1
|
Hao K, Chen F, Xu S, Xiong Y, Xu R, Huang H, Shu C, Wang H, Wang G, Reynolds GP. The role of SIRT3 in mediating the cognitive deficits and neuroinflammatory changes associated with a developmental animal model of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110914. [PMID: 38122862 DOI: 10.1016/j.pnpbp.2023.110914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/30/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
The neuroinflammatory state may contribute to the pathogenesis of many mental disorders including schizophrenia. Nicotinamide adenine dinucleotide (NAD+) is an essential cofactor for activation of proteins involved in mitochondria quality control, such as Sirtuin3 (SIRT3). Our previous study has found that NAD+ supplement could rescue early life stress (ELS)-induced neuroinflammation and down-regulation of SIRT3 in adult offspring. However, it is unclear whether SIRT3 is the key to the neuroprotective effects of NAD+ supplement in this animal model of schizophrenia. The present study used 24 h maternal separation (MS) as ELS to Wistar rat pups on the postnatal day (PND) 9. Schizophrenia-like behaviors and memory impairments were detected by behavioral tests. Microglial activation, pro-inflammatory cytokine expression, and NAD+/SIRT3 expression were detected in the prefrontal cortex and hippocampus. Meanwhile, NAM (a precursor of NAD+), and the SIRT3 activator Honokiol (HNK), and the SIRT3 inhibitor 3-TYP were used as an intervention in vivo. Our results showed that ELS could induce schizophrenia-like behaviors and M1 microglial activation, NAD+ decline, lower expression of SIRT3, and increased acetylated superoxide dismutase 2 expression at the adult stage. NAD+ supplement or HNK administration could block this process and normalize the behavioral alterations of the MS animals. 3-TYP administration in the control group and the NAM-treated MS rats caused M1 microglial activation and cognitive deficits. Our results demonstrated that SIRT3 mediated the stabilizing effect of NAD+ on normalizing M1 microglial activation and behavioral phenotypes in MS rats.
Collapse
Affiliation(s)
- Keke Hao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fashuai Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shilin Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ying Xiong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rui Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Huan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chang Shu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Institute of Neurology and Psychiatry Research, Wuhan 430060, China.
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| |
Collapse
|
2
|
Nasrolahi A, Shabani Z, Sadigh-Eteghad S, Salehi-Pourmehr H, Mahmoudi J. Stem Cell Therapy for the Treatment of Parkinson's Disease: What Promise Does it Hold? Curr Stem Cell Res Ther 2024; 19:185-199. [PMID: 36815638 DOI: 10.2174/1574888x18666230222144116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 02/24/2023]
Abstract
Parkinson's disease (PD) is a common, progressive neurodegenerative disorder characterized by substantia nigra dopamine cell death and a varied clinical picture that affects older people. Although more than two centuries have passed since the earliest attempts to find a cure for PD, it remains an unresolved problem. With this in mind, cell replacement therapy is a new strategy for treating PD. This novel approach aims to replace degenerated dopaminergic (DAergic) neurons with new ones or provide a new source of cells that can differentiate into DAergic neurons. Induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), neural stem cells (NSCs), and embryonic stem cells (ESCs) are among the cells considered for transplantation therapies. Recently disease-modifying strategies like cell replacement therapies combined with other therapeutic approaches, such as utilizing natural compounds or biomaterials, are proposed to modify the underlying neurodegeneration. In the present review, we discuss the current advances in cell replacement therapy for PD and summarize the existing experimental and clinical evidence supporting this approach.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Shabani
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Salehi-Pourmehr
- Research Center for Evidence-Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Cho YW, Park JH, Kang MJ, Kim TH. Crater-like nanoelectrode arrays for electrochemical detection of dopamine release from neuronal cells. Biomed Mater 2023; 18:065015. [PMID: 37769679 DOI: 10.1088/1748-605x/acfe69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Stem cell therapy has shown great potential in treating various incurable diseases using conventional chemotherapy. Parkinson's disease (PD)-a neurodegenerative disease-has been reported to be caused by quantitative loss or abnormal functionality of dopaminergic neurons (DAnergic neurons). To date, stem cell therapies have shown some potential in treating PD throughex vivoengraftment of stem-cell-derived neurons. However, accurately identifying the differentiation and non-invasively evaluating the functionality and maturity of DAnergic neurons are formidable challenges in stem cell therapies. These strategies are important in enhancing the efficacy of stem cell therapies. In this study, we report a novel cell cultivation platform, that is, a nanocrater-like electrochemical nanoelectrode array (NCENA) for monitoring dopamine (DA) release from neurons to detect exocytotic DA release from DAnergic neurons. In particular, the developed NCENA has a nanostructure in which three-dimensional porous gold nanopillars are uniformly arranged on conductive electrodes. The developed NCENA exhibited great DA sensing capabilities with a linear range of 0.39-150μM and a limit of detection of 1.16μM. Furthermore, the nanotopographical cues provided by the NCENA are suitable for cell cultivation with enhanced cellular adhesion. Finally, we successfully analysed the functionality and maturity of differentiated neurons on the NCENA through its excellent sensing ability for exocytotic DA.
Collapse
Affiliation(s)
- Yeon-Woo Cho
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Joon-Ha Park
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Min-Ji Kang
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
4
|
Alidoust L, Akhoondian M, Atefi AH, Keivanlou MH, Hedayati Ch M, Jafari A. Stem cell-conditioned medium is a promising treatment for Alzheimer's disease. Behav Brain Res 2023; 452:114543. [PMID: 37311523 DOI: 10.1016/j.bbr.2023.114543] [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: 03/14/2023] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND AIM Alzheimer's disease (AD), a prevalent progressive neurodegenerative disease, is mainly characterized by dementia, memory loss, and cognitive disorder. Rising research was performed to develop pharmacological or non-pharmacological approaches to treat or improve AD complications. Mesenchymal stem cells (MSCs) are stromal cells that can self-renew and exhibit multilineage differentiation. Recent evidence suggested that some of the therapeutic effects of MSCs are mediated by the secreted paracrine factors. These paracrine factors, called MSC- conditioned medium (MSC-CM), may stimulate endogenous repair, promote angio- and artery genesis, and reduce apoptosis through paracrine mechanisms. The current study aims to systematically review the advantages of MSC-CM to the development of research and therapeutic concepts for AD management. MATERIAL AND METHODS The present systematic review was performed using PubMed, Web of Science, and Scopus from April 2020 to May 2022 following the "Preferred Reporting Items for Systematic Reviews" (PRISMA) guidelines. The keywords, including "Conditioned medium OR Conditioned media OR Stem cell therapy" AND "Alzheimer's," was searched, and finally, 13 papers were extracted. RESULTS The obtained data revealed that MSC-CMs might positively affect neurodegenerative diseases prognosis, especially AD, through various mechanisms, including a decrease in neuro-inflammation, reduction of oxidative stress and Aβ formation, modulation of Microglia function and count, reduction of apoptosis, induction of synaptogenesis and neurogenesis. Also, the results showed that MSC-CM administration could significantly improve cognitive and memory function, increase the expression of neurotrophic factors, decrease the production of pro-inflammatory cytokines, improve mitochondrial function, reduce cytotoxicity, and increase neurotransmitter levels. CONCLUSION While inhibiting the induction of neuroinflammation could be considered the first therapeutic effect of CMs, the prevention of apoptosis could be regarded as the most crucial effect of CMs on AD improvement.
Collapse
Affiliation(s)
- Leila Alidoust
- Department of Genetics, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Akhoondian
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amir Homayoun Atefi
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Mojtaba Hedayati Ch
- Department of Microbiology, Virology and Microbial Toxins, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Microbial Toxins Physiology Group (MTPG), Universal Scientific Education Research Network (USERN), Rasht, Iran
| | - Adele Jafari
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
5
|
Hao K, Wang H, Zhang Y, Xie X, Huang H, Chen C, Xu S, Xu R, Shu C, Liu Z, Zhou Y, Reynolds GP, Wang G. Nicotinamide reverses deficits in puberty-born neurons and cognitive function after maternal separation. J Neuroinflammation 2022; 19:232. [PMID: 36131290 PMCID: PMC9494869 DOI: 10.1186/s12974-022-02591-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 09/04/2022] [Indexed: 01/09/2023] Open
Abstract
Background Early life stress (ELS) is associated with the development of schizophrenia later in life. The hippocampus develops significantly during childhood and is extremely reactive to stress. In rodent models, ELS can induce neuroinflammation, hippocampal neuronal loss, and schizophrenia-like behavior. While nicotinamide (NAM) can inhibit microglial inflammation, it is unknown whether NAM treatment during adolescence reduces hippocampal neuronal loss and abnormal behaviors induced by ELS. Methods Twenty-four hours of maternal separation (MS) of Wistar rat pups on post-natal day (PND)9 was used as an ELS. On PND35, animals received a single intraperitoneal injection of BrdU to label dividing neurons and were given NAM from PND35 to PND65. Behavioral testing was performed. Western blotting and immunofluorescence staining were used to detect nicotinamide adenine dinucleotide (NAD+)/Sirtuin3 (Sirt3)/superoxide dismutase 2 (SOD2) pathway-related proteins. Results Compared with controls, only MS animals in the adult stage (PND56–65) but not the adolescent stage (PND31–40) exhibited pre-pulse inhibition deficits and cognitive impairments mimicking schizophrenia symptoms. MS decreased the survival and activity of puberty-born neurons and hippocampal NAD+ and Sirt3 expression in adulthood. These observations were related to an increase in acetylated SOD2, microglial activation, and significant increases in pro-inflammatory IL-1β, TNF-α, and IL-6 expression. All the effects of MS at PND9 were reversed by administering NAM in adolescence (PND35–65). Conclusions MS may lead to schizophrenia-like phenotypes and persistent hippocampal abnormalities. NAM may be a safe and effective treatment in adolescence to restore normal hippocampal function and prevent or ameliorate schizophrenia-like behavior. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02591-y.
Collapse
Affiliation(s)
- Keke Hao
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China. .,Department of Psychiatry, Zhongxiang Hospital of Renmin Hospital of Wuhan University, Zhongxiang, 431900, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Yuejin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, China
| | - Xinhui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Huan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Cheng Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Shilin Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Rui Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Chang Shu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yuan Zhou
- Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China. .,Hubei Institute of Neurology and Psychiatry Research, Wuhan, 430060, China.
| |
Collapse
|
6
|
Notaras M, Lodhi A, Dündar F, Collier P, Sayles NM, Tilgner H, Greening D, Colak D. Schizophrenia is defined by cell-specific neuropathology and multiple neurodevelopmental mechanisms in patient-derived cerebral organoids. Mol Psychiatry 2022; 27:1416-1434. [PMID: 34789849 PMCID: PMC9095467 DOI: 10.1038/s41380-021-01316-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/03/2021] [Accepted: 09/22/2021] [Indexed: 01/02/2023]
Abstract
Due to an inability to ethically access developing human brain tissue as well as identify prospective cases, early-arising neurodevelopmental and cell-specific signatures of Schizophrenia (Scz) have remained unknown and thus undefined. To overcome these challenges, we utilized patient-derived induced pluripotent stem cells (iPSCs) to generate 3D cerebral organoids to model neuropathology of Scz during this critical period. We discovered that Scz organoids exhibited ventricular neuropathology resulting in altered progenitor survival and disrupted neurogenesis. This ultimately yielded fewer neurons within developing cortical fields of Scz organoids. Single-cell sequencing revealed that Scz progenitors were specifically depleted of neuronal programming factors leading to a remodeling of cell-lineages, altered differentiation trajectories, and distorted cortical cell-type diversity. While Scz organoids were similar in their macromolecular diversity to organoids generated from healthy controls (Ctrls), four GWAS factors (PTN, COMT, PLCL1, and PODXL) and peptide fragments belonging to the POU-domain transcription factor family (e.g., POU3F2/BRN2) were altered. This revealed that Scz organoids principally differed not in their proteomic diversity, but specifically in their total quantity of disease and neurodevelopmental factors at the molecular level. Single-cell sequencing subsequently identified cell-type specific alterations in neuronal programming factors as well as a developmental switch in neurotrophic growth factor expression, indicating that Scz neuropathology can be encoded on a cell-type-by-cell-type basis. Furthermore, single-cell sequencing also specifically replicated the depletion of BRN2 (POU3F2) and PTN in both Scz progenitors and neurons. Subsequently, in two mechanistic rescue experiments we identified that the transcription factor BRN2 and growth factor PTN operate as mechanistic substrates of neurogenesis and cellular survival, respectively, in Scz organoids. Collectively, our work suggests that multiple mechanisms of Scz exist in patient-derived organoids, and that these disparate mechanisms converge upon primordial brain developmental pathways such as neuronal differentiation, survival, and growth factor support, which may amalgamate to elevate intrinsic risk of Scz.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Aiman Lodhi
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Friederike Dündar
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Paul Collier
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Nicole M Sayles
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Hagen Tilgner
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - David Greening
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Institute & Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Dilek Colak
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA.
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| |
Collapse
|
7
|
Ren J, Li C, Zhang M, Wang H, Xie Y, Tang Y. A Step-by-Step Refined Strategy for Highly Efficient Generation of Neural Progenitors and Motor Neurons from Human Pluripotent Stem Cells. Cells 2021; 10:3087. [PMID: 34831309 PMCID: PMC8625124 DOI: 10.3390/cells10113087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/24/2021] [Accepted: 11/05/2021] [Indexed: 01/02/2023] Open
Abstract
Limited access to human neurons, especially motor neurons (MNs), was a major challenge for studying neurobiology and neurological diseases. Human pluripotent stem cells (hPSCs) could be induced as neural progenitor cells (NPCs) and further multiple neural subtypes, which provide excellent cellular sources for studying neural development, cell therapy, disease modeling and drug screening. It is thus important to establish robust and highly efficient methods of neural differentiation. Enormous efforts have been dedicated to dissecting key signalings during neural commitment and accordingly establishing reliable differentiation protocols. In this study, we refined a step-by-step strategy for rapid differentiation of hPSCs towards NPCs within merely 18 days, combining the adherent and neurosphere-floating methods, as well as highly efficient generation (~90%) of MNs from NPCs by introducing refined sets of transcription factors for around 21 days. This strategy made use of, and compared, retinoic acid (RA) induction and dual-SMAD pathway inhibition, respectively, for neural induction. Both methods could give rise to highly efficient and complete generation of preservable NPCs, but with different regional identities. Given that the generated NPCs can be differentiated into the majority of excitatory and inhibitory neurons, but hardly MNs, we thus further differentiate NPCs towards MNs by overexpressing refined sets of transcription factors, especially by adding human SOX11, whilst improving a series of differentiation conditions to yield mature MNs for good modeling of motor neuron diseases. We thus refined a detailed step-by-step strategy for inducing hPSCs towards long-term preservable NPCs, and further specified MNs based on the NPC platform.
Collapse
Affiliation(s)
- Jie Ren
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China; (J.R.); (C.L.); (M.Z.); (H.W.)
- Aging Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Chaoyi Li
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China; (J.R.); (C.L.); (M.Z.); (H.W.)
- Aging Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Mengfei Zhang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China; (J.R.); (C.L.); (M.Z.); (H.W.)
- Aging Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Huakun Wang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China; (J.R.); (C.L.); (M.Z.); (H.W.)
- Aging Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Yali Xie
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
- The Biobank of Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yu Tang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha 410008, China; (J.R.); (C.L.); (M.Z.); (H.W.)
- Aging Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
- The Biobank of Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
8
|
Talukdar S, Emdad L, Das SK, Fisher PB. GAP junctions: multifaceted regulators of neuronal differentiation. Tissue Barriers 2021; 10:1982349. [PMID: 34651545 DOI: 10.1080/21688370.2021.1982349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Gap junctions are intercellular membrane channels consisting of connexin proteins, which contribute to direct cytoplasmic exchange of small molecules, substrates and metabolites between adjacent cells. These channels play important roles in neuronal differentiation, maintenance, survival and function. Gap junctions regulate differentiation of neurons from embryonic, neural and induced pluripotent stem cells. In addition, they control transdifferentiation of neurons from mesenchymal stem cells. The expression and levels of several connexins correlate with cell cycle changes and different stages of neurogenesis. Connexins such as Cx36, Cx45, and Cx26, play a crucial role in neuronal function. Several connexin knockout mice display lethal or severely impaired phenotypes. Aberrations in connexin expression is frequently associated with various neurodegenerative disorders. Gap junctions also act as promising therapeutic targets for neuronal regenerative medicine, because of their role in neural stem cell integration, injury and remyelination.
Collapse
Affiliation(s)
- Sarmistha Talukdar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States.,Vcu Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States.,Vcu Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.,Vcu Institute of Molecular Medicine, Richmond, VA, United States.,Vcu Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| |
Collapse
|
9
|
Babaie A, Bakhshandeh B, Abedi A, Mohammadnejad J, Shabani I, Ardeshirylajimi A, Reza Moosavi S, Amini J, Tayebi L. Synergistic effects of conductive PVA/PEDOT electrospun scaffolds and electrical stimulation for more effective neural tissue engineering. Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.110051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
10
|
Tu H, Chu H, Guan S, Hao F, Xu N, Zhao Z, Liang Y. The role of the M1/M2 microglia in the process from cancer pain to morphine tolerance. Tissue Cell 2020; 68:101438. [PMID: 33220596 DOI: 10.1016/j.tice.2020.101438] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/29/2022]
Abstract
Cancer pain, especially bone cancer pain, is a pain state often caused by inflammation or dysfunctional nerves. Moreover, in the management of cancer pain, opioid especially morphine is widely used, however, it also brings severe side effects such as morphine tolerance to the patient (Deandrea et al., 2008). A growing body of literatures demonstrated that neuroinflammation is mediated by microglia. As the macrophages like immune cells, microglia play an important role in the pathogenesis of cancer pain and morphine tolerance. Microglia acquire different activation states to regulate the function of these cells. As to M1 phenotype, microglia release pro-inflammatory cytokines and neurotoxic molecules that promote inflammation and cytotoxic reactions. Conversely, when microglia represent M2 phenotypes secreting anti-inflammatory cytokines and nutrient factors that promote the function of repair, regeneration and restore homeostasis. A better understanding of microglia activation in cancer pain and morphine tolerance is crucial for the development of hypothesized neuroprotective drugs. Targeting microglia different polarization states by the inhibition of their deleterious pro-inflammatory neurotoxicity and/or enhancing their beneficial anti-inflammatory protective function seems to be an effective treatment for cancer pain and morphine tolerance.
Collapse
Affiliation(s)
- Houan Tu
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, 6 Tongfu Road, Qingdao, Shandong 266034, China
| | - Haichen Chu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Sen Guan
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, 6 Tongfu Road, Qingdao, Shandong 266034, China
| | - Fengxi Hao
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Na Xu
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Zhiping Zhao
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, 59 Hai Er Road, Qingdao, Shandong 266061, China
| | - Yongxin Liang
- Department of Anesthesiology, Women's and Children's Hospital Affiliated to Qingdao University, 6 Tongfu Road, Qingdao, Shandong 266034, China.
| |
Collapse
|
11
|
Jin T, Gu J, Xia H, Chen H, Xu X, Li Z, Yue Y, Gui Y. Differential Expression of microRNA Profiles and Wnt Signals in Stem Cell-Derived Exosomes During Dopaminergic Neuron Differentiation. DNA Cell Biol 2020; 39:2143-2153. [PMID: 33064572 DOI: 10.1089/dna.2020.5931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The role of secreted exosomes during dopaminergic (DA) neuron differentiation is still unknown. To investigate the roles of exosomes in DA neuron fate specification, we profiled exosomal microRNAs (miRNAs) during DA neuron differentiation of epiblast-derived stem cells (EpiSCs). There were 26 miRNAs differentially expressed (relative fold >2, p < 0.05) in EpiSC-derived exosomes at 0, 2, 4, 6, 8, 10, 12, and 14 days of DA epiblast differentiation. Among them, 23 exosomic miRNAs were significantly increased, including miR-124, miR-132, miR-133b, miR-218, miR-9, miR-34b, miR-34c, and miR-135a2, while three exosomic miRNAs (miR-214, miR-7a, and miR-302b) were decreased, when compared with control samples. Bioinformatics analysis by DIANA-mirPath demonstrated that extracellular matrix-receptor interaction, signaling pathways regulating pluripotency of stem cells, FoxO signaling pathway, DA synapse, Wnt signaling pathway, GABAergic synapse, and neurotrophin signaling pathway were significantly enriched in DA differentiation-related miRNA signature (all p-values <0.012). Furthermore, messenger RNAs for nine DA neuronal markers tyrosine hydroxylase (TH), Nr4a2, Pitx3, Drd1a, Lmx1a, Lmx1b, Foxa1, Dmrt5, and Slc18a2 were significantly increased expressed over time in exosomes derived from differentiated EpiSCs. Interestingly, adding with exosomes derived from EpiSC induction experiment resulted in a twofold increase of TH-positive neurons production (35% vs. 17%, p < 0.01) during DA neuronal differentiation from mouse embryonic stem cells (ESCs). In summary, our results suggested exosomal miRNAs are potential regulators of DA neuron differentiation. More importantly, EpiSC-derived exosomes could promote the generation of DA neuron differentiation from ESCs.
Collapse
Affiliation(s)
- Tao Jin
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiachen Gu
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongbo Xia
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Neurology, The First People's Hospital of Fuyang, Hangzhou, China
| | - Huimin Chen
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Neurology, School of Medicine, Shaoxing University, Shaoxing, China
| | - Xiaomin Xu
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zongshan Li
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yumei Yue
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yaxing Gui
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Isolation, Culture, and Functional Characterization of Human Embryonic Stem Cells: Current Trends and Challenges. Stem Cells Int 2018; 2018:1429351. [PMID: 30254679 PMCID: PMC6142731 DOI: 10.1155/2018/1429351] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/19/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022] Open
Abstract
Human embryonic stem cells (hESCs) hold great potential for the treatment of various degenerative diseases. Pluripotent hESCs have a great ability to undergo unlimited self-renewal in culture and to differentiate into all cell types in the body. The journey of hESC research is not that smooth, as it has faced several challenges which are limited to not only tumor formation and immunorejection but also social, ethical, and political aspects. The isolation of hESCs from the human embryo is considered highly objectionable as it requires the destruction of the human embryo. The issue was debated and discussed in both public and government platforms, which led to banning of hESC research in many countries around the world. The banning has negatively affected the progress of hESC research as many federal governments around the world stopped research funding. Afterward, some countries lifted the ban and allowed the funding in hESC research, but the damage has already been done on the progress of research. Under these unfavorable conditions, still some progress was made to isolate, culture, and characterize hESCs using different strategies. In this review, we have summarized various strategies used to successfully isolate, culture, and characterize hESCs. Finally, hESCs hold a great promise for clinical applications with proper strategies to minimize the teratoma formation and immunorejection and better cell transplantation strategies.
Collapse
|
13
|
Joshi R, Fuller B, Mosadegh B, Tavana H. Stem cell colony interspacing effect on differentiation to neural cells. J Tissue Eng Regen Med 2018; 12:2041-2054. [PMID: 30058271 DOI: 10.1002/term.2739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/20/2018] [Accepted: 07/11/2018] [Indexed: 01/30/2023]
Abstract
Efforts to enhance the efficiency of neural differentiation of stem cells are primarily focused on exogenous modulation of physical niche parameters such as surface topography and extracellular matrix proteins, or addition of certain growth factors or small molecules to culture media. We report a novel neurogenic niche to enhance the neural differentiation of embryonic stem cells (ESCs) without any external intervention by micropatterning ESCs into spatially organized colonies of controlled size and interspacing. Using an aqueous two-phase system cell microprinting technology, we generated pairs of uniformly sized isolated ESC colonies at defined interspacing distances over a layer of differentiation-inducing stromal cells. Our comprehensive analysis of temporal expression of neural genes and proteins of cells in colony pairs showed that interspacing two colonies at approximately 0.66 times the colony diameter (0.66D) significantly enhanced neural differentiation of ESCs. Cells in these colonies displayed higher expression of neural genes and proteins and formed thick neurite bundles between the two colonies. A computational model of spatial distribution of soluble factors of cells in interspaced colony pairs showed that the enhanced neural differentiation is due to the presence of stable concentration gradients of soluble signalling factors between the two colonies. Our results indicate that culturing ESCs in colony pairs with defined interspacing is a promising approach to efficiently derive neural cells. Additionally, this approach provides a platform for quantitative studies of molecular mechanisms that regulate neurogenesis of stem cells.
Collapse
Affiliation(s)
- Ramila Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio
| | - Brendan Fuller
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio
| | - Bobak Mosadegh
- Department of Radiology, Dalio Institute of Cardiovascular Imaging, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, New York
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio
| |
Collapse
|
14
|
Lai SW, Chen JH, Lin HY, Liu YS, Tsai CF, Chang PC, Lu DY, Lin C. Regulatory Effects of Neuroinflammatory Responses Through Brain-Derived Neurotrophic Factor Signaling in Microglial Cells. Mol Neurobiol 2018; 55:7487-7499. [PMID: 29427085 DOI: 10.1007/s12035-018-0933-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/25/2018] [Indexed: 11/26/2022]
Abstract
Inhibition of microglial over-activation is an important strategy to counter balance neurodegenerative progression. We previously demonstrated that the adenosine monophosphate-activated protein kinase (AMPK) may be a therapeutic target in mediating anti-neuroinflammatory responses in microglia. Brain-derived neurotrophic factor (BDNF) is one of the major neurotrophic factors produced by astrocytes to maintain the development and survival of neurons in the brain, and have recently been shown to modulate homeostasis of neuroinflammation. Therefore, the present study focused on BDNF-mediated neuroinflammatory responses and may provide an endogenous regulation of neuroinflammation. Among the tested neuroinflammation, epigallocatechin gallate (EGCG) and minocycline exerted BDNF upregulation to inhibit COX-2 and proinflammatory mediator expressions. Furthermore, both EGCG and minocycline upregulated BDNF expression in microglia through AMPK signaling. In addition, minocycline and EGCG also increased expressions of erythropoietin (EPO) and sonic hedgehog (Shh). In the endogenous modulation of neuroinflammation, astrocyte-conditioned medium (AgCM) also decreased the expression of COX-2 and upregulated BDNF expression in microglia. The anti-inflammatory effects of BDNF were mediated through EPO/Shh in microglia. Our results indicated that the BDNF-EPO-Shh novel-signaling pathway underlies the regulation of inflammatory responses and may be regarded as a potential therapeutic target in neurodegenerative diseases. This study also reveals a better understanding of an endogenous crosstalk between astrocytes and microglia to regulate anti-inflammatory actions, which could provide a novel strategy for the treatment of neuroinflammation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sheng-Wei Lai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Hsiao-Yun Lin
- Department of Pharmacology, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
| | - Yu-Shu Liu
- Department of Pharmacology, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, Taiwan.
- Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
15
|
Joshi R, Buchanan JC, Tavana H. Self-regulatory factors of embryonic stem cells in co-culture with stromal cells enhance neural differentiation. Integr Biol (Camb) 2017; 9:418-426. [PMID: 28406502 PMCID: PMC5498101 DOI: 10.1039/c7ib00038c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Embryonic stem cells (ESCs), due to their intrinsic capability to generate somatic cells of all three germ layers, are potential sources of neural cells for cell replacement therapies. However, the empirical differentiation protocols and the lack of mechanistic understanding of the neural differentiation of ESCs have limited the utility of ESCs as a developmental model or as a cell source for neural cell populations for replacement therapies. Co-culturing ESCs with stromal cells is one of the extensively used methods to induce neural differentiation. Despite several studies to identify neural inducing factors in stromal cell induced neural differentiation, the self-regulatory effects of ESCs in the neural differentiation process remain unexplored. For the first time, we elucidate the self-regulatory role of mESCs in their neural cell differentiation by supplementing conditioned media from differentiating mESCs to mESC-PA6 co-cultures and quantitatively evaluating the change in neural differentiation. Moreover, we use statistical tools to analyze the expression of various growth and trophic factors and distinguish the factors produced primarily by PA6 cells versus mESCs in co-culture. We observe that addition of the medium containing mESC-secreted factors to a single mESC colony co-cultured with PA6 cells significantly enhances the neural differentiation of mESCs compares to the medium extracted from the stromal cells only. Hierarchical clustering of gene expression data from PA6 and co-cultured mESCs segregates two groups of factors that are produced by the stromal cells and differentiating mESCs. Identifying the major soluble factors that drive and regulate the neural differentiation process in the mESC-PA6 co-culture niche will help understand molecular mechanisms of neural development. Moreover, it can be a major step toward developing novel protocols to differentiate stem cells with mESC derived factor supplementation without using feeder cells and with greater efficiency compared to existing approaches.
Collapse
Affiliation(s)
- R. Joshi
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, USA
| | - J. C. Buchanan
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, USA
| | - H. Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, Ohio 44325, USA
| |
Collapse
|
16
|
Pires AO, Teixeira FG, Mendes-Pinheiro B, Serra SC, Sousa N, Salgado AJ. Old and new challenges in Parkinson's disease therapeutics. Prog Neurobiol 2017; 156:69-89. [PMID: 28457671 DOI: 10.1016/j.pneurobio.2017.04.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 03/15/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic neurons and/or loss od neuronal projections, in several dopaminergic networks. Current treatments for idiopathic PD rely mainly on the use of pharmacologic agents to improve motor symptomatology of PD patients. Nevertheless, so far PD remains an incurable disease. Therefore, it is of utmost importance to establish new therapeutic strategies for PD treatment. Over the last 20 years, several molecular, gene and cell/stem-cell therapeutic approaches have been developed with the aim of counteracting or retarding PD progression. The scope of this review is to provide an overview of PD related therapies and major breakthroughs achieved within this field. In order to do so, this review will start by focusing on PD characterization and current treatment options covering thereafter molecular, gene and cell/stem cell-based therapies that are currently being studied in animal models of PD or have recently been tested in clinical trials. Among stem cell-based therapies, those using MSCs as possible disease modifying agents for PD therapy and, specifically, the MSCs secretome contribution to meet the clinical challenge of counteracting or retarding PD progression, will be more deeply explored.
Collapse
Affiliation(s)
- Ana O Pires
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - F G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - B Mendes-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Sofia C Serra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
17
|
Xia N, Fang F, Zhang P, Cui J, Tep-Cullison C, Hamerley T, Lee HJ, Palmer T, Bothner B, Lee JH, Pera RR. A Knockin Reporter Allows Purification and Characterization of mDA Neurons from Heterogeneous Populations. Cell Rep 2017; 18:2533-2546. [PMID: 28273465 DOI: 10.1016/j.celrep.2017.02.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/07/2016] [Accepted: 02/06/2017] [Indexed: 12/26/2022] Open
Abstract
Generation of midbrain dopaminergic (mDA) neurons from human pluripotent stem cells provides a platform for inquiry into basic and translational studies of Parkinson's disease (PD). However, heterogeneity in differentiation in vitro makes it difficult to identify mDA neurons in culture or in vivo following transplantation. Here, we report the generation of a human embryonic stem cell (hESC) line with a tyrosine hydroxylase (TH)-RFP (red fluorescent protein) reporter. We validated that RFP faithfully mimicked TH expression during differentiation. Use of this TH-RFP reporter cell line enabled purification of mDA-like neurons from heterogeneous cultures with subsequent characterization of neuron transcriptional and epigenetic programs (global binding profiles of H3K27ac, H3K4me1, and 5-hydroxymethylcytosine [5hmC]) at four different stages of development. We anticipate that the tools and data described here will contribute to the development of mDA neurons for applications in disease modeling and/or drug screening and cell replacement therapies for PD.
Collapse
Affiliation(s)
- Ninuo Xia
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT 59717, USA; Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Fang Fang
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT 59717, USA; Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Pengbo Zhang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jun Cui
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT 59717, USA; Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chhavy Tep-Cullison
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tim Hamerley
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Hyun Joo Lee
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Theo Palmer
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Jin Hyung Lee
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Renee Reijo Pera
- Department of Cell Biology and Neurosciences, Montana State University, Bozeman, MT 59717, USA; Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
18
|
Modeling Axonal Defects in Hereditary Spastic Paraplegia with Human Pluripotent Stem Cells. ACTA ACUST UNITED AC 2016; 11:339-354. [PMID: 27956894 DOI: 10.1007/s11515-016-1416-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cortical motor neurons, also known as upper motor neurons, are large projection neurons whose axons convey signals to lower motor neurons to control the muscle movements. Degeneration of cortical motor neuron axons is implicated in several debilitating disorders, including hereditary spastic paraplegia (HSP) and amyotrophic lateral sclerosis (ALS). Since the discovery of the first HSP gene, SPAST that encodes spastin, over 70 distinct genetic loci associated with HSP have been identified. How the mutations of these functionally diverse genes result in axonal degeneration and why certain axons are affected in HSP remains largely unknown. The development of induced pluripotent stem cell (iPSC) technology has provided researchers an excellent resource to generate patient-specific human neurons to model human neuropathologic processes including axonal defects. METHODS In this article, we will frst review the pathology and pathways affected in the common forms of HSP subtypes by searching the PubMed database. We will then summurize the findings and insights gained from studies using iPSC-based models, and discuss the challenges and future directions. RESULTS HSPs, a heterogeneous group of genetic neurodegenerative disorders, are characterized by lower extremity weakness and spasticity that result from retrograde axonal degeneration of cortical motor neurons. Recently, iPSCs have been generated from several common forms of HSP including SPG4, SPG3A, and SPG11 patients. Neurons derived from HSP iPSCs exhibit disease-relevant axonal defects, such as impaired neurite outgrowth, increased axonal swellings, and reduced axonal transport. CONCLUSION These patient-derived neurons offer unique tools to study the pathogenic mechanisms and explore the treatments for rescuing axonal defects in HSP, as well as other diseases involving axonopathy.
Collapse
|
19
|
The Preclinical Research Progress of Stem Cells Therapy in Parkinson's Disease. BIOMED RESEARCH INTERNATIONAL 2016; 2016:5683097. [PMID: 27379248 PMCID: PMC4917676 DOI: 10.1155/2016/5683097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 04/21/2016] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a type of degenerative disorder of the basal ganglia, causing tremor at rest, muscle rigidity hypokinesia, and dementia. The effectiveness of drug treatments gradually diminishes because the conversion to dopamine within the brain is increasingly disrupted by the progressive degeneration of the dopaminergic terminals. After long-term treatment, most patients with PD suffer from disability that cannot be satisfactorily controlled. To solve these issues, stem cells have recently been used for cell therapy of PD. In this review, the characteristics of different stem cells and their therapeutic effects on PD treatment will be discussed.
Collapse
|
20
|
Xu X, Huang J, Li J, Liu L, Han C, Shen Y, Zhang G, Jiang H, Lin Z, Xiong N, Wang T. Induced pluripotent stem cells and Parkinson's disease: modelling and treatment. Cell Prolif 2016; 49:14-26. [PMID: 26748765 DOI: 10.1111/cpr.12229] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/23/2015] [Indexed: 02/06/2023] Open
Abstract
Many neurodegenerative disorders, such as Parkinson's disease (PD), are characterized by progressive neuronal loss in different regions of the central nervous system, contributing to brain dysfunction in the relevant patients. Stem cell therapy holds great promise for PD patients, including with foetal ventral mesencephalic cells, human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Moreover, stem cells can be used to model neurodegenerative diseases in order to screen potential medication and explore their mechanisms of disease. However, related ethical issues, immunological rejection and lack of canonical grafting protocols limit common clinical use of stem cells. iPSCs, derived from reprogrammed somatic cells, provide new hope for cell replacement therapy. In this review, recent development in stem cell treatment for PD, using hiPSCs, as well as the potential value of hiPSCs in modelling for PD, have been summarized for application of iPSCs technology to clinical translation for PD treatment.
Collapse
Affiliation(s)
- Xiaoyun Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jie Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Han
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Shen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guoxin Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haiyang Jiang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Division of Alcohol and Drug Abuse, Mailman Neuroscience Research Center, McLean Hospital, Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
21
|
Xu L, He D, Bai Y. Microglia-Mediated Inflammation and Neurodegenerative Disease. Mol Neurobiol 2015; 53:6709-6715. [PMID: 26659872 DOI: 10.1007/s12035-015-9593-4] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023]
Abstract
Microglia are the main effectors in the inflammatory process of the central nervous system. As the first line of defense, microglia play an important role in the inflammatory reaction. When there is pathogen invasion or cell debris, microglia will be activated rapidly and remove it, while releasing the inflammatory cytokines to mediate inflammatory reaction. Activated microglia were found surrounding lesions of various neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, muscular amyotrophic lateral sclerosis, and multiple sclerosis. Microglia, the effectors of neuronal degeneration and necrosis, are involved in the removal of necrotic neurons. But over activated microglia may accelerate the process of some neurodegenerative diseases. Activated microglia can release cytotoxic factor and cytokines. Some of them may cause further damage to neuron, and some of them can regulate inflammatory cells to gather to the lesion. Microglia-mediated inflammation was considered to be the possible mechanism for the occurrence or deterioration of neurodegenerative diseases. Therefore, inhibiting the activity of microglia appropriately may be an effective way for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ling Xu
- The Department of Neurology, Xin Hua Hospital Affiliated Dalian University, Dalian University, Dalian, 116021, China
| | - Dan He
- Department of Cardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116022, China
| | - Ying Bai
- The Department of Neurology, Xin Hua Hospital Affiliated Dalian University, Dalian University, Dalian, 116021, China.
| |
Collapse
|
22
|
Cherry JF, Bennett NK, Schachner M, Moghe PV. Engineered N-cadherin and L1 biomimetic substrates concertedly promote neuronal differentiation, neurite extension and neuroprotection of human neural stem cells. Acta Biomater 2014; 10:4113-26. [PMID: 24914828 DOI: 10.1016/j.actbio.2014.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/26/2014] [Accepted: 06/01/2014] [Indexed: 02/05/2023]
Abstract
We investigated the design of neurotrophic biomaterial constructs for human neural stem cells, guided by neural developmental cues of N-cadherin and L1 adhesion molecules. Polymer substrates fabricated either as two-dimensional (2-D) films or three-dimensional (3-D) microfibrous scaffolds were functionalized with fusion chimeras of N-cadherin-Fc alone and in combination with L1-Fc, and the effects on differentiation, neurite extension and survival of H9 human-embryonic-stem-cell-derived neural stem cells (H9-NSCs) were quantified. Combinations of N-cadherin and L1-Fc co-operatively enhanced neuronal differentiation profiles, indicating the critical nature of the two complementary developmental cues. Notably, substrates presenting low levels of N-cadherin-Fc concentrations, combined with proportionately higher L1-Fc concentration, most enhanced neurite outgrowth and the degree of MAP2+ and neurofilament-M+ H9-NSCs. Low N-cadherin-Fc alone promoted improved cell survival following oxidative stress, compared to higher concentrations of N-cadherin-Fc alone or combinations with L1-Fc. Pharmacological and antibody blockage studies revealed that substrates presenting low levels of N-cadherin are functionally competent so long as they elicit a threshold signal mediated by homophilic N-cadherin and fibroblast growth factor signaling. Overall, these studies highlight the ability of optimal combinations of N-cadherin and L1 to recapitulate a "neurotrophic" microenvironment that enhances human neural stem cell differentiation and neurite outgrowth. Additionally, 3-D fibrous scaffolds presenting low N-cadherin-Fc further enhanced the survival of H9-NSCs compared to equivalent 2-D films. This indicates that similar biofunctionalization approaches based on N-cadherin and L1 can be translated to 3-D "transplantable" scaffolds with enhanced neurotrophic behaviors. Thus, the insights from this study have fundamental and translational impacts for neural-stem-cell-based regenerative medicine.
Collapse
Affiliation(s)
- Jocie F Cherry
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Neal K Bennett
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Melitta Schachner
- W.M. Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA; Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou 515041, People's Republic of China
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA; Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
23
|
Zhang P, Xia N, Reijo Pera RA. Directed dopaminergic neuron differentiation from human pluripotent stem cells. J Vis Exp 2014:51737. [PMID: 25285746 DOI: 10.3791/51737] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dopaminergic (DA) neurons in the substantia nigra pars compacta (also known as A9 DA neurons) are the specific cell type that is lost in Parkinson's disease (PD). There is great interest in deriving A9 DA neurons from human pluripotent stem cells (hPSCs) for regenerative cell replacement therapy for PD. During neural development, A9 DA neurons originate from the floor plate (FP) precursors located at the ventral midline of the central nervous system. Here, we optimized the culture conditions for the stepwise differentiation of hPSCs to A9 DA neurons, which mimics embryonic DA neuron development. In our protocol, we first describe the efficient generation of FP precursor cells from hPSCs using a small molecule method, and then convert the FP cells to A9 DA neurons, which could be maintained in vitro for several months. This efficient, repeatable and controllable protocol works well in human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) from normal persons and PD patients, in which one could derive A9 DA neurons to perform in vitro disease modeling and drug screening and in vivo cell transplantation therapy for PD.
Collapse
Affiliation(s)
- Pengbo Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine;
| | - Ninuo Xia
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine
| | - Renee A Reijo Pera
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine; Department of Obstetrics and Gynecology, Stanford University School of Medicine
| |
Collapse
|
24
|
Cell based therapies in Parkinson's Disease. Ann Neurosci 2014; 18:76-83. [PMID: 25205926 PMCID: PMC4117039 DOI: 10.5214/ans.0972.7531.1118209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 04/09/2011] [Accepted: 04/30/2011] [Indexed: 12/27/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder after Alzheimer’s disease. It is characterized by bradykinesia, hypokinesia/ akinesia, rigidity, tremor, and postural instability, caused by dopaminergic (DA) striatal denervation. The prevalence of PD increases from 50 years of age with steep rise after age 60 years. Current treatment of PD relies heavily on replacing lost dopamine either with its precursor L-dopa or dopamine agonists (ropinirole, pramipexole, bromocriptine, lisuride etc). Other pharmacological measures like catechol-O-methyltrasferase (COMT) inhibitors like entacopone, telcapone and monoamine oxidase B (MAO-B) inhibitors like selegiline and rasagiline are also useful, while L-dopa remains the gold standard in the treatment of PD. Emerging therapies are focusing on cell based therapeutics derived from various sources.
Collapse
|
25
|
Hsieh WT, Chiang BH. A well-refined in vitro model derived from human embryonic stem cell for screening phytochemicals with midbrain dopaminergic differentiation-boosting potential for improving Parkinson's disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:6326-6336. [PMID: 24933592 DOI: 10.1021/jf501640a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Stimulation of endogenous neurogenesis is a potential approach to compensate for loss of dopaminergic neurons of substantia nigra compacta nigra (SNpc) in patients with Parkinson's disease (PD). This objective was to establish an in vitro model by differentiating pluripotent human embryonic stem cells (hESCs) into midbrain dopaminergic (mDA) neurons for screening phytochemicals with mDA neurogenesis-boosting potentials. Consequently, a five-stage differentiation process was developed. The derived cells expressed many mDA markers including tyrosine hydroxylase (TH), β-III tubulin, and dopamine transporter (DAT). The voltage-gated ion channels and dopamine release were also examined for verifying neuron function, and the dopamine receptor agonists bromocriptine and 7-hydroxy-2-(dipropylamino)tetralin (7-OH-DPAT) were used to validate our model. Then, several potential phytochemicals including green tea catechins and ginsenosides were tested using the model. Finally, ginsenoside Rb1 was identified as the most potent phytochemical which is capable of upregulating neurotrophin expression and inducing mDA differentiation.
Collapse
Affiliation(s)
- Wen-Ting Hsieh
- Institute of Food Science and Technology, National Taiwan University , No. 1, Roosevelt Road, Section 4, Taipei, Taiwan ROC
| | | |
Collapse
|
26
|
Young GT, Gutteridge A, Fox HDE, Wilbrey AL, Cao L, Cho LT, Brown AR, Benn CL, Kammonen LR, Friedman JH, Bictash M, Whiting P, Bilsland JG, Stevens EB. Characterizing human stem cell-derived sensory neurons at the single-cell level reveals their ion channel expression and utility in pain research. Mol Ther 2014; 22:1530-1543. [PMID: 24832007 PMCID: PMC4435594 DOI: 10.1038/mt.2014.86] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 05/02/2014] [Indexed: 12/25/2022] Open
Abstract
The generation of human sensory neurons by directed differentiation of pluripotent stem cells opens new opportunities for investigating the biology of pain. The inability to generate this cell type has meant that up until now their study has been reliant on the use of rodent models. Here, we use a combination of population and single-cell techniques to perform a detailed molecular, electrophysiological, and pharmacological phenotyping of sensory neurons derived from human embryonic stem cells. We describe the evolution of cell populations over 6 weeks of directed differentiation; a process that results in the generation of a largely homogeneous population of neurons that are both molecularly and functionally comparable to human sensory neurons derived from mature dorsal root ganglia. This work opens the prospect of using pluripotent stem-cell–derived sensory neurons to study human neuronal physiology and as in vitro models for drug discovery in pain and sensory disorders.
Collapse
Affiliation(s)
| | | | - Heather DE Fox
- Pfizer Neusentis, Cambridge, UK; Current address: Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | - Julia H Friedman
- Oncology Research Unit, Pfizer Global Research and Development, Pearl River, NY, USA
| | | | | | | | | |
Collapse
|
27
|
Binan L, Ajji A, De Crescenzo G, Jolicoeur M. Approaches for Neural Tissue Regeneration. Stem Cell Rev Rep 2013; 10:44-59. [DOI: 10.1007/s12015-013-9474-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
28
|
Zeng X, Couture LA. Pluripotent stem cells for Parkinson's disease: progress and challenges. Stem Cell Res Ther 2013; 4:25. [PMID: 23672848 PMCID: PMC3707048 DOI: 10.1186/scrt173] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is a common debilitating neurodegenerative disease. The motor symptoms of PD are caused mainly by a progressive loss of dopaminergic neurons from the substania nigra, resulting in a loss of dopamine production. Current therapies are palliative and, in the long term, ineffective. In addition, some can result in significant clinical side effects. The relatively localized pathology of PD makes it an ideal candidate for cell replacement therapy. Initial efforts focused on fetal cell transplantation, and significant clinical benefit lasting more than 10 years has been reported in some cases. However, the approach is controversial and results have been inconsistent. Inherent limitations of this approach for widespread use are the limited availability and variability of transplant material. In contrast, the self-renewal and differentiation potential of human pluripotent stem cells (hPSCs) make them a promising alternative cell source for cell replacement therapy for PD. Efforts in the past decade have demonstrated that hPSCs can be induced to differentiate in culture to functional dopaminergic neurons. Studies in delivering these cells into PD animal models have demonstrated survival, engraftment, and behavioral deficit improvements. Several groups are developing these cells with clinical trials in mind. Here, we review the state of the technology and consider the suitability of current manufacturing processes, cell purity, and tumorgenicity for clinical testing.
Collapse
|
29
|
Belinsky GS, Sirois CL, Rich MT, Short SM, Moore AR, Gilbert SE, Antic SD. Dopamine receptors in human embryonic stem cell neurodifferentiation. Stem Cells Dev 2013; 22:1522-40. [PMID: 23286225 DOI: 10.1089/scd.2012.0150] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We tested whether dopaminergic drugs can improve the protocol for in vitro differentiation of H9 human embryonic stem cells (hESCs) into dopaminergic neurons. The expression of 5 dopamine (DA) receptor subtypes (mRNA and protein) was analyzed at each protocol stage (1, undifferentiated hESCs; 2, embryoid bodies [EBs]; 3, neuroepithelial rosettes; 4, expanding neuroepithelium; and 5, differentiating neurons) and compared to human fetal brain (gestational week 17-19). D2-like DA receptors (D2, D3, and D4) predominate over the D1-like receptors (D1 and D5) during derivation of neurons from hESCs. D1 was the receptor subtype with the lowest representation in each protocol stage (Stages 1-5). D1/D5-agonist SKF38393 and D2/D3/D4-agonist quinpirole (either alone or combined) evoked Ca(2+) responses, indicating functional receptors in hESCs. To identify when receptor activation causes a striking effect on hESC neurodifferentiation, and what ligands and endpoints are most interesting, we varied the timing, duration, and drug in the culture media. Dopaminergic agonists or antagonists were administered either early (Stages 1-3) or late (Stages 4-5). Early DA exposure resulted in more neuroepithelial colonies, more neuronal clusters, and more TH(+) clusters. The D1/D5 antagonist SKF83566 had a strong effect on EB morphology and the expression of midbrain markers. Late exposure to DA resulted in a modest increase in TH(+) neuron clusters (∼75%). The increase caused by DA did not occur in the presence of dibutyryl cAMP (dbcAMP), suggesting that DA acts through the cAMP pathway. However, a D2-antagonist (L741) decreased TH(+) cluster counts. Electrophysiological parameters of the postmitotic neurons were not significantly affected by late DA treatment (Stages 4-5). The mRNA of mature neurons (VGLUT1 and GAD1) and the midbrain markers (GIRK2, LMX1A, and MSX1) were lower in hESCs treated by DA or a D2-antagonist. When hESCs were neurodifferentiated on PA6 stromal cells, DA also increased expression of tyrosine hydroxylase. Although these results are consistent with DA's role in potentiating DA neurodifferentiation, dopaminergic treatments are generally less efficient than dbcAMP alone.
Collapse
Affiliation(s)
- Glenn S Belinsky
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Stem cells and the treatment of Parkinson's disease. Exp Neurol 2013; 260:3-11. [PMID: 23298521 DOI: 10.1016/j.expneurol.2012.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 12/18/2012] [Accepted: 12/26/2012] [Indexed: 02/06/2023]
Abstract
Progress in Parkinson's disease (PD) research has been hampered by the lack of an appropriate model which exhibits the core pathology seen in the human brain. Recent advances in deriving cells with neuronal phenotypes from patients with neurodegenerative disorders through cellular reprogramming offer a unique tool for disease modelling and may help shed light on the molecular pathogenesis that drives the progression of the disease. This technology may also help in establishing platforms for drug screening and open up exciting new prospects for cell grafting. In this review, we will discuss progress made in differentiating stem cells into authentic dopamine neurons and where we stand with respect to clinical trials with these cells in patients with PD. We will also examine the various approaches used in cellular reprogramming and their differentiation into patient-specific midbrain dopamine neurons, with an emphasis particularly on modelling familial cases of PD to recapitulate disease phenotypes. This review will highlight some of the challenges that need to be addressed for this technology to have any potential clinical application in cell therapy and personalised medicine.
Collapse
|
31
|
Needham K, Minter RL, Shepherd RK, Nayagam BA. Challenges for stem cells to functionally repair the damaged auditory nerve. Expert Opin Biol Ther 2013; 13:85-101. [PMID: 23094991 PMCID: PMC3543850 DOI: 10.1517/14712598.2013.728583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION In the auditory system, a specialized subset of sensory neurons are responsible for correctly relaying precise pitch and temporal cues to the brain. In individuals with severe-to-profound sensorineural hearing impairment these sensory auditory neurons can be directly stimulated by a cochlear implant, which restores sound input to the brainstem after the loss of hair cells. This neural prosthesis therefore depends on a residual population of functional neurons in order to function effectively. AREAS COVERED In severe cases of sensorineural hearing loss where the numbers of auditory neurons are significantly depleted, the benefits derived from a cochlear implant may be minimal. One way in which to restore function to the auditory nerve is to replace these lost neurons using differentiated stem cells, thus re-establishing the neural circuit required for cochlear implant function. Such a therapy relies on producing an appropriate population of electrophysiologically functional neurons from stem cells, and on these cells integrating and reconnecting in an appropriate manner in the deaf cochlea. EXPERT OPINION Here we review progress in the field to date, including some of the key functional features that stem cell-derived neurons would need to possess and how these might be enhanced using electrical stimulation from a cochlear implant.
Collapse
Affiliation(s)
- Karina Needham
- University of Melbourne, Department of Otolaryngology, East Melbourne, Australia.
| | | | | | | |
Collapse
|
32
|
Alwin Prem Anand A, Gowri Sankar S, Kokila Vani V. Immortalization of neuronal progenitors using SV40 large T antigen and differentiation towards dopaminergic neurons. J Cell Mol Med 2012; 16:2592-610. [PMID: 22863662 PMCID: PMC4118228 DOI: 10.1111/j.1582-4934.2012.01607.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 07/16/2012] [Indexed: 01/19/2023] Open
Abstract
Transplantation is common in clinical practice where there is availability of the tissue and organ. In the case of neurodegenerative disease such as Parkinson's disease (PD), transplantation is not possible as a result of the non-availability of tissue or organ and therefore, cell therapy is an innovation in clinical practice. However, the availability of neuronal cells for transplantation is very limited. Alternatively, immortalized neuronal progenitors could be used in treating PD. The neuronal progenitor cells can be differentiated into dopaminergic phenotype. Here in this article, the current understanding of the molecular mechanisms involved in the differentiation of dopaminergic phenotype from the neuronal progenitors immortalized with SV40 LT antigen is discussed. In addition, the methods of generating dopaminergic neurons from progenitor cells and the factors that govern their differentiation are elaborated. Recent advances in cell-therapy based transplantation in PD patients and future prospects are discussed.
Collapse
|
33
|
Politis M, Lindvall O. Clinical application of stem cell therapy in Parkinson's disease. BMC Med 2012; 10:1. [PMID: 22216957 PMCID: PMC3261810 DOI: 10.1186/1741-7015-10-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 01/04/2012] [Indexed: 01/29/2023] Open
Abstract
Cell replacement therapies in Parkinson's disease (PD) aim to provide long-lasting relief of patients' symptoms. Previous clinical trials using transplantation of human fetal ventral mesencephalic (hfVM) tissue in the striata of PD patients have provided proof-of-principle that such grafts can restore striatal dopaminergic (DA-ergic) function. The transplants survive, reinnervate the striatum, and generate adequate symptomatic relief in some patients for more than a decade following operation. However, the initial clinical trials lacked homogeneity of outcomes and were hindered by the development of troublesome graft-induced dyskinesias in a subgroup of patients. Although recent knowledge has provided insights for overcoming these obstacles, it is unlikely that transplantation of hfVM tissue will become routine treatment for PD owing to problems with tissue availability and standardization of the grafts. The main focus now is on producing DA-ergic neuroblasts for transplantation from stem cells (SCs). There is a range of emerging sources of SCs for generating a DA-ergic fate in vitro. However, the translation of these efforts in vivo currently lacks efficacy and sustainability. A successful, clinically competitive SC therapy in PD needs to produce long-lasting symptomatic relief without side effects while counteracting PD progression.
Collapse
Affiliation(s)
- Marios Politis
- Centre for Neuroscience, Department of Medicine, Imperial College London, Hammersmith Hospital, DuCane Road, London W12 0NN, UK.
| | | |
Collapse
|
34
|
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide, classically characterized by a triad of motor features: bradykinesia, rigidity and resting tremor. Neurodegeneration in PD critically involves the dopaminergic neurons of the substantia nigra pars compacta, which results in a severe reduction in dopamine levels in the dorsal striatum. However, the disease also exhibits extensive non-nigral pathology and as many non-motor as motor features. Nevertheless, owing to the relatively circumscribed nature of the nigrostriatal lesion in PD, dopaminergic cell transplantation has emerged as a potentially reparative therapy for the disease. Sources for such cells are varied and include the developing ventral mesencephalon, several autologous somatic cell types, embryonic stem cells and induced pluripotent stem cells. In this article, we review the origins of dopaminergic transplantation for PD and the emergent hunt for a suitable long-term source of transplantable dopaminergic neurons.
Collapse
Affiliation(s)
- Sean C Dyson
- Cambridge University Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.
| | | |
Collapse
|
35
|
Kim J, Kim JS, Kim HE, Jeon YJ, Kim DW, Soh Y, Seo KS, Lee HK, Choi NJ, Chung HM, Lee DS, Chae JI. Proteomic analysis of phosphotyrosyl proteins in human embryonic stem cell-derived neural stem cells. Neurosci Lett 2011; 499:158-63. [PMID: 21640791 DOI: 10.1016/j.neulet.2011.05.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 05/04/2011] [Accepted: 05/17/2011] [Indexed: 01/07/2023]
Abstract
Phosphorylation can reveal essential cell functions, such as cell differentiation, signal transduction, metabolic maintenance and cell division. The aim of this study was to investigate phosphorylated protein expression changes during neuronal lineage differentiation from hESCs. To measure the phosphorylated protein expression change during neuronal differentiation, we performed a comparative phosphoproteome analysis using 2-DE after MALDI-TOF MS and an MS/MS protein identification method, making a comparison between neural lineage differentiating cells and normal embryoid bodies (EBs) differentiated from human embryonic stem cells (hESCs) and profiling constituent phosphorylated proteins. Of 36 differentially expressed protein spots, 12 spots were shown to be up-regulated in differentiating neural cells. Specifically, the 7 up-regulated proteins of the 12 have potential roles in neuronal differentiation or neuronal damage recovery, including ACTB, heterogeneous nuclear ribonucleoprotein A2B1 (hnRNP A2B1), heterogeneous nuclear ribonucleoprotein L (hnRNP L), SET, chaperonin-containing TCP-1, vimentin and voltage-dependent anion channel protein 1 (VDAC1). These proteins are discussed further below.
Collapse
Affiliation(s)
- Jumi Kim
- CHA Bio & Diostech Co., Ltd., 606-16 Yeoksam 1 dong, Gangnam gu, Seoul 135-907, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Morizane A, Darsalia V, Guloglu MO, Hjalt T, Carta M, Li JY, Brundin P. A simple method for large-scale generation of dopamine neurons from human embryonic stem cells. J Neurosci Res 2011; 88:3467-78. [PMID: 20981866 DOI: 10.1002/jnr.22515] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dopamine (DA) neurons derived from human embryonic stem cells (hESCs) are potentially valuable in drug screening and as a possible source of donor tissue for transplantation in Parkinson's disease. However, existing culture protocols that promote the differentiation of DA neurons from hESCs are complex, involving multiple steps and having unreliable results between cultures. Here we report a simple and highly reproducible culture protocol that induces expandable DA neuron progenitors from hESCs in attached cultures. We found that the hESC-derived neuronal progenitors retain their full capacity to generate DA neurons after repeated passaging in the presence of basic fibroblast growth factor (bFGF) and medium conditioned with PA6 stromal cells. Using immunocytochemistry and RT-PCR, we found that the differentiated DA neurons exhibit a midbrain phenotype and express, e.g., Aldh1a, Ptx3, Nurr1, and Lmx1a. Using HPLC, we monitored their production of DA. We then demonstrated that the expanded progenitors are possible to cryopreserve without loosing the dopaminergic phenotype. With our protocol, we obtained large and homogeneous populations of dopaminergic progenitors and neurons. We conclude that our protocol can be used to generate human DA neurons suitable for the study of disease mechanisms, toxicology, drug screening, and intracerebral transplantation.
Collapse
Affiliation(s)
- Asuka Morizane
- Neuronal Survival Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | | | | | | | | | | | | |
Collapse
|
37
|
Belinsky GS, Moore AR, Short SM, Rich MT, Antic SD. Physiological properties of neurons derived from human embryonic stem cells using a dibutyryl cyclic AMP-based protocol. Stem Cells Dev 2011; 20:1733-46. [PMID: 21226567 DOI: 10.1089/scd.2010.0501] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neurons derived from human embryonic stem cells hold promise for the therapy of neurological diseases. Quality inspection of human embryonic stem cell-derived neurons has often been based on immunolabeling for neuronal markers. Here we put emphasis on their physiological properties. Electrophysiological measurements were carried out systematically at different stages of neuronal in vitro development, including the very early stage, neuroepithelial rosettes. Developing human neurons are able to generate action potentials (APs) as early as 10 days after the start of differentiation. Tyrosine hydroxylase (TH)-positive (putative dopaminergic, DA) neurons tend to aggregate into clumps, and their overall yield per coverslip is relatively low (8.3%) because of areas void of DA neurons. On the same in vitro day, neighboring neurons can be in very different stages of differentiation, including repetitive AP firing, single full-size AP, and abortive AP. Similarly, the basic electrophysiological parameters (resting membrane potential, input resistance, peak sodium, and peak potassium currents) are scattered in a wide range. Visual appearance of differentiating neurons, and number of primary and secondary dendrites cannot be used to predict the peak sodium current or AP firing properties of cultured neurons. Approximately 13% of neurons showed evidence of hyperpolarization-induced current (I(h)), a characteristic of DA neurons; however, no neurons with repetitive APs showed I(h). The electrophysiological measurements thus indicate that a standard DA differentiation (dibutyryl cyclic AMP-based) protocol, applied for 2-5 weeks, produces a heterogeneous ensemble of mostly immature neurons. The overall quality of human neurons under present conditions (survival factors were not used) begins to deteriorate after 12 days of differentiation.
Collapse
Affiliation(s)
- Glenn S Belinsky
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut 06030-3401, USA
| | | | | | | | | |
Collapse
|
38
|
Narsinh KH, Sun N, Sanchez-Freire V, Lee AS, Almeida P, Hu S, Jan T, Wilson KD, Leong D, Rosenberg J, Yao M, Robbins RC, Wu JC. Single cell transcriptional profiling reveals heterogeneity of human induced pluripotent stem cells. J Clin Invest 2011; 121:1217-21. [PMID: 21317531 DOI: 10.1172/jci44635] [Citation(s) in RCA: 222] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 12/15/2010] [Indexed: 01/03/2023] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) are promising candidate cell sources for regenerative medicine. However, despite the common ability of hiPSCs and hESCs to differentiate into all 3 germ layers, their functional equivalence at the single cell level remains to be demonstrated. Moreover, single cell heterogeneity amongst stem cell populations may underlie important cell fate decisions. Here, we used single cell analysis to resolve the gene expression profiles of 362 hiPSCs and hESCs for an array of 42 genes that characterize the pluripotent and differentiated states. Comparison between single hESCs and single hiPSCs revealed markedly more heterogeneity in gene expression levels in the hiPSCs, suggesting that hiPSCs occupy an alternate, less stable pluripotent state. hiPSCs also displayed slower growth kinetics and impaired directed differentiation as compared with hESCs. Our results suggest that caution should be exercised before assuming that hiPSCs occupy a pluripotent state equivalent to that of hESCs, particularly when producing differentiated cells for regenerative medicine aims.
Collapse
Affiliation(s)
- Kazim H Narsinh
- Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Malgrange B, Borgs L, Grobarczyk B, Purnelle A, Ernst P, Moonen G, Nguyen L. Using human pluripotent stem cells to untangle neurodegenerative disease mechanisms. Cell Mol Life Sci 2011; 68:635-49. [PMID: 20976521 PMCID: PMC11115022 DOI: 10.1007/s00018-010-0557-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/14/2010] [Accepted: 10/04/2010] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cells, including embryonic (hES) and induced pluripotent stem cells (hiPS), retain the ability to self-renew indefinitely, while maintaining the capacity to differentiate into all cell types of the nervous system. While human pluripotent cell-based therapies are unlikely to arise soon, these cells can currently be used as an inexhaustible source of committed neurons to perform high-throughput screening and safety testing of new candidate drugs. Here, we describe critically the available methods and molecular factors that are used to direct the differentiation of hES or hiPS into specific neurons. In addition, we discuss how the availability of patient-specific hiPS offers a unique opportunity to model inheritable neurodegenerative diseases and untangle their pathological mechanisms, or to validate drugs that would prevent the onset or the progression of these neurological disorders.
Collapse
|
40
|
Lam HJ, Patel S, Wang A, Chu J, Li S. In vitro regulation of neural differentiation and axon growth by growth factors and bioactive nanofibers. Tissue Eng Part A 2011; 16:2641-8. [PMID: 20367289 DOI: 10.1089/ten.tea.2009.0414] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human embryonic stem cell (ESC)-derived neural cells are a potential cell source for neural tissue regeneration. Understanding the biochemical and biophysical regulation of neural differentiation and axon growth will help us develop cell therapies and bioactive scaffolds. We demonstrated that basic fibroblast growth factor (bFGF) and epidermal growth factor (EGF) had different effects on human ESC differentiation into neural cells. EGF was more effective in inducing expression of neuron and glial markers and cell extensions. In addition to biochemical cues, poly(l-lactic acid) scaffolds with aligned nanofibers increased axon growth from ESC-derived neural cells, demonstrating the significant effects of biophysical guidance at nanoscale. To combine the biochemical and biophysical cues, bFGF and EGF were either adsorbed or bound to heparin on nanofibrous scaffolds. EGF, but not bFGF, was effectively adsorbed onto nanofibers. However, adsorbed EGF and bFGF did not effectively enhance axon growth. In contrast, immobilization of bFGF or EGF onto nanofibers using heparin as the adapter molecule significantly promoted axon growth. This study elucidated the effect of bFGF and EGF in neural differentiation and axon growth, and demonstrated a method to immobilize active bFGF and EGF onto aligned nanofibers to promote neural tissue regeneration.
Collapse
Affiliation(s)
- Hayley J Lam
- Department of Bioengineering, University of California at Berkeley, Berkeley, California 94720, USA
| | | | | | | | | |
Collapse
|
41
|
Xie X, Hiona A, Lee AS, Cao F, Huang M, Li Z, Cherry A, Pei X, Wu JC. Effects of long-term culture on human embryonic stem cell aging. Stem Cells Dev 2010; 20:127-38. [PMID: 20629482 DOI: 10.1089/scd.2009.0475] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In recent years, human embryonic stem (hES) cells have become a promising cell source for regenerative medicine. Although hES cells have the ability for unlimited self-renewal, potential adverse effects of long-term cell culture upon hES cells must be investigated before therapeutic applications of hES cells can be realized. Here we investigated changes in molecular profiles associated with young (<60 passages) and old (>120 passages) cells of the H9 hES cell line as well as young (<85 passages) and old (>120 passages) cells of the PKU1 hES cell line. Our results show that morphology, stem cell markers, and telomerase activity do not differ significantly between young and old passage cells. Cells from both age groups were also shown to differentiate into derivatives of all 3 germ layers upon spontaneous differentiation in vitro. Interestingly, mitochondrial dysfunction was found to occur with prolonged culture. Old passage cells of both the H9 and PKU1 lines were characterized by higher mitochondrial membrane potential, larger mitochondrial morphology, and higher reactive oxygen species content than their younger counterparts. Teratomas derived from higher passage cells were also found to have an uneven preference for differentiation compared with tumors derived from younger cells. These findings suggest that prolonged culture of hES cells may negatively impact mitochondrial function and possibly affect long-term pluripotency.
Collapse
Affiliation(s)
- Xiaoyan Xie
- Department of Medicine, Stanford University School of Medicine , Stanford, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hwang DY, Kim DS, Kim DW. Human ES and iPS cells as cell sources for the treatment of Parkinson's disease: current state and problems. J Cell Biochem 2010; 109:292-301. [PMID: 20014069 DOI: 10.1002/jcb.22411] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cell therapy using human embryonic stem cells (hESCs) is a promising therapeutic option for Parkinson's disease (PD), an incurable neurodegenerative disease. A prerequisite for clinical application of hESCs for PD is an efficient and strict differentiation of hESCs into midbrain dopamine (mDA) neuron-like cells, which would be directly translated into high effectiveness of the therapy with minimum risk of undesirable side effects. Due to fruitful efforts from many laboratories, a variety of strategies for improving efficiency of dopaminergic differentiation from hESCs have been developed, mostly by optimizing culture conditions, genetic modification, and modulating intracellular signaling pathways. The rapid advances in the fields of dopaminergic differentiation of hESCs, prevention of tumor formation, and establishment of safe human induced pluripotent stem cells (hiPSCs) would open the door to highly effective, tumor-free, and immune rejection-free cell therapy for PD in the near future.
Collapse
Affiliation(s)
- Dong-Youn Hwang
- Stem Cell Research Center, 21C Frontier R&D Program of Ministry of Education, Science and Technology, Yonsei University Medical Center, Seoul, South Korea.
| | | | | |
Collapse
|
43
|
Sonntag KC, Simunovic F, Sanchez-Pernaute R. Stem cells and cell replacement therapy for Parkinson's disease. JOURNAL OF NEURAL TRANSMISSION. SUPPLEMENTUM 2010:287-99. [PMID: 20411787 DOI: 10.1007/978-3-211-92660-4_24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by a progressive degeneration of the midbrain dopamine (DA) neurons in the substantia nigra pars compacta (SNc) that predominantly affects the ventral population projecting to the dorsal striatum and leads to a gradual dysfunction of the motor system. There is currently no cure for PD. Pharmacological and surgical (e.g. deep brain stimulation) interventions can alleviate some of the symptoms, but lose their efficacy over time. The distinct loss of DA neurons in the SN offers the opportunity to assay neuronal cell replacement, and the clinical transplantation of fetal midbrain neuroblasts in PD patients has shown that this approach is feasible. However, there are multiple problems associated with the use of fetus-derived material, including limited availability. DA neurons derived from stem cells (SC) represent an alternative and unlimited cell source for cell replacement therapies. Currently, human pluripotent SC, such as embryonic (ES), and most recently, induced pluripotent stem cells (iPS), and multipotent (tissue-specific) adult SC are available, although the methodology for a reliable and efficient production of DA neurons necessary for biomedical applications is still underdeveloped. Here, we discuss some essentials for SC and SC-derived DA neurons to become therapeutic agents.
Collapse
Affiliation(s)
- K-C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, MRC 223 115 Mill Street, Belmont, MA 02478, USA.
| | | | | |
Collapse
|
44
|
Anisimov SV. Cell-based therapeutic approaches for Parkinson's disease: progress and perspectives. Rev Neurosci 2010; 20:347-81. [PMID: 20397620 DOI: 10.1515/revneuro.2009.20.5-6.347] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Motor dysfunctions in Parkinson's disease are believed to be primarily due to the degeneration of dopaminergic neurons located in the substantia nigra pars compacta. Because a single-type cell population is depleted, Parkinson's disease is considered a primary target for cell replacement-based therapeutic strategies. Extensive studies have confirmed transplantation of donor neurons could be beneficial, yet identifying an alternative cell source is clearly essential. Human embryonic stem cells (hESCs) have been proposed as a renewable source of dopaminergic neurons for transplantation in Parkinson's disease; other potential sources could include neural stem cells (hNSCs) and adult mesenchymal stem cells (hMSCs). However, numerous difficulties avert practical application of stem cell-based therapeutic approaches for the treatment of Parkinson's disease. Among the latter, ethical, safety (including xeno- and tumor formation-associated risks) and technical issues stand out. This review aims to provide a balanced and updated outlook on various issues associated with stem cells in regard to their potential in the treatment of Parkinson's disease. Essential features of the individual stem cell subtypes, principles of available differentiation protocols, transplantation, and safety issues are discussed extensively.
Collapse
Affiliation(s)
- Sergey V Anisimov
- Department of Intracellular Signalling and Transport, Institute of Cytology, Russian Academy of Sciences and Research, Saint-Petersburg, Russia.
| |
Collapse
|
45
|
Swistowska AM, da Cruz AB, Han Y, Swistowski A, Liu Y, Shin S, Zhan M, Rao MS, Zeng X. Stage-specific role for shh in dopaminergic differentiation of human embryonic stem cells induced by stromal cells. Stem Cells Dev 2010; 19:71-82. [PMID: 19788370 DOI: 10.1089/scd.2009.0107] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Stromal cells have been used to induce dopaminergic differentiation of mouse, primate, and human embryonic stem cells (hESCs), but the mechanism that governs this induction is unknown. In this manuscript, we show that medium conditioned by the stromal cell line PA6 (PA6-CM) can induce dopaminergic differentiation in neural stem cells (NSCs) derived from hESCs but not directly from hESCs, indicating that soluble factors produced by PA6 cells act at the NSC stage to specify a dopaminergic fate. To identify such soluble factors, we analyzed the transcriptomes of PA6 cells, NSCs, and dopaminergic populations induced by PA6-CM from hESC-derived NSCs. We focused our analysis on growth factors expressed by PA6 and receptors expressed by NSCs, and generated a list of growth factors and receptors that are differentially expressed. Some of the growth factor/receptor pairs are categorized into the Shh, Wnt5A, TGFbeta, and IGF pathways. The expression of genes activated by these pathways in dopaminergic populations was analyzed to confirm that these signals were likely candidates for specifying dopaminergic fate. Results were verified for Shh by using perturbation agents such as cyclopamine to show that Shh is indeed one of the active agents in PA6-CM, and by showing that Shh and FGF8 can substitute for PA6-CM at the NSC induction stage. We conclude that PA6-CM can induce dopaminergic differentiation in hESCs in a stage-specific manner. Shh is likely an important soluble dopaminergic inducing factor secreted by stromal cells and acts after the neural fate determination.
Collapse
|
46
|
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: 1.2] [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
|
47
|
Meyer AK, Maisel M, Hermann A, Stirl K, Storch A. Restorative approaches in Parkinson's Disease: Which cell type wins the race? J Neurol Sci 2010; 289:93-103. [DOI: 10.1016/j.jns.2009.08.024] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
48
|
Donaldson AE, Cai J, Yang M, Iacovitti L. Human amniotic fluid stem cells do not differentiate into dopamine neurons in vitro or after transplantation in vivo. Stem Cells Dev 2009; 18:1003-12. [PMID: 19049321 DOI: 10.1089/scd.2008.0300] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although embryonic stem (ES) cells can generate dopamine (DA) neurons that are potentially useful as a cell replacement therapy in Parkinson's disease (PD), associated ethical and practical concerns remain major stumbling blocks to their eventual use in humans. In this study, we examined human amniotic fluid stem (hAFS) cells derived from routine amniocenteses for their potential to give rise to DA neurons in vitro and following transplantation into the 6-hydroxydopamine-lesioned rat brain. We show that undifferentiated hAFS cells constitutively expressed mRNAs and proteins typical of stem cells but also cell derivatives of all three germ layers, including neural progenitors/neurons (nestin, beta-tubulin III, neurofilament). Additionally, these cells expressed mRNAs of an immature DA phenotype (Lmx1a, Pitx-3, Nurr1, Aldh1a1) but not the corresponding proteins. Importantly, treatment with DA differentiation factors using a variety of protocols did not further promote the development of fully differentiated DA neurons from hAFS cells. Thus, Lmx1a, Aldh1a1, AADC, TH, and DAT proteins were not detected in hAFS cells in culture or after transplantation into the PD rat brain. Moreover, by 3 weeks after implantation, there were no surviving AFS cells in the graft, likely as a result of an acute immunorejection response, as evidenced by the abundant presence of CD11+ macrophage/microglia and reactive GFAP+ astrocytes in the host brain. Taken together, these results suggest that further studies will be needed to improve differentiation procedures in culture and to prolong cell survival in vivo if hAFS cells are to be useful as replacement cells in PD.
Collapse
Affiliation(s)
- Angela E Donaldson
- Department of Neurology, Farber Institute for the Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | |
Collapse
|
49
|
Naghdi M, Tiraihi T, Namin SAM, Arabkheradmand J. Transdifferentiation of bone marrow stromal cells into cholinergic neuronal phenotype: a potential source for cell therapy in spinal cord injury. Cytotherapy 2009; 11:137-52. [PMID: 19253075 DOI: 10.1080/14653240802716582] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND AIMS Cholinergic neurons are very important cells in spinal cord injuries because of the deficits in motor, autonomic and sensory neurons. In this study, bone marrow stromal cells (BMSC) were evaluated as a source of cholinergic neurons in a rat model of contusive spinal cord injury. METHODS BMSC were isolated from adult rats and transdifferentiated into cholinergic neuronal cells. The BMSC were pre-induced with beta-mercaptoethanol (BME), while the induction was done with nerve growth factor (NGF). Neurofilament (NF)-68, -160 and -200 immunostaining was used for evaluating the transdifferentiation of BMSC into a neuronal phenotype. NeuroD expression, a marker for neuroblast differentiation, and Oct-4 expression, a marker for stemness, were evaluated by reverse transcriptase (RT)-polymerase chain reaction (PCR). Choline acetyl transferase (ChAT) immunoreactivity was used for assessing the cholinergic neuronal phenotype. Anti-microtubule-associated protein-2 (MAP-2) and anti-synapsin I antibodies were used as markers for the tendency for synptogenesis. Finally, the induced cells were transplanted into the contused spinal cord and locomotion was evaluated with the Basso-Beattie-Bresnahan (BBB) test. RESULTS At the induction stage, there was a decline in the expression of NF-68 associated with a sustained increase in the expression of NF-200, NF-160, ChAT and synapsin I, whereas MAP-2 expression was variable. Transplanted cells were detected 6 weeks after their injection intraspinally and were associated with functional recovery. CONCLUSIONS The transdifferentiation of BMSC into a cholinergic phenotype is feasible for replacement therapy in spinal cord injury.
Collapse
Affiliation(s)
- Majid Naghdi
- Department of Anatomical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | |
Collapse
|
50
|
Abraham S, Eroshenko N, Rao RR. Role of bioinspired polymers in determination of pluripotent stem cell fate. Regen Med 2009; 4:561-78. [PMID: 19580405 DOI: 10.2217/rme.09.23] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human pluripotent stem cells, including embryonic and induced pluripotent stem cells, hold enormous potential for the treatment of many diseases, owing to their ability to generate cell types useful for therapeutic applications. Currently, many stem cell culture propagation and differentiation systems incorporate animal-derived components for promoting self-renewal and differentiation. However, use of these components is labor intensive, carries the risk of xenogeneic contamination and yields compromised experimental results that are difficult to duplicate. From a biomaterials perspective, the generation of an animal- and cell-free biomimetic microenvironment that provides the appropriate physical and chemical cues for stem cell self-renewal or differentiation into specialized cell types would be ideal. This review presents the use of natural and synthetic polymers that support propagation and differentiation of stem cells, in an attempt to obtain a clear understanding of the factors responsible for the determination of stem cell fate.
Collapse
Affiliation(s)
- Sheena Abraham
- Department of Chemical & Life Science Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | | | | |
Collapse
|