established a simple and efficient method to promote the viability and induce the differentiation of NSCs by stimulating with magnesium. METHODS The proliferation and differentiation of NSCs was determined by MTT assay and immunostaining. The behavior alteration was measured by rotorod test and Morris water maze. RESULTS Magnesium enhanced proliferation in NSCs. The ratio of Nestin+, Ki67+ and GFAP+ progenitor cells was increased in the presence of magnesium. Besides, magnesium induced the glial differentiation instead of neuronal differentiation in NSCs. By contrast, transplantation of Mg2+-treated NSCs in vivo generated more neurons. In established PD models, transplantation of Mg2+-treated NSCs could improve the symptoms and recover the memory. CONCLUSION We established a simple and efficient way to promote the proliferation and induce the differentiation of NSCs. More importantly, this may also facilitate to develop a new method to neural disorder treatment.
Collapse
Affiliation(s)
- Chao Wu
- a Department of Neurosurgery , Tengzhou Central People's Hospital , Shandong , China
| | - Lan-De Xue
- b Department of Periodontology , Jinan Stomatological Hospital , Jinan , China
| | - Lin-Wang Su
- c Oral medicine , Liaocheng Peoples Hospital , Liaocheng , China
| | - Jian-Li Xie
- d Department of Prosthodontics , Jinan Stomatological Hospital , Jinan , China
| | - Huan Jiang
- e Department of Endodontics , Jinan Stomatological Hospital , Jinan , China
| | - Xi-Jiao Yu
- e Department of Endodontics , Jinan Stomatological Hospital , Jinan , China
| | - Hong-Mei Liu
- e Department of Endodontics , Jinan Stomatological Hospital , Jinan , China
| |
Collapse
|
11
|
Moriarty N, Cabré S, Alamilla V, Pandit A, Dowd E. Encapsulation of young donor age dopaminergic grafts in a GDNF-loaded collagen hydrogel further increases their survival, reinnervation, and functional efficacy after intrastriatal transplantation in hemi-Parkinsonian rats. Eur J Neurosci 2018; 49:487-496. [PMID: 30054941 DOI: 10.1111/ejn.14090] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/10/2018] [Accepted: 07/23/2018] [Indexed: 11/30/2022]
Abstract
Biomaterials have been shown to significantly improve the outcome of cellular reparative approaches for Parkinson's disease in experimental studies because of their ability to provide transplanted cells with a supportive microenvironment and shielding from the host immune system. However, given that the margin for improvement in such reparative therapies is considerable, further studies are required to fully investigate and harness the potential of biomaterials in this context. Given that several recent studies have demonstrated improved brain repair in Parkinsonian models when using dopaminergic grafts derived from younger foetal donors, we hypothesized that encapsulating these cells in a supportive biomaterial would further improve their reparative efficacy. Thus, this study aimed to determine the impact of a GDNF-loaded collagen hydrogel on the survival, reinnervation, and functional efficacy of dopaminergic neurons derived from young donors. To do so, hemi-Parkinsonian (6-hydroxydopamine-lesioned) rats received intrastriatal transplants of embryonic day 12 cells extracted from the rat ventral mesencephalon either alone, in a collagen hydrogel, with GDNF, or in a GDNF-loaded collagen hydrogel. Methamphetamine-induced rotational behaviour was assessed at three weekly intervals for a total of 12 weeks, after which rats were sacrificed for postmortem assessment of graft survival. We found that, following intrastriatal transplantation to the lesioned striatum, the GDNF-loaded collagen hydrogel significantly increased the survival (4-fold), reinnervation (5.4-fold), and functional efficacy of the embryonic day 12 dopaminergic neurons. In conclusion, this study further demonstrates the significant potential of biomaterial hydrogel scaffolds for cellular brain repair approaches in neurodegenerative diseases such as Parkinson's disease.
Collapse
Affiliation(s)
- Niamh Moriarty
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Sílvia Cabré
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Verónica Alamilla
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Abhay Pandit
- CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| |
Collapse
|
12
|
Magnesium lithospermate B promotes proliferation and differentiation of neural stem cells in vitro and enhances neurogenesis in vivo. Tissue Cell 2018; 53:8-14. [PMID: 30060831 DOI: 10.1016/j.tice.2018.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/21/2018] [Accepted: 05/22/2018] [Indexed: 11/22/2022]
Abstract
Multipotent neural stem cells could generate neurons and glial cells. Wide studies have been conducted to disclose the mechanism underlying neural stem cell differentiation and multiple factors have been identified in this field, one of which is bioactive components including natural herbal medicine. In this study, we found that magnesium lithospermate B is able to promote proliferation of neural stem cell in vitro. Besides, magnesium lithospermate B also induces generation of more neuronal cells and less glial cells. The in vivo studies indicates that magnesium lithospermate B enhances local neurogenesis since more Ki67+ and Thy1+ cells are observed in hippocampal region with injection of magnesium lithospermate B. Interestingly, enhancing proliferation and neurogenesis occurs in medial forebrain bundle of Parkinson's Disease model and behavioral studies demonstrates that motor function is significantly improved in magnesium lithospermate B-treated disease models. Furthermore, we also found that effect of MLB on proliferation and differentiation of NSCs was mediated by PI3K/Akt signaling. Collectively, our study shows the important role of magnesium lithospermate B in neural stem cell proliferation and differentiation, accordingly providing a simple and efficient method to induce the neuronal cell generation in neurodegenerative disease model.
Collapse
|
13
|
Qi L, Tang Y, He W, Pan H, Jiang W, Wang L, Deng W. Lithium chloride promotes neuronal differentiation of rat neural stem cells and enhances neural regeneration in Parkinson's disease model. Cytotechnology 2017; 69:277-287. [PMID: 28120140 DOI: 10.1007/s10616-016-0056-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 12/16/2016] [Indexed: 01/17/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neural degenerative disease, affecting millions of people globally. Great progress has been made in the PD treatment, and one of the most promising one is the stem cell-based therapy. Thus, studies on the differentiation of neural stem cells (NSCs) are important to the advancement in PD therapy. In this study, we used the rat NSCs to elucidate the role of Lithium in the proliferation and differentiation of NSCs by immunostaining against Ki67 and BrdU analysis as well as immunostaining against specific neuronal markers. We concluded that lithium chloride (LiCl) treatment could enhance the proliferation in NSCs and promote the dopaminergic neuronal differentiation of NSCs in vitro. This process was potentially mediated by Wnt signaling pathway. Using the 6-OHDA-induced PD models, we provided evidence to show that LiCl had the capacity to enhance the proliferation in NSCs and differentiation towards dopaminergic neurons in vivo. The beneficial effect of LiCl treatment was further validated by the fact that the motor function as well as learning and memory was improved in the PD models through Rotarod test and Morris water maze analysis. The learning and memory improvement was further supported by the increase in dendrite spine density in PD models receiving LiCl-treated NSCs. Through this study, we concluded that Lithium plays an important role in promoting NSCs' neuronal differentiation in vitro and improving the symptoms of PD models in vivo. It is of great significance that this work showed the potential application of Lithium in the PD therapy in the future.
Collapse
Affiliation(s)
- Li Qi
- Department of Neurology, The 181st Center Hospital of the People's Liberation Army, No. 1 Xin Qiao Yuan Rd., Guilin, 541002, Guangxi, China.
| | - Yonggang Tang
- Department of Neurology, The 181st Center Hospital of the People's Liberation Army, No. 1 Xin Qiao Yuan Rd., Guilin, 541002, Guangxi, China
| | - Wei He
- Department of Neurology, The 181st Center Hospital of the People's Liberation Army, No. 1 Xin Qiao Yuan Rd., Guilin, 541002, Guangxi, China
| | - Honghua Pan
- Department of Neurology, The 181st Center Hospital of the People's Liberation Army, No. 1 Xin Qiao Yuan Rd., Guilin, 541002, Guangxi, China
| | - Wenxian Jiang
- Department of Neurology, The 181st Center Hospital of the People's Liberation Army, No. 1 Xin Qiao Yuan Rd., Guilin, 541002, Guangxi, China
| | - Lin Wang
- Department of Neurology, The 181st Center Hospital of the People's Liberation Army, No. 1 Xin Qiao Yuan Rd., Guilin, 541002, Guangxi, China
| | - Weilin Deng
- Department of Neurology, The 181st Center Hospital of the People's Liberation Army, No. 1 Xin Qiao Yuan Rd., Guilin, 541002, Guangxi, China
| |
Collapse
|
14
|
Lu J, Yao XQ, Luo X, Wang Y, Chung SK, Tang HX, Cheung CW, Wang XY, Meng C, Li Q. Monosialoganglioside 1 may alleviate neurotoxicity induced by propofol combined with remifentanil in neural stem cells. Neural Regen Res 2017; 12:945-952. [PMID: 28761428 PMCID: PMC5514870 DOI: 10.4103/1673-5374.208589] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Monosialoganglioside 1 (GM1) is the main ganglioside subtype and has neuroprotective properties in the central nervous system. In this study, we aimed to determine whether GM1 alleviates neurotoxicity induced by moderate and high concentrations of propofol combined with remifentanil in the immature central nervous system. Hippocampal neural stem cells were isolated from newborn Sprague-Dawley rats and treated with remifentanil (5, 10, 20 ng/mL) and propofol (1.0, 2.5, 5.0 μg/mL), and/or GM1 (12.5, 25, 50 μg/mL). GM1 reversed combined propofol and remifentanil-induced decreases in the percentage of 5-bromodeoxyuridine(+) cells and also reversed the increase in apoptotic cell percentage during neural stem cell proliferation and differentiation. However, GM1 with combined propofol and remifentanil did not affect β-tubulin(+) or glial fibrillary acidic protein(+) cell percentage during neural stem cell differentiation. In conclusion, we show that GM1 alleviates the damaging effects of propofol combined with remifentanil at moderate and high exposure concentrations in neural stem cells in vitro, and exerts protective effects on the immature central nervous system.
Collapse
Affiliation(s)
- Jiang Lu
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Xue-Qin Yao
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Xin Luo
- Department of Anesthesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.,Laboratory and Clinical Research Institute for Pain, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Sookja Kim Chung
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - He-Xin Tang
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Chi Wai Cheung
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.,Laboratory and Clinical Research Institute for Pain, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xian-Yu Wang
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Chen Meng
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Qing Li
- Anesthesiology Research Institute of Hubei University of Medicine, Shiyan, Hubei Province, China.,Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| |
Collapse
|
15
|
Ma X, Zhou Y, Chai Y, Wang X, Huang X. Stat3 Controls Maturation and Terminal Differentiation in Mouse Hippocampal Neurons. J Mol Neurosci 2016; 61:88-95. [DOI: 10.1007/s12031-016-0820-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/17/2016] [Indexed: 12/14/2022]
|
16
|
Somaa FA, Bye CR, Thompson LH, Parish CL. Meningeal cells influence midbrain development and the engraftment of dopamine progenitors in Parkinsonian mice. Exp Neurol 2015; 267:30-41. [PMID: 25708989 DOI: 10.1016/j.expneurol.2015.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/30/2015] [Accepted: 02/09/2015] [Indexed: 01/09/2023]
Abstract
Dopaminergic neuroblasts, isolated from ventral midbrain fetal tissue, have been shown to structurally and functionally integrate, and alleviate Parkinsonian symptoms following transplantation. The use of donor tissue isolated at an age younger than conventionally employed can result in larger grafts - a consequence of improved cell survival and neuroblast proliferation at the time of implantation. However studies have paid little attention to removal of the meninges from younger tissue, due to its age-dependent tight attachment to the underlying brain. Beyond the protection of the central nervous system, the meninges act as a signaling center, secreting a variety of trophins to influence neural development and additionally impact on neural repair. However it remains to be elucidated what influence these cells have on ventral midbrain development and grafted dopaminergic neuroblasts. Here we examined the temporal role of meningeal cells in graft integration in Parkinsonian mice and, using in vitro approaches, identified the mechanisms underlying the roles of meningeal cells in midbrain development. We demonstrate that young (embryonic day 10), but not older (E12), meningeal cells promote dopaminergic differentiation as well as neurite growth and guidance within grafts and during development. Furthermore we identify stromal derived factor 1 (SDF1), secreted by the meninges and acting on the CXCR4 receptor present on dopaminergic progenitors, as a contributory mediator in these effects. These findings identify new and important roles for the meningeal cells, and SDF1/CXCR4 signaling, in ventral midbrain development as well as neural repair following cell transplantation into the Parkinsonian brain.
Collapse
Affiliation(s)
- Fahad A Somaa
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christopher R Bye
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
17
|
Li Q, Lu J, Wang X. Propofol and remifentanil at moderate and high concentrations affect proliferation and differentiation of neural stem/progenitor cells. Neural Regen Res 2015; 9:2002-7. [PMID: 25598783 PMCID: PMC4283284 DOI: 10.4103/1673-5374.145384] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2014] [Indexed: 12/27/2022] Open
Abstract
Propofol and remifentanil alter intracellular Ca2+ concentration ([Ca2+]i) in neural stem/progenitor cells by activating γ-aminobutyric acid type A receptors and by reducing testosterone levels. However, whether this process affects neural stem/progenitor cell proliferation and differentiation remains unknown. In the present study, we applied propofol and remifentanil, alone or in combination, at low, moderate or high concentrations (1, 2–2.5 and 4–5 times the clinically effective blood drug concentration), to neural stem/progenitor cells from the hippocampi of newborn rat pups. Low concentrations of propofol, remifentanil or both had no noticeable effect on cell proliferation or differentiation; however, moderate and high concentrations of propofol and/or remifentanil markedly suppressed neural stem/progenitor cell proliferation and differentiation, and induced a decrease in [Ca2+]i during the initial stage of neural stem/progenitor cell differentiation. We therefore propose that propofol and remifentanil interfere with the proliferation and differentiation of neural stem/progenitor cells by altering [Ca2+]i. Our findings suggest that propofol and/or remifentanil should be used with caution in pediatric anesthesia.
Collapse
Affiliation(s)
- Qing Li
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China ; Anesthesiology Research Institude, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Jiang Lu
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China ; Anesthesiology Research Institude, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Xianyu Wang
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China ; Anesthesiology Research Institude, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| |
Collapse
|
18
|
Higuchi A, Ling QD, Kumar SS, Chang Y, Alarfaj AA, Munusamy MA, Murugan K, Hsu ST, Umezawa A. Physical cues of cell culture materials lead the direction of differentiation lineages of pluripotent stem cells. J Mater Chem B 2015; 3:8032-8058. [DOI: 10.1039/c5tb01276g] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Differentiation methods of hPSCs into specific cell lineages. Differentiation of hPSCsviaEB formation (types AB, A–D) or without EB formation (types E–H).
Collapse
Affiliation(s)
- Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University
- Taoyuan 32001
- Taiwan
- National Research Institute for Child Health and Development
- Center for Regenerative Medicine
| | - Qing-Dong Ling
- Cathay Medical Research Institute
- Cathay General Hospital
- Taipei
- Taiwan
- Graduate Institute of Systems Biology and Bioinformatics
| | - S. Suresh Kumar
- Department of Medical Microbiology and Parasitology
- Universiti Putra Malaysia
- Selangor
- Malaysia
| | - Yung Chang
- Department of Chemical Engineering
- R&D Center for Membrane Technology
- Chung Yuan Christian University
- Taoyuan
- Taiwan
| | - Abdullah A. Alarfaj
- Department of Botany and Microbiology
- College of Science
- King Saud University
- Riyadh
- Saudi Arabia
| | - Murugan A. Munusamy
- Department of Botany and Microbiology
- College of Science
- King Saud University
- Riyadh
- Saudi Arabia
| | - Kadarkarai Murugan
- Division of Entomology
- Department of Zoology
- School of Life Sciences
- Bharathiar University
- Coimbatore 641046
| | - Shih-Tien Hsu
- Department of Internal Medicine
- Taiwan Landseed Hospital
- Taoyuan
- Taiwan
| | - Akihiro Umezawa
- National Research Institute for Child Health and Development
- Center for Regenerative Medicine
- Tokyo 157-8535
- Japan
| |
Collapse
|
19
|
Upadhyay G, Shankar S, Srivastava RK. Stem Cells in Neurological Disorders: Emerging Therapy with Stunning Hopes. Mol Neurobiol 2014; 52:610-25. [DOI: 10.1007/s12035-014-8883-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 08/27/2014] [Indexed: 12/14/2022]
|
20
|
Addington CP, Pauken CM, Caplan MR, Stabenfeldt SE. The role of SDF-1α-ECM crosstalk in determining neural stem cell fate. Biomaterials 2014; 35:3263-72. [PMID: 24438907 DOI: 10.1016/j.biomaterials.2013.12.102] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 12/28/2013] [Indexed: 02/04/2023]
Abstract
The consequences of central nervous system injury are far-reaching and debilitating and, while an endogenous repair response to neural injury has been observed in recent years, the mechanisms behind this response remain unclear. Neural progenitor/stem cell (NPSC) migration to the site of injury from the neural stem cell niches (e.g. subventricular zone and hippocampus) has been observed to be vasophilic in nature. While the chemotactic stimuli directing NPSC homing to injury is not well established, it is thought to be due in part to an increasing gradient of chemotactic cytokines, such as stromal cell-derived factor 1α (SDF-1α). Based on these recent findings, we hypothesize that critical crosstalk between SDF-1α and the extracellular matrix (ECM) drives injury-induced NPSC behavior. In this study, we investigated the effect of SDF-1α and ECM substrates (Matrigel, laminin, and vitronectin) on the migration, differentiation, and proliferation of NPSCs in vitro using standard assays. The results demonstrated that SDF-1α and laminin-based ECM (Matrigel and laminin) significantly and synergistically enhanced NPSC migration and acute neuronal differentiation. These effects were significantly attenuated with the addition of AMD3100 (an antagonist against the SDF-1α receptor, CXCR4). SDF-1α alone significantly increased NPSC proliferation regardless of ECM substrate, however no synergy was observed between SDF-1α and the ECM. These results serve to elucidate the relationship between adhesive and soluble signaling factors of interest and their effect on NPSC behavior following neural injury. Furthermore, these results better inform the next generation of biomaterials aimed at stimulating endogenous neural regeneration for neural injury and neurodegenerative diseases.
Collapse
Affiliation(s)
- Caroline P Addington
- School of Biological and Health Systems Engineering, Arizona State University, P.O. Box 879709, Tempe, AZ 85287-9709, USA
| | - Christine M Pauken
- School of Biological and Health Systems Engineering, Arizona State University, P.O. Box 879709, Tempe, AZ 85287-9709, USA
| | - Michael R Caplan
- School of Biological and Health Systems Engineering, Arizona State University, P.O. Box 879709, Tempe, AZ 85287-9709, USA
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, P.O. Box 879709, Tempe, AZ 85287-9709, USA.
| |
Collapse
|
21
|
Jha RM, Chrenek R, Magnotti LM, Cardozo DL. The isolation, differentiation, and survival in vivo of multipotent cells from the postnatal rat filum terminale. PLoS One 2013; 8:e65974. [PMID: 23762453 PMCID: PMC3675200 DOI: 10.1371/journal.pone.0065974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 05/03/2013] [Indexed: 01/25/2023] Open
Abstract
Neural stem cells (NSCs) are undifferentiated cells in the central nervous system (CNS) that are capable of self-renewal and can be induced to differentiate into neurons and glia. Current sources of mammalian NSCs are confined to regions of the CNS that are critical to normal function and surgically difficult to access, which limits their therapeutic potential in human disease. We have found that the filum terminale (FT), a previously unexplored, expendable, and easily accessible tissue at the caudal end of the spinal cord, is a source of multipotent cells in postnatal rats and humans. In this study, we used a rat model to isolate and characterize the potential of these cells. Neurospheres derived from the rat FT are amenable to in vitro expansion in the presence of a combination of growth factors. These proliferating, FT-derived cells formed neurospheres that could be induced to differentiate into neural progenitor cells, neurons, astrocytes, and oligodendrocytes by exposure to serum and/or adhesive substrates. Through directed differentiation using sonic hedgehog and retinoic acid in combination with various neurotrophic factors, FT-derived neurospheres generated motor neurons that were capable of forming neuromuscular junctions in vitro. In addition, FT-derived progenitors that were injected into chick embryos survived and could differentiate into both neurons and glia in vivo.
Collapse
Affiliation(s)
- Ruchira M. Jha
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ryan Chrenek
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Laura M. Magnotti
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| | - David L. Cardozo
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
22
|
Song L, Liu P, Han C, Liu Y, Zou W, Piao H, Wang Y, Liu J. A novel approach to facilitate dopaminergic neuron generation from stem-cells: The combination of genetic modification and signaling factors within a three-dimensional perfusion microbioreactor. Med Hypotheses 2013; 80:407-10. [DOI: 10.1016/j.mehy.2012.12.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 12/04/2012] [Accepted: 12/29/2012] [Indexed: 12/12/2022]
|
23
|
Zhang HY, Zhang X, Wang ZG, Shi HX, Wu FZ, Lin BB, Xu XL, Wang XJ, Fu XB, Li ZY, Shen CJ, Li XK, Xiao J. Exogenous basic fibroblast growth factor inhibits ER stress-induced apoptosis and improves recovery from spinal cord injury. CNS Neurosci Ther 2012; 19:20-9. [PMID: 23082997 DOI: 10.1111/cns.12013] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 08/26/2012] [Accepted: 08/27/2012] [Indexed: 01/13/2023] Open
Abstract
AIM To investigate the mechanism of endoplasmic reticulum (ER) stress-induced apoptosis as well as the protective action of basic fibroblast growth factor (bFGF) both in vivo and in vitro. METHODS AND RESULTS ER stress-induced apoptosis was involved in the injuries of spinal cord injury (SCI) model rat. bFGF administration improved the recovery and increased the survival of neurons in spinal cord lesions in model rat. The protective effect of bFGF is related to the inhibition of CHOP, GRP78 and caspase-12, which are ER stress-induced apoptosis response proteins. bFGF administration also increased the survival of neurons and the expression of growth-associated protein 43 (GAP43), which is related to neural regeneration. The protective effect of bFGF is related to the activation of downstream signals, PI3K/Akt/GSK-3β and ERK1/2, especially in the ER stress cell model. CONCLUSIONS This is the first study to illustrate that the role of bFGF in SCI recovery is related to the inhibition of ER stress-induced cell death via the activation of downstream signals. Our work also suggested a new trend for bFGF drug development in central neural system injuries, which are involved in chronic ER stress-induced apoptosis.
Collapse
Affiliation(s)
- Hong-Yu Zhang
- School of Pharmacy, Key Laboratory of Biotechnology and Pharmaceutical Engineering, Wenzhou Medical College, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Tan X, Zhang L, Qin J, Tian M, Zhu H, Dong C, Zhao H, Jin G. Transplantation of neural stem cells co-transfected with Nurr1 and Brn4 for treatment of Parkinsonian rats. Int J Dev Neurosci 2012; 31:82-7. [PMID: 23085081 DOI: 10.1016/j.ijdevneu.2012.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 09/17/2012] [Accepted: 10/08/2012] [Indexed: 12/31/2022] Open
Abstract
Neural stem cells (NSCs) tranplantation has great potential for the treatment of neurodegenerative disease such as Parkinson's disease (PD). However, the usage of NSCs is limited because the differentiation of NSCs into specific dopaminergic neurons has proven difficult. We have recently demonstrated that transgenic expression of Nurr1 could induce the differentiation of NSCs into tyrosine hydroxylase (TH) immunoreactive dopaminergic neurons, and forced co-expression of Nurr1 with Brn4 caused a dramatic increase in morphological and phenotypical maturity of these neurons. In this study, we investigated the effect of transplanted NSCs in PD model rats. The results showed that overexpression of Nurr1 promoted NSCs to differentiate into dopaminergic neurons in vivo, increased the level of dopamine (DA) neurotransmitter in the striatum, resulting in behavioral improvement of PD rats. Importantly, co-expression of Nurr1 and Brn4 in NSCs significantly increased the maturity and viability of dopaminergic neurons, further raised the DA amount in the striatum and reversed the behavioral deficit of the PD rats. Our findings provide a new potential and strategy for the use of NSCs in cell replacement therapy for PD.
Collapse
Affiliation(s)
- Xuefeng Tan
- Department of Anatomy and Neurobiology, the Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Guan S, Zhang XL, Lin XM, Liu TQ, Ma XH, Cui ZF. Chitosan/gelatin porous scaffolds containing hyaluronic acid and heparan sulfate for neural tissue engineering. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2012; 24:999-1014. [DOI: 10.1080/09205063.2012.731374] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Shui Guan
- a Stem Cell and Tissue Engineering Laboratory , Dalian University of Technology , Dalian , China
| | - Xiu-Li Zhang
- b School of Pharmaceutical Sciences, Binzhou Medical University , Yantai, Shandong , China
| | - Xiao-Min Lin
- a Stem Cell and Tissue Engineering Laboratory , Dalian University of Technology , Dalian , China
| | - Tian-Qing Liu
- a Stem Cell and Tissue Engineering Laboratory , Dalian University of Technology , Dalian , China
| | - Xue-Hu Ma
- a Stem Cell and Tissue Engineering Laboratory , Dalian University of Technology , Dalian , China
| | - Zhan-Feng Cui
- c Department of Engineering Science , Oxford University , Oxford , UK
| |
Collapse
|
26
|
Regulation of neural stem cell differentiation by transcription factors HNF4-1 and MAZ-1. Mol Neurobiol 2012; 47:228-40. [PMID: 22944911 DOI: 10.1007/s12035-012-8335-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 08/16/2012] [Indexed: 10/27/2022]
Abstract
Neural stem cells (NSCs) are promising candidates for a variety of neurological diseases due to their ability to differentiate into neurons, astrocytes, and oligodentrocytes. During this process, Rho GTPases are heavily involved in neuritogenesis, axon formation and dendritic development, due to their effects on the cytoskeleton through downstream effectors. The activities of Rho GTPases are controlled by Rho-GDP dissociation inhibitors (Rho-GDIs). As shown in our previous study, these are also involved in the differentiation of NSCs; however, little is known about the underlying regulatory mechanism. Here, we describe how the transcription factors hepatic nuclear factor (HNF4-1) and myc-associated zinc finger protein (MAZ-1) regulate the expression of Rho-GDIγ in the stimulation of NSC differentiation. Using a transfection of cis-element double-stranded oligodeoxynucleotides (ODNs) strategy, referred to as "decoy" ODNs, we examined the effects of HNF4-1 and MAZ-1 on NSC differentiation in the NSC line C17.2. Our results show that HNF4-1 and MAZ-1 decoy ODNs significantly knock down Rho-GDIγ gene transcription, leading to NSC differentiation towards neurons. We observed that HNF4-1 and MAZ-1 decoy ODNs are able enter to the cell nucleolus and specifically bind to their target transcription factors. Furthermore, the expression of Rho-GDIγ-mediated genes was identified, suggesting that the regulatory mechanism for the differentiation of NSCs is triggered by the transcription factors MAZ-1 and HNF4-1. These findings indicate that HNF4-1 and MAZ-1 regulate the expression of Rho-GDIγ and contribute to the differentiation of NSCs. Our findings provide a new perspective within regulatory mechanism research during differentiation of NSCs, especially the clinical application of transcription factor decoys in vivo, suggesting potential therapeutic strategies for neurodegenerative disease.
Collapse
|
27
|
Lu J, Lu K, Li D. Changes in expression and secretion patterns of fibroblast growth factor 8 and Sonic Hedgehog signaling pathway molecules during murine neural stem/progenitor cell differentiation in vitro. Neural Regen Res 2012; 7:1688-94. [PMID: 25624789 PMCID: PMC4302448 DOI: 10.3969/j.issn.1673-5374.2012.22.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 05/03/2012] [Indexed: 12/21/2022] Open
Abstract
In the present study, we investigated the dynamic expression of fibroblast growth factor 8 and Sonic Hedgehog signaling pathway related factors in the process of in vitro hippocampal neural stem/progenitor cell differentiation from embryonic Sprague-Dawley rats or embryonic Kunming species mice, using fluorescent quantitative reverse transcription-PCR and western blot analyses. Results demonstrated that the dynamic expression of fibroblast growth factor 8 was similar to fibroblast growth factor receptor 1 expression but not to other fibroblast growth factor receptors. Enzyme-linked immunosorbent assay demonstrated that fibroblast growth factor 8 and Sonic Hedgehog signaling pathway protein factors were secreted by neural cells into the intercellular niche. Our experimental findings indicate that fibroblast growth factor 8 and Sonic Hedgehog expression may be related to the differentiation of neural stem/progenitor cells.
Collapse
Affiliation(s)
- Jiang Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China ; Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Kehuan Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| | - Dongsheng Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| |
Collapse
|
28
|
Lu J, Li D, Lu K. Distribution and localization of fibroblast growth factor-8 in rat brain and nerve cells during neural stem/progenitor cell differentiation. Neural Regen Res 2012; 7:1455-62. [PMID: 25657679 PMCID: PMC4308775 DOI: 10.3969/j.issn.1673-5374.2012.19.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 05/13/2012] [Indexed: 11/24/2022] Open
Abstract
The present study explored the distribution and localization of fibroblast growth factor-8 and its potential receptor, fibroblast growth factor receptor-3, in adult rat brain in vivo and in nerve cells during differentiation of neural stem/progenitor cells in vitro. Immunohistochemistry was used to examine the distribution of fibroblast growth factor-8 in adult rat brain in vivo. Localization of fibroblast growth factor-8 and fibroblast growth factor receptor-3 in cells during neural stem/progenitor cell differentiation in vitro was detected by immunofluorescence. Flow cytometry and immunofluorescence were used to evaluate the effect of an anti-fibroblast growth factor-8 antibody on neural stem/progenitor cell differentiation and expansion in vitro. Results from this study confirmed that fibroblast growth factor-8 was mainly distributed in adult midbrain, namely the substantia nigra, compact part, dorsal tier, substantia nigra and reticular part, but was not detected in the forebrain comprising the caudate putamen and striatum. Unusual results were obtained in retrosplenial locations of adult rat brain. We found that fibroblast growth factor-8 and fibroblast growth factor receptor-3 were distributed on the cell membrane and in the cytoplasm of nerve cells using immunohistochemistry and immunofluorescence analyses. We considered that the distribution of fibroblast growth factor-8 and fibroblast growth factor receptor-3 in neural cells corresponded to the characteristics of fibroblast growth factor-8, a secretory factor. Addition of an anti-fibroblast growth factor-8 antibody to cultures significantly affected the rate of expansion and differentiation of neural stem/progenitor cells. In contrast, addition of recombinant fibroblast growth factor-8 to differentiation medium promoted neural stem/progenitor cell differentiation and increased the final yields of dopaminergic neurons and total neurons. Our study may help delineate the important roles of fibroblast growth factor-8 in brain activities and neural stem/progenitor cell differentiation.
Collapse
Affiliation(s)
- Jiang Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China ; Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Dongsheng Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei Province, China
| | - Kehuan Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning 530004, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
29
|
Relucio J, Menezes MJ, Miyagoe-Suzuki Y, Takeda S, Colognato H. Laminin regulates postnatal oligodendrocyte production by promoting oligodendrocyte progenitor survival in the subventricular zone. Glia 2012; 60:1451-67. [PMID: 22706957 DOI: 10.1002/glia.22365] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 05/10/2012] [Indexed: 01/28/2023]
Abstract
The laminin family of extracellular matrix proteins are expressed broadly during embryonic brain development, but are enriched at ventricular and pial surfaces where laminins mediate radial glial attachment during corticogenesis. In the adult brain, however, laminin distribution is restricted, yet is found within the vascular basal lamina and associated fractones of the ventricular zone (VZ)-subventricular zone (SVZ) stem cell niche, where laminins regulate adult neural progenitor cell proliferation. It remains unknown, however, if laminins regulate the wave of oligodendrogenesis that occurs in the neonatal/early postnatal VZ-SVZ. Here we report that Lama2, the gene that encodes the laminin α2-subunit, regulates postnatal oligodendrogenesis. At birth, Lama2-/- mice had significantly higher levels of dying oligodendrocyte progenitor cells (OPCs) in the OPC germinal zone of the dorsal SVZ. This translated into fewer OPCs, both in the dorsal SVZ well as in an adjacent developing white matter tract, the corpus callosum. In addition, intermediate progenitor cells that give rise to OPCs in the Lama2-/- VZ-SVZ were mislocalized and proliferated nearer to the ventricle surface. Later, delays in oligodendrocyte maturation (with accompanying OPC accumulation), were observed in the Lama2-/- corpus callosum, leading to dysmyelination by postnatal day 21. Together these data suggest that prosurvival laminin interactions in the developing postnatal VZ-SVZ germinal zone regulate the ability, or timing, of oligodendrocyte production to occur appropriately.
Collapse
Affiliation(s)
- Jenne Relucio
- Department of Pharmacology, Stony Brook University, Stony Brook, New York, 11794, USA
| | | | | | | | | |
Collapse
|
30
|
Higuchi A, Ling QD, Hsu ST, Umezawa A. Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation. Chem Rev 2012; 112:4507-40. [PMID: 22621236 DOI: 10.1021/cr3000169] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, 32001 Taiwan.
| | | | | | | |
Collapse
|
31
|
Human Umbilical Cord-Derived Schwann-Like Cell Transplantation Combined with Neurotrophin-3 Administration in Dyskinesia of Rats with Spinal Cord Injury. Neurochem Res 2011; 36:783-92. [DOI: 10.1007/s11064-011-0402-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2011] [Indexed: 10/25/2022]
|
32
|
Li M, Zhang SZ, Guo YW, Cai YQ, Yan ZJ, Zou Z, Jiang XD, Ke YQ, He XY, Jin ZL, Lu GH, Su DQ. Human umbilical vein-derived dopaminergic-like cell transplantation with nerve growth factor ameliorates motor dysfunction in a rat model of Parkinson's disease. Neurochem Res 2010; 35:1522-9. [PMID: 20658188 DOI: 10.1007/s11064-010-0211-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2010] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells are capable of differentiating into dopaminergic-like cells, but currently no report has been available to describe the induction of human umbilical vein mesenchymal stem cells (HUVMSCs) into dopaminergic-like cells. In this study, we induced HUVMSCs in vitro into neurospheres constituted by neural stem-like cells, and further into cells bearing strong morphological, phenotypic and functional resemblances with dopaminergic-like cells. These HUVMSC-derived dopaminergic-like cells, after grafting into the brain of a rat model of Parkinson's disease (PD), showed a partial therapeutic effect in terms of the behavioral improvement. Nerve growth factor was reported to improve the local microenvironment of the grafted cells, and we therefore further tested the effect of dopaminergic-like cell grafting combined with nerve growth factor (NGF) administration at the site of cell transplantation. The results showed that NGF administration significantly promoted the survival of the grafted cells in the host brain and enhanced the content of dopaminergic in the local brain tissue. Behavioral test demonstrated a significant improvement of the motor function of the PD rats after dopaminergic-like cell grafting with NGF administration as compared with that of rats receiving the cell grafting only. These results suggest that transplantation of the dopaminergic-like cells combined with NGF administration may represent a new strategy of stem cell therapy for PD.
Collapse
Affiliation(s)
- Ming Li
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Fricker-Gates RA, Gates MA. Stem cell-derived dopamine neurons for brain repair in Parkinson’s disease. Regen Med 2010; 5:267-78. [DOI: 10.2217/rme.10.3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
One of the prospects for a curative treatment for Parkinson’s disease is to replace the lost dopaminergic neurons. Preclinical and clinical trials have demonstrated that dissected fetal dopaminergic neurons have the potential to markedly improve motor function in animal models and Parkinson’s disease patients. However, this source of cells will never be sufficient to use as a widespread therapy. Over the last 20 years, scientists have been searching for other reliable sources of midbrain dopamine neurons, and stem cells appear to be strong candidates. This article reviews the potential of different types of stem cells, from embryonic to adult to induced pluripotent stem cells, to see how well the cells can be differentiated into fully functional dopamine neurons, which cells might be the best candidates and how much more research is required before stem cell technology might be translated to a clinical therapy for Parkinson’s disease.
Collapse
|
34
|
Gu S, Huang H, Bi J, Yao Y, Wen T. Combined treatment of neurotrophin-3 gene and neural stem cells is ameliorative to behavior recovery of Parkinson's disease rat model. Brain Res 2009; 1257:1-9. [DOI: 10.1016/j.brainres.2008.12.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 11/15/2008] [Accepted: 12/01/2008] [Indexed: 10/21/2022]
|
35
|
Yamada T, Sawada R, Tsuchiya T. The effect of sulfated hyaluronan on the morphological transformation and activity of cultured human astrocytes. Biomaterials 2008; 29:3503-13. [PMID: 18533253 DOI: 10.1016/j.biomaterials.2008.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 03/26/2008] [Indexed: 10/22/2022]
Abstract
We demonstrated the effect of synthesized sulfated hyaluronan (SHya), which is composed of a sulfated group and hyaluronan, and basic fibroblast growth factor 2 (FGF-2) on normal human astrocytes (NHA) activity and its morphological transformation in vitro study. Astrocyte is a kind of glial cell and stellated astrocyte (activating astrocyte) supports axons network, neurons survival and synaptic plasticity. Treatment of SHya hardly affected NHA proliferation. However combination treatment of SHya and FGF-2 increased NHA proliferation. Treatment of SHya promoted transformation of normal astrocyte into a stella morphology (stellation) and combination treatment of SHya and FGF-2 promoted stellation than that of SHya only. Treatment of SHya increased glial fibrillary acidic protein (GFAP), nestin mRNA and GFAP protein expression in the stellated NHA. The cell-cell adhesion of NHA increased by treatment of SHya. Treatment of SHya increased heparin-binding trophic factors FGF-2, midkine, and some other trophic factors mRNA level in the NHA. These results suggested that the treatment of SHya promoted NHA activity due to enhancing neurotrophins production and the morphological transformation of NHA and the effect of SHya on astrocytes partly involved FGF-2 activity. These findings indicate that SHya may be involved in the astrocyte activity and support neurons survivals.
Collapse
Affiliation(s)
- Takashi Yamada
- Division of Medical Devices, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | | | | |
Collapse
|