1
|
Wu Z, Zhang J, Gao H, Li W. Astragaloside IV Treats Parkinson's Disease by Regulating the Proliferation and Differentiation of NSCs through the SHH-Nurr1 Pathway. Stem Cells Int 2024; 2024:2792909. [PMID: 39257865 PMCID: PMC11387078 DOI: 10.1155/2024/2792909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
Recently, there has been a surge of interest in enhancing the differentiation of neural stem cells (NSCs) and supplementing dopamine neurons as a potential treatment for Parkinson's disease, the second most prevalent neurodegenerative disorder. Two factors, sonic hedgehog (SHH) and nuclear receptor-related 1 protein (Nurr1), have been identified as influential in NSCs differentiation. Additionally, Astragaloside IV (AS-IV), an active compound derived from Astragalus, has also been discovered to impact NSCs differentiation. To assess the effects of AS-IV on cell activity, CCK-8 and flow cytometry techniques were employed. Meanwhile, western blotting, immunofluorescence, and real-time PCR were utilized to detect protein expression both in vivo and in vitro. Furthermore, siRNA assay was used to verify the association between SHH and Nurr1 and to investigate whether AS-IV exerts its effects through this pathway. The experimental findings revealed that AS-IV enhances cell activity and promotes the expression of differentiation proteins related to NSCs. Furthermore, the relationship between the SHH-Nurr1 pathway was confirmed, demonstrating that AS-IV induces NSCs differentiation via this pathway. Consequently, SHH, acting as the upstream signaling pathway of Nurr1, influences its expression, while AS-IV regulates the proliferation and differentiation of NSCs by modulating the SHH-Nurr1 pathway.
Collapse
Affiliation(s)
- Zicong Wu
- Encephalopathy Department Shanghai Municipal Hospital of Traditional Chinese Medicine Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Jianing Zhang
- Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Han Gao
- Encephalopathy Department Shanghai Municipal Hospital of Traditional Chinese Medicine Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Wentao Li
- Encephalopathy Department Shanghai Municipal Hospital of Traditional Chinese Medicine Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| |
Collapse
|
2
|
Jiang T, Huang J, Xu B, Ge Z, Li Y, Wei L, Yu L, Li J. Human amniotic epithelial stem cell-derived dopaminergic neuron-like cells ameliorate motor dysfunction in a rat model of Parkinson's disease. Life Sci 2024; 351:122816. [PMID: 38862064 DOI: 10.1016/j.lfs.2024.122816] [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: 02/19/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
AIMS Parkinson's disease (PD) remains a substantial clinical challenge due to the progressive loss of midbrain dopaminergic (DA) neurons in nigrostriatal pathway. In this study, human amniotic epithelial stem cells (hAESCs)-derived dopaminergic neuron-like cells (hAESCs-DNLCs) were generated, with the aim of providing new therapeutic approach to PD. MATERIALS AND METHODS hAESCs, which were isolated from discarded placentas, were induced to differentiate into hAESCs-DNLCs by following a "two stages" induction protocol. The differentiation efficiency was assessed by quantitative real-time PCR (qRT-PCR), immunocytochemistry (ICC), and ELISA. Immunogenicity, cell viability and tumorigenicity of hAESCs-DNLC were analyzed before in vivo experiments. Subsequently, hAESCs-DNLCs were transplanted into PD rats, behavioral tests were monitored after graft, and the regeneration of DA neurons was detected by immunohistochemistry (IHC). Furthermore, to trace hAESCs-DNLCs in vivo, cells were pre-labeled with PKH67 green fluorescence. KEY FINDINGS hAESCs were positive for pluripotent markers and highly expressed neural stem cells (NSCs) markers. Based on this, we established an induction method reliably generates hAESCs-DNLCs, which was evidenced by epithelium-to-neuron morphological changes, elevated expressions of neuronal and DA neuronal markers, and increased secretion of dopamine. Moreover, hAESCs-DNLCs maintained high cell viability, no tumorigenicity and low immunogenicity, suggesting hAESCs-DNLCs an attractive implant for PD therapy. Transplantation of hAESCs-DNLCs into PD rats significantly ameliorated motor disorders, as well as enhanced the reinnervation of TH+ DA neurons in nigrostriatal pathway. SIGNIFICANCE Our study has demonstrated evident therapeutic effects of hAESCs-DNLCs, and provides a safe and promising solution for PD.
Collapse
Affiliation(s)
- Tuoying Jiang
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, PR China
| | - Jianan Huang
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, PR China; Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, Zhejiang Province, PR China
| | - Bo Xu
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, PR China
| | - Zhen Ge
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310013, Zhejiang Province, PR China
| | - Yi Li
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, PR China
| | - Leiting Wei
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, PR China
| | - Luyang Yu
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, PR China.
| | - Jinying Li
- MOE Laboratory of Biosystems Homeostasis & Protection & College of Life Sciences-iCell Biotechnology Regenerative Biomedicine Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, Zhejiang Province, PR China; College of Traditional Chinese Medicine and Health Industry, Lishui University, Lishui 323000, Zhejiang Province, PR China.
| |
Collapse
|
3
|
Rybiczka-Tešulov M, Garritsen O, Venø MT, Wieg L, Dijk RV, Rahimi K, Gomes-Duarte A, Wit MD, van de Haar LL, Michels L, van Kronenburg NCH, van der Meer C, Kjems J, Vangoor VR, Pasterkamp RJ. Circular RNAs regulate neuron size and migration of midbrain dopamine neurons during development. Nat Commun 2024; 15:6773. [PMID: 39117691 PMCID: PMC11310423 DOI: 10.1038/s41467-024-51041-1] [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: 05/04/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Midbrain dopamine (mDA) neurons play an essential role in cognitive and motor behaviours and are linked to different brain disorders. However, the molecular mechanisms underlying their development, and in particular the role of non-coding RNAs (ncRNAs), remain incompletely understood. Here, we establish the transcriptomic landscape and alternative splicing patterns of circular RNAs (circRNAs) at key developmental timepoints in mouse mDA neurons in vivo using fluorescence-activated cell sorting followed by short- and long-read RNA sequencing. In situ hybridisation shows expression of several circRNAs during early mDA neuron development and post-transcriptional silencing unveils roles for different circRNAs in regulating mDA neuron morphology. Finally, in utero electroporation and time-lapse imaging implicate circRmst, a circRNA with widespread morphological effects, in the migration of developing mDA neurons in vivo. Together, these data for the first time suggest a functional role for circRNAs in developing mDA neurons and characterise poorly defined aspects of mDA neuron development.
Collapse
Affiliation(s)
- Mateja Rybiczka-Tešulov
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Oxana Garritsen
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Morten T Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
- Omiics ApS, Aarhus N, Denmark
| | - Laura Wieg
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Roland van Dijk
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Karim Rahimi
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
- Department of Genetics, Blavatnik Institute, Harvard Medical School, MA, Boston, USA
| | - Andreia Gomes-Duarte
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lieke L van de Haar
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Berlin Institute for Medical Systems Biology, Max Delbrück Center, Berlin, Germany
| | - Lars Michels
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- VectorY Therapeutics, Matrix Innovation Center VI, Amsterdam, The Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan van der Meer
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C, Denmark
| | - Vamshidhar R Vangoor
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
4
|
Lin L, Deng J, Peng J, Cui J, Wang L, Zhang M, Gao J, Li F, Shi Y, Lv M. Structural insights into the recognition of the A/T-rich motif in target gene promoters by the LMX1a homeobox domain. FEBS J 2024; 291:2792-2810. [PMID: 38465368 DOI: 10.1111/febs.17118] [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: 07/18/2023] [Revised: 01/16/2024] [Accepted: 03/01/2024] [Indexed: 03/12/2024]
Abstract
LIM homeodomain transcription factor 1-alpha (LMX1a) is a neuronal lineage-specific transcription activator that plays an essential role during the development of midbrain dopaminergic (mDA) neurons. LMX1a induces the expression of multiple key genes, which ultimately determine the morphology, physiology, and functional identity of mDA neurons. This function of LMX1a is dependent on its homeobox domain. Here, we determined the structures of the LMX1a homeobox domain in complex with the promoter sequences of the Wnt family member 1 (WNT1) or paired like homeodomain 3 (Pitx3) gene, respectively. The complex structures revealed that the LMX1a homeobox domain employed its α3 helix and an N-terminal loop to achieve specific target recognition. The N-terminal loop (loop1) interacted with the minor groove of the double-stranded DNA (dsDNA), whereas the third α-helix (α3) was tightly packed into the major groove of the dsDNA. Structure-based mutations in the α3 helix of the homeobox domain significantly reduced the binding affinity of LMX1a to dsDNA. Moreover, we identified a nonsyndromic hearing loss (NSHL)-related mutation, R199, which yielded a more flexible loop and disturbed the recognition in the minor groove of dsDNA, consistent with the molecular dynamics (MD) simulations. Furthermore, overexpression of Lmx1a promoted the differentiation of SH-SY5Y cells and upregulated the transcription of WNT1 and PITX3 genes. Hence, our work provides a detailed elucidation of the specific recognition between the LMX1a homeobox domain and its specific dsDNA targets, which represents valuable information for future investigations of the functional pathways that are controlled by LMX1a during mDA neuron development.
Collapse
Affiliation(s)
- Liqing Lin
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Jie Deng
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Junhui Peng
- Laboratory of Evolutionary Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Jing Cui
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Lei Wang
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Min Zhang
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Jia Gao
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Fudong Li
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Yunyu Shi
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| | - Mengqi Lv
- Division of Life Sciences and Medicine, Hefei National Research Center for Cross Disciplinary Science, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, China
| |
Collapse
|
5
|
Catlin JP, Tooley CES. Exploring potential developmental origins of common neurodegenerative disorders. Biochem Soc Trans 2024; 52:1035-1044. [PMID: 38661189 PMCID: PMC11440815 DOI: 10.1042/bst20230422] [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: 09/11/2023] [Revised: 03/12/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
In the United States, it is now estimated that 6.7 million people over the age of 65 are afflicted by Alzheimer's disease (AD), over 1 million people are living with Parkinson's disease (PD), and over 200 000 have or are at risk for developing Huntington's disease (HD). All three of these neurodegenerative diseases result in the ultimate death of distinct neuronal subtypes, and it is widely thought that age-related damage is the single biggest contributing factor to this neuronal death. However, recent studies are now suggesting that developmental defects during early neurogenesis could also play a role in the pathology of neurodegenerative diseases. Loss or overexpression of proteins associated with HD, PD, and AD also result in embryonic phenotypes but whether these developmental defects slowly unmask over time and contribute to age-related neurodegeneration remains highly debated. Here, we discuss known links between embryonic neurogenesis and neurodegenerative disorders (including common signaling pathways), potential compensatory mechanisms that could delay presentation of neurodegenerative disorders, and the types of model systems that could be used to study these links in vivo.
Collapse
Affiliation(s)
- James P. Catlin
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Christine E. Schaner Tooley
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
6
|
Rifes P, Kajtez J, Christiansen JR, Schörling A, Rathore GS, Wolf DA, Heuer A, Kirkeby A. Forced LMX1A expression induces dorsal neural fates and disrupts patterning of human embryonic stem cells into ventral midbrain dopaminergic neurons. Stem Cell Reports 2024; 19:830-838. [PMID: 38759646 PMCID: PMC11390620 DOI: 10.1016/j.stemcr.2024.04.010] [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: 11/24/2022] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/19/2024] Open
Abstract
The differentiation of human pluripotent stem cells into ventral mesencephalic dopaminergic (DA) fate is relevant for the treatment of Parkinson's disease. Shortcuts to obtaining DA cells through direct reprogramming often include forced expression of the transcription factor LMX1A. Although reprogramming with LMX1A can generate tyrosine hydroxylase (TH)-positive cells, their regional identity remains elusive. Using an in vitro model of early human neural tube patterning, we report that forced LMX1A expression induced a ventral-to-dorsal fate shift along the entire neuroaxis with the emergence of roof plate fates despite the presence of ventralizing molecules. The LMX1A-expressing progenitors gave rise to grafts containing roof plate-derived choroid plexus cysts as well as ectopically induced TH-positive neurons of a forebrain identity. Early activation of LMX1A prior to floor plate specification was necessary for the dorsalizing effect. Our work suggests using caution in employing LMX1A for the induction of DA fate, as this factor may generate roof plate rather than midbrain fates.
Collapse
Affiliation(s)
- Pedro Rifes
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Janko Kajtez
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Josefine Rågård Christiansen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Alrik Schörling
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark; Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; Wallenberg Center for Molecular Medicine, Department of Experimental Medical Sciences, Lund University, 22184 Lund, Sweden
| | - Gaurav Singh Rathore
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Daniel A Wolf
- Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Andreas Heuer
- Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; Behavioural Neuroscience Laboratory, Department of Experimental Medical Sciences, Lund University, 22184 Lund, Sweden
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark; Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; Wallenberg Center for Molecular Medicine, Department of Experimental Medical Sciences, Lund University, 22184 Lund, Sweden.
| |
Collapse
|
7
|
Bernal-Conde LD, Peña-Martínez V, Morato-Torres CA, Ramos-Acevedo R, Arias-Carrión Ó, Padilla-Godínez FJ, Delgado-González A, Palomero-Rivero M, Collazo-Navarrete O, Soto-Rojas LO, Gómez-Chavarín M, Schüle B, Guerra-Crespo M. Alpha-Synuclein Gene Alterations Modulate Tyrosine Hydroxylase in Human iPSC-Derived Neurons in a Parkinson's Disease Animal Model. Life (Basel) 2024; 14:728. [PMID: 38929711 PMCID: PMC11204703 DOI: 10.3390/life14060728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Parkinson's disease (PD) caused by SNCA gene triplication (3XSNCA) leads to early onset, rapid progression, and often dementia. Understanding the impact of 3XSNCA and its absence is crucial. This study investigates the differentiation of human induced pluripotent stem cell (hiPSC)-derived floor-plate progenitors into dopaminergic neurons. Three different genotypes were evaluated in this study: patient-derived hiPSCs with 3XSNCA, a gene-edited isogenic line with a frame-shift mutation on all SNCA alleles (SNCA 4KO), and a normal wild-type control. Our aim was to assess how the substantia nigra pars compacta (SNpc) microenvironment, damaged by 6-hydroxydopamine (6-OHDA), influences tyrosine hydroxylase-positive (Th+) neuron differentiation in these genetic variations. This study confirms successful in vitro differentiation into neuronal lineage in all cell lines. However, the SNCA 4KO line showed unusual LIM homeobox transcription factor 1 alpha (Lmx1a) extranuclear distribution. Crucially, both 3XSNCA and SNCA 4KO lines had reduced Th+ neuron expression, despite initial successful neuronal differentiation after two months post-transplantation. This indicates that while the SNpc environment supports early neuronal survival, SNCA gene alterations-either amplification or knock-out-negatively impact Th+ dopaminergic neuron maturation. These findings highlight SNCA's critical role in PD and underscore the value of hiPSC models in studying neurodegenerative diseases.
Collapse
Affiliation(s)
- Luis Daniel Bernal-Conde
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Verónica Peña-Martínez
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - C. Alejandra Morato-Torres
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA;
| | - Rodrigo Ramos-Acevedo
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Óscar Arias-Carrión
- Movement and Sleep Disorders Unit, Dr. Manuel Gea González General Hospital, Mexico City 14080, Mexico;
| | - Francisco J. Padilla-Godínez
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Alexa Delgado-González
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Marcela Palomero-Rivero
- Neurodevelopment and Physiology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico;
| | - Omar Collazo-Navarrete
- National Laboratory of Genomic Resources, Institute of Biomedical Research, National Autonomous University of Mexico, Mexico City 04510, Mexico;
| | - Luis O. Soto-Rojas
- Laboratory of Molecular Pathogenesis, Laboratory 4, Building A4, Medical Surgeon Career, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Mexico City 54090, Mexico;
| | - Margarita Gómez-Chavarín
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, USA;
| | - Magdalena Guerra-Crespo
- Laboratory of Regenerative Medicine, Physiology Department, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (L.D.B.-C.); (V.P.-M.); (C.A.M.-T.); (R.R.-A.); (F.J.P.-G.); (A.D.-G.); (M.G.-C.)
- Molecular Neuropathology Department, Neuroscience Division, Institute of Cell Physiology, National Autonomous University of Mexico, Mexico City 04510, Mexico
| |
Collapse
|
8
|
Umeyama T, Matsuda T, Nakashima K. Lineage Reprogramming: Genetic, Chemical, and Physical Cues for Cell Fate Conversion with a Focus on Neuronal Direct Reprogramming and Pluripotency Reprogramming. Cells 2024; 13:707. [PMID: 38667322 PMCID: PMC11049106 DOI: 10.3390/cells13080707] [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: 04/02/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Although lineage reprogramming from one cell type to another is becoming a breakthrough technology for cell-based therapy, several limitations remain to be overcome, including the low conversion efficiency and subtype specificity. To address these, many studies have been conducted using genetics, chemistry, physics, and cell biology to control transcriptional networks, signaling cascades, and epigenetic modifications during reprogramming. Here, we summarize recent advances in cellular reprogramming and discuss future directions.
Collapse
Affiliation(s)
- Taichi Umeyama
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Taito Matsuda
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | | |
Collapse
|
9
|
Zhang L, Yang H. Research progress of neural stem cells as a source of dopaminergic neurons for cell therapy in Parkinson's disease. Mol Biol Rep 2024; 51:347. [PMID: 38400887 DOI: 10.1007/s11033-024-09294-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, the most characteristic pathological feature is the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compactus (SNpc) of the mesencephalon, along with reduced dopamine content in the striatum. Researchers have been searching for drugs and therapies to treat PD in decades. However, no approach could stop the progression of the disease, and even some of them caused adverse clinical side effects. PD has a well-defined lesion. Therefore, it is considered to be one of the most curable central nervous system diseases by cell replacement treatment. Fetal ventral mesencephalic tissue transplantation has been used to treat patients with PD and obtained positive treatment results. However, ethical issues, such as limited donor tissue, and side effects including graft-induced dyskinesias, limit its clinical applications. Neural stem cell (NSC) transplantation is a viable therapy choice because it possesses multipotency, self-renewal ability, and differentiation into DA neurons, which may substitute for lost DA neurons and slow down the neurodegenerative process in PD. Studies that investigated the delivery of NSCs by using animal models of PD revealed survival, migration, and even amelioration of behavioral deficits. Here, the research progress of NSCs or NSC-derived DA neurons in treating PD was reviewed, and the practicability of present manufacturing processes for clinical testing was considered. This review is expected to offer ideas for practical strategies to solve the present technical and biological problems related to the clinical application of NSCs in PD.
Collapse
Affiliation(s)
- Lingling Zhang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, 555 East Youyi Road, Beilin District, Xi'an, 710054, China.
| | - Hao Yang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, 555 East Youyi Road, Beilin District, Xi'an, 710054, China
| |
Collapse
|
10
|
Li Y, Zhu M, Chen WX, Luo J, Li X, Cao Y, Zheng M, Ma S, Xiao Z, Zhang Y, Jiang L, Wang X, Tan T, Li X, Gong Q, Xiong X, Wang J, Tang M, Li M, Tang YP. A novel mutation in intron 1 of Wnt1 causes developmental loss of dopaminergic neurons in midbrain and ASD-like behaviors in rats. Mol Psychiatry 2023; 28:3795-3805. [PMID: 37658228 PMCID: PMC10730402 DOI: 10.1038/s41380-023-02223-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 09/03/2023]
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders with a strong genetic liability. Despite extensive studies, however, the underlying pathogenic mechanism still remains elusive. In the present study, we identified a homozygous mutation in the intron 1 of Wnt1 via large-scale screening of ASD risk/causative genes and verified that this mutation created a new splicing donor site in the intron 1, and consequently, a decrease of WNT1 expression. Interestingly, humanized rat models harboring this mutation exhibited robust ASD-like behaviors including impaired ultrasonic vocalization (USV), decreased social interactions, and restricted and repetitive behaviors. Moreover, in the substantia nigra compacta (SNpc) and the ventral tegmental area (VTA) of mutant rats, dopaminergic (DAergic) neurons were dramatically lost, together with a comparable decrease in striatal DAergic fibers. Furthermore, using single-cell RNA sequencing, we demonstrated that the decreased DAergic neurons in these midbrain areas might attribute to a shift of the boundary of the local pool of progenitor cells from the hypothalamic floor plate to the midbrain floor plate during the early embryonic stage. Moreover, treatments of mutant rats with levodopa could attenuate the impaired USV and social interactions almost completely, but not the restricted and repetitive behaviors. Our results for the first time documented that the developmental loss of DAergic neurons in the midbrain underlies the pathogenesis of ASD, and that the abnormal progenitor cell patterning is a cellular underpinning for this developmental DAergic neuronal loss. Importantly, the effective dopamine therapy suggests a translational significance in the treatment of ASD.
Collapse
Affiliation(s)
- Yongyi Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Mingwei Zhu
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wen-Xiong Chen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jing Luo
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
- School of Basic Medicine, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yangyang Cao
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Meng Zheng
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhilan Xiao
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yani Zhang
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Linyan Jiang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiumin Wang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ting Tan
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xia Li
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Qian Gong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaoli Xiong
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jun Wang
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Mingxi Tang
- Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Ya-Ping Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Department of Child Health, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
11
|
Toh HSY, Choo XY, Sun AX. Midbrain organoids-development and applications in Parkinson's disease. OXFORD OPEN NEUROSCIENCE 2023; 2:kvad009. [PMID: 38596240 PMCID: PMC10913847 DOI: 10.1093/oons/kvad009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/31/2023] [Indexed: 04/11/2024]
Abstract
Human brain development is spatially and temporally complex. Insufficient access to human brain tissue and inadequacy of animal models has limited the study of brain development and neurodegenerative diseases. Recent advancements of brain organoid technology have created novel opportunities to model human-specific neurodevelopment and brain diseases. In this review, we discuss the use of brain organoids to model the midbrain and Parkinson's disease. We critically evaluate the extent of recapitulation of PD pathology by organoids and discuss areas of future development that may lead to the model to become a next-generation, personalized therapeutic strategy for PD and beyond.
Collapse
Affiliation(s)
- Hilary S Y Toh
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
| | - Xin Yi Choo
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
| | - Alfred Xuyang Sun
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
- National Neuroscience Institute, 11 Jln Tan Tock Seng, Singapore
| |
Collapse
|
12
|
Kim W, Tripathi M, Kim C, Vardhineni S, Cha Y, Kandi SK, Feitosa M, Kholiya R, Sah E, Thakur A, Kim Y, Ko S, Bhatia K, Manohar S, Kong YB, Sindhu G, Kim YS, Cohen B, Rawat DS, Kim KS. An optimized Nurr1 agonist provides disease-modifying effects in Parkinson's disease models. Nat Commun 2023; 14:4283. [PMID: 37463889 DOI: 10.1038/s41467-023-39970-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 07/05/2023] [Indexed: 07/20/2023] Open
Abstract
The nuclear receptor, Nurr1, is critical for both the development and maintenance of midbrain dopamine neurons, representing a promising molecular target for Parkinson's disease (PD). We previously identified three Nurr1 agonists (amodiaquine, chloroquine and glafenine) that share an identical chemical scaffold, 4-amino-7-chloroquinoline (4A7C), suggesting a structure-activity relationship. Herein we report a systematic medicinal chemistry search in which over 570 4A7C-derivatives were generated and characterized. Multiple compounds enhance Nurr1's transcriptional activity, leading to identification of an optimized, brain-penetrant agonist, 4A7C-301, that exhibits robust neuroprotective effects in vitro. In addition, 4A7C-301 protects midbrain dopamine neurons in the MPTP-induced male mouse model of PD and improves both motor and non-motor olfactory deficits without dyskinesia-like behaviors. Furthermore, 4A7C-301 significantly ameliorates neuropathological abnormalities and improves motor and olfactory dysfunctions in AAV2-mediated α-synuclein-overexpressing male mouse models. These disease-modifying properties of 4A7C-301 may warrant clinical evaluation of this or analogous compounds for the treatment of patients with PD.
Collapse
Affiliation(s)
- Woori Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Mohit Tripathi
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Chunhyung Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | | | - Young Cha
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | | | - Melissa Feitosa
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Rohit Kholiya
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Eric Sah
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Anuj Thakur
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Yehan Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Sanghyeok Ko
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Kaiya Bhatia
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Sunny Manohar
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Young-Bin Kong
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Gagandeep Sindhu
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Yoon-Seong Kim
- Institute for Neurological Therapeutics, Rutgers University, Piscataway, NJ, 08854, USA
| | - Bruce Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Diwan S Rawat
- Department of Chemistry, University of Delhi, Delhi, 110007, India.
| | - Kwang-Soo Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA.
- Molecular Neurobiology Laboratory, Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
13
|
Ji X, Zhou S, Wang N, Wang J, Wu Y, Duan Y, Ni P, Zhang J, Yu S. Cerebral-Organoid-Derived Exosomes Alleviate Oxidative Stress and Promote LMX1A-Dependent Dopaminergic Differentiation. Int J Mol Sci 2023; 24:11048. [PMID: 37446226 DOI: 10.3390/ijms241311048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
The remarkable advancements related to cerebral organoids have provided unprecedented opportunities to model human brain development and diseases. However, despite their potential significance in neurodegenerative diseases such as Parkinson's disease (PD), the role of exosomes from cerebral organoids (OExo) has been largely unknown. In this study, we compared the effects of OExo to those of mesenchymal stem cell (MSC)-derived exosomes (CExo) and found that OExo shared similar neuroprotective effects to CExo. Our findings showed that OExo mitigated H2O2-induced oxidative stress and apoptosis in rat midbrain astrocytes by reducing excess ROS production, antioxidant depletion, lipid peroxidation, mitochondrial dysfunction, and the expression of pro-apoptotic genes. Notably, OExo demonstrated superiority over CExo in promoting the differentiation of human-induced pluripotent stem cells (iPSCs) into dopaminergic (DA) neurons. This was attributed to the higher abundance of neurotrophic factors, including neurotrophin-4 (NT-4) and glial-cell-derived neurotrophic factor (GDNF), in OExo, which facilitated the iPSCs' differentiation into DA neurons in an LIM homeobox transcription factor 1 alpha (LMX1A)-dependent manner. Our study provides novel insight into the biological properties of cerebral organoids and highlights the potential of OExo in the treatment of neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
- Xingrui Ji
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Shaocong Zhou
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Nana Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Jingwen Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yue Wu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yuhan Duan
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Penghao Ni
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Jingzhong Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| | - Shuang Yu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
- School of Medical Imaging, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
14
|
Nie L, Yao D, Chen S, Wang J, Pan C, Wu D, Liu N, Tang Z. Directional induction of neural stem cells, a new therapy for neurodegenerative diseases and ischemic stroke. Cell Death Discov 2023; 9:215. [PMID: 37393356 DOI: 10.1038/s41420-023-01532-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023] Open
Abstract
Due to the limited capacity of the adult mammalian brain to self-repair and regenerate, neurological diseases, especially neurodegenerative disorders and stroke, characterized by irreversible cellular damage are often considered as refractory diseases. Neural stem cells (NSCs) play a unique role in the treatment of neurological diseases for their abilities to self-renew and form different neural lineage cells, such as neurons and glial cells. With the increasing understanding of neurodevelopment and advances in stem cell technology, NSCs can be obtained from different sources and directed to differentiate into a specific neural lineage cell phenotype purposefully, making it possible to replace specific cells lost in some neurological diseases, which provides new approaches to treat neurodegenerative diseases as well as stroke. In this review, we outline the advances in generating several neuronal lineage subtypes from different sources of NSCs. We further summarize the therapeutic effects and possible therapeutic mechanisms of these fated specific NSCs in neurological disease models, with special emphasis on Parkinson's disease and ischemic stroke. Finally, from the perspective of clinical translation, we compare the strengths and weaknesses of different sources of NSCs and different methods of directed differentiation, and propose future research directions for directed differentiation of NSCs in regenerative medicine.
Collapse
Affiliation(s)
- Luwei Nie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dabao Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Shiling Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jingyi Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chao Pan
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Dongcheng Wu
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, 430030, China
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, 430030, China
| | - Na Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
15
|
Wang X, Chen X, Liu G, Cai H, Le W. The Crucial Roles of Pitx3 in Midbrain Dopaminergic Neuron Development and Parkinson's Disease-Associated Neurodegeneration. Int J Mol Sci 2023; 24:ijms24108614. [PMID: 37239960 DOI: 10.3390/ijms24108614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
The degeneration of midbrain dopaminergic (mDA) neurons, particularly in the substantia nigra pars compacta (SNc), is one of the most prominent pathological hallmarks of Parkinson's disease (PD). To uncover the pathogenic mechanisms of mDA neuronal death during PD may provide therapeutic targets to prevent mDA neuronal loss and slow down the disease's progression. Paired-like homeodomain transcription factor 3 (Pitx3) is selectively expressed in the mDA neurons as early as embryonic day 11.5 and plays a critical role in mDA neuron terminal differentiation and subset specification. Moreover, Pitx3-deficient mice exhibit some canonical PD-related features, including the profound loss of SNc mDA neurons, a dramatic decrease in striatal dopamine (DA) levels, and motor abnormalities. However, the precise role of Pitx3 in progressive PD and how this gene contributes to mDA neuronal specification during early stages remains unclear. In this review, we updated the latest findings on Pitx3 by summarizing the crosstalk between Pitx3 and its associated transcription factors in mDA neuron development. We further explored the potential benefits of Pitx3 as a therapeutic target for PD in the future. To better understand the transcriptional network of Pitx3 in mDA neuron development may provide insights into Pitx3-related clinical drug-targeting research and therapeutic approaches.
Collapse
Affiliation(s)
- Xin Wang
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 611731, China
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 611731, China
| | - Guangdong Liu
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 611731, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 611731, China
| |
Collapse
|
16
|
Gamit N, Dharmarajan A, Sethi G, Warrier S. Want of Wnt in Parkinson's disease: Could sFRP disrupt interplay between Nurr1 and Wnt signaling? Biochem Pharmacol 2023; 212:115566. [PMID: 37088155 DOI: 10.1016/j.bcp.2023.115566] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Nuclear receptor related 1 (Nurr1) is a transcription factor known to regulate the development and maintenance of midbrain dopaminergic (mDA) neurons. Reports have confirmed that defect or obliteration of Nurr1 results in neurodegeneration and motor function impairment leading to Parkinson's disease (PD). Studies have also indicated that Nurr1 regulates the expression of alpha-synuclein (α-SYN) and mutations in Nurr1 cause α-SYN overexpression, thereby increasing the risk of PD. Nurr1 is modulated via various pathways including Wnt signaling pathway which is known to play an important role in neurogenesis and deregulation of it contributes to PD pathogenesis. Both Wnt/β-catenin dependent and independent pathways are implicated in the activation of Nurr1 and subsequent downregulation of α-SYN. This review highlights the interaction between Nurr1 and Wnt signaling pathways in mDA neuronal development. We further hypothesize how modulation of Wnt signaling pathway by its antagonist, secreted frizzled related proteins (sFRPs) could be a potential route to treat PD.
Collapse
Affiliation(s)
- Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600 116, India; School of Pharmacy and Biomedical Sciences, Curtin Medical School, Curtin University, Perth, Western Australia 6102, Australia; Curtin Health and Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia; School of Human Sciences, Faculty of Life and Physical Sciences, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore 117 600, Singapore
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India; Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India.
| |
Collapse
|
17
|
Abstract
The midbrain dopamine (mDA) system is composed of molecularly and functionally distinct neuron subtypes that mediate specific behaviours and are linked to various brain diseases. Considerable progress has been made in identifying mDA neuron subtypes, and recent work has begun to unveil how these neuronal subtypes develop and organize into functional brain structures. This progress is important for further understanding the disparate physiological functions of mDA neurons and their selective vulnerability in disease, and will ultimately accelerate therapy development. This Review discusses recent advances in our understanding of molecularly defined mDA neuron subtypes and their circuits, ranging from early developmental events, such as neuron migration and axon guidance, to their wiring and function, and future implications for therapeutic strategies.
Collapse
|
18
|
Embryoid Body Cells from Human Embryonic Stem Cells Overexpressing Dopaminergic Transcription Factors Survive and Initiate Neurogenesis via Neural Rosettes in the Substantia Nigra. Brain Sci 2023; 13:brainsci13020329. [PMID: 36831872 PMCID: PMC9954545 DOI: 10.3390/brainsci13020329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Transplantation of immature dopaminergic neurons or neural precursors derived from embryonic stem cells (ESCs) into the substantia nigra pars compacta (SNpc) is a potential therapeutic approach for functional restitution of the nigrostriatal pathway in Parkinson's disease (PD). However, further studies are needed to understand the effects of the local microenvironment on the transplanted cells to improve survival and specific differentiation in situ. We have previously reported that the adult SNpc sustains a neurogenic microenvironment. Non-neuralized embryoid body cells (EBCs) from mouse ESCs (mESCs) overexpressing the dopaminergic transcription factor Lmx1a gave rise to many tyrosine hydroxylase (Th+) cells in the intact and damaged adult SNpc, although only for a short-term period. Here, we extended our study by transplanting EBCs from genetically engineered naive human ESC (hESC), overexpressing the dopaminergic transcription factors LMX1A, FOXA2, and OTX2 (hESC-LFO), in the SNpc. Unexpectedly, no graft survival was observed in wild-type hESC EBCs transplants, whereas hESC-LFO EBCs showed viability in the SNpc. Interestingly, neural rosettes, a developmental hallmark of neuroepithelial tissue, emerged at 7- and 15-days post-transplantation (dpt) from the hESC-LFO EBCs. Neural rosettes expressed specification dopaminergic markers (Lmx1a, Otx2), which gave rise to several Th+ cells at 30 dpt. Our results suggest that the SNpc enables the robust initiation of neural differentiation of transplanted human EBCs prompted to differentiate toward the midbrain dopaminergic phenotype.
Collapse
|
19
|
Yeap YJ, Teddy TJW, Lee MJ, Goh M, Lim KL. From 2D to 3D: Development of Monolayer Dopaminergic Neuronal and Midbrain Organoid Cultures for Parkinson's Disease Modeling and Regenerative Therapy. Int J Mol Sci 2023; 24:ijms24032523. [PMID: 36768843 PMCID: PMC9917335 DOI: 10.3390/ijms24032523] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Parkinson's Disease (PD) is a prevalent neurodegenerative disorder that is characterized pathologically by the loss of A9-specific dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain. Despite intensive research, the etiology of PD is currently unresolved, and the disease remains incurable. This, in part, is due to the lack of an experimental disease model that could faithfully recapitulate the features of human PD. However, the recent advent of induced pluripotent stem cell (iPSC) technology has allowed PD models to be created from patient-derived cells. Indeed, DA neurons from PD patients are now routinely established in many laboratories as monolayers as well as 3D organoid cultures that serve as useful toolboxes for understanding the mechanism underlying PD and also for drug discovery. At the same time, the iPSC technology also provides unprecedented opportunity for autologous cell-based therapy for the PD patient to be performed using the patient's own cells as starting materials. In this review, we provide an update on the molecular processes underpinning the development and differentiation of human pluripotent stem cells (PSCs) into midbrain DA neurons in both 2D and 3D cultures, as well as the latest advancements in using these cells for drug discovery and regenerative medicine. For the novice entering the field, the cornucopia of differentiation protocols reported for the generation of midbrain DA neurons may seem daunting. Here, we have distilled the essence of the different approaches and summarized the main factors driving DA neuronal differentiation, with the view to provide a useful guide to newcomers who are interested in developing iPSC-based models of PD.
Collapse
Affiliation(s)
- Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Tng J. W. Teddy
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Interdisciplinary Graduate Programme (IGP-Neuroscience), Nanyang Technological University, Singapore 639798, Singapore
| | - Mok Jung Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Micaela Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- National Neuroscience Institute, Singapore 308433, Singapore
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK
- Department of Anatomy, Shanxi Medical University, Taiyuan 030001, China
- Correspondence:
| |
Collapse
|
20
|
Cha Y, Park TY, Leblanc P, Kim KS. Current Status and Future Perspectives on Stem Cell-Based Therapies for Parkinson's Disease. J Mov Disord 2023; 16:22-41. [PMID: 36628428 PMCID: PMC9978267 DOI: 10.14802/jmd.22141] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 01/12/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, affecting 1%-2% of the population over the age of 65. As the population ages, it is anticipated that the burden on society will significantly escalate. Although symptom reduction by currently available pharmacological and/or surgical treatments improves the quality of life of many PD patients, there are no treatments that can slow down, halt, or reverse disease progression. Because the loss of a specific cell type, midbrain dopamine neurons in the substantia nigra, is the main cause of motor dysfunction in PD, it is considered a promising target for cell replacement therapy. Indeed, numerous preclinical and clinical studies using fetal cell transplantation have provided proof of concept that cell replacement therapy may be a viable therapeutic approach for PD. However, the use of human fetal cells remains fraught with controversy due to fundamental ethical, practical, and clinical limitations. Groundbreaking work on human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, coupled with extensive basic research in the stem cell field offers promising potential for hPSC-based cell replacement to become a realistic treatment regimen for PD once several major issues can be successfully addressed. In this review, we will discuss the prospects and challenges of hPSC-based cell therapy for PD.
Collapse
Affiliation(s)
- Young Cha
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Tae-Yoon Park
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Pierre Leblanc
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Kwang-Soo Kim
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| |
Collapse
|
21
|
Abdelrahman S, Alsanie WF, Khan ZN, Albalawi HI, Felimban RI, Moretti M, Steiner N, Chaudhary AG, Hauser CAE. A Parkinson's disease model composed of 3D bioprinted dopaminergic neurons within a biomimetic peptide scaffold. Biofabrication 2022; 14. [PMID: 35793642 DOI: 10.1088/1758-5090/ac7eec] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/06/2022] [Indexed: 11/12/2022]
Abstract
Parkinson's disease (PD) is a progressive neurological disorder that affects movement. It is associated with lost dopaminergic (DA) neurons in thesubstantia nigra, a process that is not yet fully understood. To understand this deleterious disorder, there is an immense need to develop efficientin vitrothree-dimensional (3D) models that can recapitulate complex organs such as the brain. However, due to the complexity of neurons, selecting suitable biomaterials to accommodate them is challenging. Here, we report on the fabrication of functional DA neuronal 3D models using ultrashort self-assembling tetrapeptide scaffolds. Our peptide-based models demonstrate biocompatibility both for primary mouse embryonic DA neurons and for human DA neurons derived from human embryonic stem cells. DA neurons encapsulated in these scaffolds responded to 6-hydroxydopamine, a neurotoxin that selectively induces loss of DA neurons. Using multi-electrode arrays, we recorded spontaneous activity in DA neurons encapsulated within these 3D peptide scaffolds for more than 1 month without decrease of signal intensity. Additionally, vascularization of our 3D models in a co-culture with endothelial cells greatly promoted neurite outgrowth, leading to denser network formation. This increase of neuronal networks through vascularization was observed for both primary mouse DA and cortical neurons. Furthermore, we present a 3D bioprinted model of DA neurons inspired by the mouse brain and created with an extrusion-based 3D robotic bioprinting system that was developed during previous studies and is optimized with time-dependent pulsing by microfluidic pumps. We employed a hybrid fabrication strategy that relies on an external mold of the mouse brain construct that complements the shape and size of the desired bioprinted model to offer better support during printing. We hope that our 3D model provides a platform for studies of the pathogenesis of PD and other neurodegenerative disorders that may lead to better understanding and more efficient treatment strategies.
Collapse
Affiliation(s)
- Sherin Abdelrahman
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia.,Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratories Sciences, Faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia.,Center of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, Taif, Saudi Arabia
| | - Zainab N Khan
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia.,Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Hamed I Albalawi
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia.,Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Raed I Felimban
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.,Center of Innovation in Personalized Medicine (CIPM), 3D Bioprinting Unit, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Manola Moretti
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia.,Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Nadia Steiner
- Biological and Environmental Science and Engineering (BESE), Laboratory of Cellular Imaging and Energetics (LCIE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Adeel G Chaudhary
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia.,Center of Innovation in Personalized Medicine (CIPM), 3D Bioprinting Unit, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia.,Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
22
|
McComish SF, MacMahon Copas AN, Caldwell MA. Human Brain-Based Models Provide a Powerful Tool for the Advancement of Parkinson’s Disease Research and Therapeutic Development. Front Neurosci 2022; 16:851058. [PMID: 35651633 PMCID: PMC9149087 DOI: 10.3389/fnins.2022.851058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease and affects approximately 2–3% of the population over the age of 65. PD is characterised by the loss of dopaminergic neurons from the substantia nigra, leading to debilitating motor symptoms including bradykinesia, tremor, rigidity, and postural instability. PD also results in a host of non-motor symptoms such as cognitive decline, sleep disturbances and depression. Although existing therapies can successfully manage some motor symptoms for several years, there is still no means to halt progression of this severely debilitating disorder. Animal models used to replicate aspects of PD have contributed greatly to our current understanding but do not fully replicate pathological mechanisms as they occur in patients. Because of this, there is now great interest in the use of human brain-based models to help further our understanding of disease processes. Human brain-based models include those derived from embryonic stem cells, patient-derived induced neurons, induced pluripotent stem cells and brain organoids, as well as post-mortem tissue. These models facilitate in vitro analysis of disease mechanisms and it is hoped they will help bridge the existing gap between bench and bedside. This review will discuss the various human brain-based models utilised in PD research today and highlight some of the key breakthroughs they have facilitated. Furthermore, the potential caveats associated with the use of human brain-based models will be detailed.
Collapse
Affiliation(s)
- Sarah F. McComish
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Adina N. MacMahon Copas
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Maeve A. Caldwell
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Maeve A. Caldwell,
| |
Collapse
|
23
|
Xia N, Cabin DE, Fang F, Reijo Pera RA. Parkinson's Disease: Overview of Transcription Factor Regulation, Genetics, and Cellular and Animal Models. Front Neurosci 2022; 16:894620. [PMID: 35600613 PMCID: PMC9115107 DOI: 10.3389/fnins.2022.894620] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/12/2022] [Indexed: 01/21/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, affecting nearly 7-10 million people worldwide. Over the last decade, there has been considerable progress in our understanding of the genetic basis of PD, in the development of stem cell-based and animal models of PD, and in management of some clinical features. However, there remains little ability to change the trajectory of PD and limited knowledge of the underlying etiology of PD. The role of genetics versus environment and the underlying physiology that determines the trajectory of the disease are still debated. Moreover, even though protein aggregates such as Lewy bodies and Lewy neurites may provide diagnostic value, their physiological role remains to be fully elucidated. Finally, limitations to the model systems for probing the genetics, etiology and biology of Parkinson's disease have historically been a challenge. Here, we review highlights of the genetics of PD, advances in understanding molecular pathways and physiology, especially transcriptional factor (TF) regulators, and the development of model systems to probe etiology and potential therapeutic applications.
Collapse
Affiliation(s)
- Ninuo Xia
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Deborah E. Cabin
- McLaughlin Research Institute for Biomedical Sciences, Inc., Great Falls, MT, United States
| | - Fang Fang
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Renee A. Reijo Pera
- McLaughlin Research Institute for Biomedical Sciences, Inc., Great Falls, MT, United States
| |
Collapse
|
24
|
Li X, Hu X, Jiang Y, Wang D, Wang T, Li B. Autonomic behavioral impairment induced by simazine exposure during early life of male mouse is mediated by Lmx1a/Wnt1 pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:776-788. [PMID: 34936186 DOI: 10.1002/tox.23442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/06/2021] [Accepted: 12/11/2021] [Indexed: 06/14/2023]
Abstract
Simazine is a widely used herbicide and known as an environmental estrogen. Multiple studies have proved simazine can induced the degeneration of dopaminergic neuron resulting in a degenerative disease-like syndrome. Herein, we explored the neurotoxicity of simazine on the dopaminergic nervous system of embryos and weaned offspring during the maternal gestation period or the maternal gestation and lactation periods. We found that simazine disturbed the crucial components expression involved in Lmx1a/Wnt1 pathway of dopaminergic neuron in embryonic and weaned offspring. Furthermore, morphological and behavioral tests performed on weaned male offspring treated by simazine suggested that the grip strength, autonomic exploring, and the space sense ability were weakened, as well as the pathological damage of dopaminergic neuron was clearly observed. But, the same neurotoxicity of simazine is less significantly observed in female offspring. Our findings will provide reliable reference for the determination of environmental limits and new insight into the pathogenesis of nonfamilial neurodegenerative diseases related to environmental risk factors.
Collapse
Affiliation(s)
- Xueting Li
- Department of Hygienic Toxicology, College of Public Health, Harbin Medical University, Harbin, China
- Department of Epidemiology, College of Public Health, Harbin Medical University, Harbin, China
| | - Xiaomeng Hu
- Department of Hygienic Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| | - Yujia Jiang
- Department of Hygienic Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| | - Dandan Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, China
| | - Ting Wang
- Department of Hygienic Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| | - Baixiang Li
- Department of Hygienic Toxicology, College of Public Health, Harbin Medical University, Harbin, China
| |
Collapse
|
25
|
Spotting-based differentiation of functional dopaminergic progenitors from human pluripotent stem cells. Nat Protoc 2022; 17:890-909. [PMID: 35140411 DOI: 10.1038/s41596-021-00673-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
To fully realize the potential of human pluripotent stem cells (hPSCs) for both therapeutic and research purposes, it is critical to follow an efficient and reliable in vitro differentiation method that is based on optimal physical, chemical and developmental cues. This highly reproducible protocol describes how to grow hPSCs such as human induced pluripotent and embryonic stem cells in a physically confined area ('spot') and efficiently differentiate them into a highly enriched population of healthy and functional midbrain dopamine progenitors (mDAPs) and midbrain dopamine neurons (mDANs). The protocol takes 28 d, during which cells first grow and differentiate in spots for 14 d and then are replated and further differentiated for a further 14 d as a monolayer culture. We describe how to produce mDAPs, control the quality of cells and cryopreserve mDAPs without loss of viability. Previously we showed that mDANs generated by this 'spotting'-based method exhibit gene expression and (electro)physiological properties typical of A9 mDANs lost in Parkinson's disease brains and can rescue motor defects when transplanted into the striatum of 6-hydroxydopamine-lesioned rats. This protocol is scalable for production of mDAPs under good manufacturing practice conditions and was also previously successfully used to generate cells for the first autologous cell replacement therapy of a patient with Parkinson's disease without the need for immune suppression. We anticipate this protocol could also be readily adapted to use spotting-based culture to further optimize the differentiation of hPSC to alternative differentiated cell types.
Collapse
|
26
|
Lohoff T, Ghazanfar S, Missarova A, Koulena N, Pierson N, Griffiths JA, Bardot ES, Eng CHL, Tyser RCV, Argelaguet R, Guibentif C, Srinivas S, Briscoe J, Simons BD, Hadjantonakis AK, Göttgens B, Reik W, Nichols J, Cai L, Marioni JC. Integration of spatial and single-cell transcriptomic data elucidates mouse organogenesis. Nat Biotechnol 2022; 40:74-85. [PMID: 34489600 PMCID: PMC8763645 DOI: 10.1038/s41587-021-01006-2] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Molecular profiling of single cells has advanced our knowledge of the molecular basis of development. However, current approaches mostly rely on dissociating cells from tissues, thereby losing the crucial spatial context of regulatory processes. Here, we apply an image-based single-cell transcriptomics method, sequential fluorescence in situ hybridization (seqFISH), to detect mRNAs for 387 target genes in tissue sections of mouse embryos at the 8-12 somite stage. By integrating spatial context and multiplexed transcriptional measurements with two single-cell transcriptome atlases, we characterize cell types across the embryo and demonstrate that spatially resolved expression of genes not profiled by seqFISH can be imputed. We use this high-resolution spatial map to characterize fundamental steps in the patterning of the midbrain-hindbrain boundary (MHB) and the developing gut tube. We uncover axes of cell differentiation that are not apparent from single-cell RNA-sequencing (scRNA-seq) data, such as early dorsal-ventral separation of esophageal and tracheal progenitor populations in the gut tube. Our method provides an approach for studying cell fate decisions in complex tissues and development.
Collapse
Affiliation(s)
- T Lohoff
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Epigenetics Programme, Babraham Institute, Cambridge, UK
| | - S Ghazanfar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - A Missarova
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK
| | - N Koulena
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - N Pierson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - J A Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Genomics Plc, Cambridge, UK
| | - E S Bardot
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - C-H L Eng
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - R C V Tyser
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - R Argelaguet
- Epigenetics Programme, Babraham Institute, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK
| | - C Guibentif
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Sahlgrenska Center for Cancer Research, Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - S Srinivas
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - J Briscoe
- The Francis Crick Institute, London, UK
| | - B D Simons
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- The Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
| | - A-K Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - B Göttgens
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - W Reik
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Epigenetics Programme, Babraham Institute, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
| | - J Nichols
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - L Cai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - J C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
| |
Collapse
|
27
|
Oosterveen T, Garção P, Moles-Garcia E, Soleilhavoup C, Travaglio M, Sheraz S, Peltrini R, Patrick K, Labas V, Combes-Soia L, Marklund U, Hohenstein P, Panman L. Pluripotent stem cell derived dopaminergic subpopulations model the selective neuron degeneration in Parkinson's disease. Stem Cell Reports 2021; 16:2718-2735. [PMID: 34678205 PMCID: PMC8581055 DOI: 10.1016/j.stemcr.2021.09.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 01/06/2023] Open
Abstract
In Parkinson’s disease (PD), substantia nigra (SN) dopaminergic (DA) neurons degenerate, while related ventral tegmental area (VTA) DA neurons remain relatively unaffected. Here, we present a methodology that directs the differentiation of mouse and human pluripotent stem cells toward either SN- or VTA-like DA lineage and models their distinct vulnerabilities. We show that the level of WNT activity is critical for the induction of the SN- and VTA-lineage transcription factors Sox6 and Otx2, respectively. Both WNT signaling modulation and forced expression of these transcription factors can drive DA neurons toward the SN- or VTA-like fate. Importantly, the SN-like lineage enriched DA cultures recapitulate the selective sensitivity to mitochondrial toxins as observed in PD, while VTA-like neuron-enriched cultures are more resistant. Furthermore, a proteomics approach led to the identification of compounds that alter SN neuronal survival, demonstrating the utility of our strategy for disease modeling and drug discovery. Derivation of distinct dopaminergic subpopulations from pluripotent stem cells Wnt signaling inhibitors promote SN dopaminergic neuron specification Modeling selective vulnerability of SN dopaminergic neurons in vitro Proteomics reveals pathways that promote SN dopaminergic neuron survival
Collapse
Affiliation(s)
- Tony Oosterveen
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Pedro Garção
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Emma Moles-Garcia
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Clement Soleilhavoup
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Marco Travaglio
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Shahida Sheraz
- Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Rosa Peltrini
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Kieran Patrick
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Valerie Labas
- PRC, INRA, CNRS, University of Tours, IFCE, Nouzilly, France
| | | | - Ulrika Marklund
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | - Lia Panman
- MRC Toxicology Unit, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| |
Collapse
|
28
|
Khajebishak Y, Alivand M, Faghfouri AH, Moludi J, Payahoo L. The effects of vitamins and dietary pattern on epigenetic modification of non-communicable diseases. INT J VITAM NUTR RES 2021. [PMID: 34643416 DOI: 10.1024/0300-9831/a000735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background: Non-communicable diseases (NCDs) have received more attention because of high prevalence and mortality rate. Besides genetic and environmental factors, the epigenetic abnormality is also involved in the pathogenesis of NCDs. Methylation of DNA, chromatin remodeling, modification of histone, and long non-coding RNAs are the main components of epigenetic phenomena. Methodology: In this review paper, the mechanistic role of vitamins and dietary patterns on epigenetic modification was discussed. All papers indexed in scientific databases, including PubMed, Scopus, Embase, Google Scholar, and Elsevier were searched during 2000 - 2021 using, vitamins, diet, epigenetic repression, histones, methylation, acetylation, and NCDs as keywords. Results: The components of healthy dietary patterns like Mediterranean and dietary approaches to stop hypertension diets have a beneficial effect on epigenetic hemostasis. Both quality and quantity of dietary components influence epigenetic phenomena. A diet with calorie deficiency in protein content and methyl-donor agents in a long time, with a high level of fat, disrupts epigenetic hemostasis and finally, causes genome instability. Also, soluble and insoluble vitamins have an obvious role in epigenetic modifications. Most vitamins interact directly with methylation, acetylation, and phosphorylation pathways of histone and DNA. However, numerous indirect functions related to the cell cycle stability and genome integrity have been recognized. Conclusion: Considering the crucial role of a healthy diet in epigenetic homeostasis, adherence to a healthy dietary pattern containing enough levels of vitamin and avoiding the western diet seems to be necessary. Having a healthy diet and consuming the recommended dietary level of vitamins can also contribute to epigenetic stability.
Collapse
Affiliation(s)
- Yaser Khajebishak
- Department of Nutrition and Food Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mohammadreza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Jalal Moludi
- School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Laleh Payahoo
- Department of Nutrition and Food Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| |
Collapse
|
29
|
Effect of NTN and Lmx1 α on the Notch Signaling Pathway during the Differentiation of Human Bone Marrow Mesenchymal Stem Cells into Dopaminergic Neuron-Like Cells. PARKINSONS DISEASE 2021; 2021:6676709. [PMID: 34373779 PMCID: PMC8349261 DOI: 10.1155/2021/6676709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/30/2021] [Accepted: 07/22/2021] [Indexed: 11/20/2022]
Abstract
Human bone marrow mesenchymal stem cells (h-BMSCs) have the potential to differentiate into dopaminergic neuron-like cells to treat Parkinson's disease. The Notch signaling pathway has been implicated in the regulation of cell fate decisions such as differentiation of BMSCs. This study investigated changes in the expression of Notch-related genes in the differentiation of BMSCs in vitro into dopaminergic (DA) neuron-like cells. BMSCs transfected with empty lentiviral vectors served as the control group and those transfected with NTN and Lmx1α recombinant lentiviral vectors served as the experimental group. After induction and culture of NTN and Lmx1α-transfected h-BMSCs for 21 days, the cells exhibited features of dopaminergic neuron-like cells, which were observed by transmission and scanning electron microscopy and verified by immunofluorescence of tyrosine hydroxylase (TH) and dopamine transporter (DAT). These induced cells could secrete dopamine and had basic action potentials. Expression of the neural stem cell (NSC) markers, including octamer-binding protein (Oct4), paired box gene 6 (Pax6), and sex determining region Y-box 1 (SOX1), increased on day 14 of induction and decreased on day 21 of induction during differentiation. The human Notch signaling pathway PCR array showed a differential expression of Notch-related genes during the differentiation of h-BMSCs into DA neuron-like cells in vitro relative to that in the control group. In conclusion, h-BMSCs overexpressing NTN and Lmx1α can successfully be induced to differentiate into dopaminergic neuron-like cells with a neuronal phenotype exhibiting fundamental biological functions in vitro, and NTN and Lmx1α may affect the expression of Notch-related genes during differentiation.
Collapse
|
30
|
Ng YH, Chanda S, Janas JA, Yang N, Kokubu Y, Südhof TC, Wernig M. Efficient generation of dopaminergic induced neuronal cells with midbrain characteristics. Stem Cell Reports 2021; 16:1763-1776. [PMID: 34171286 PMCID: PMC8282497 DOI: 10.1016/j.stemcr.2021.05.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 05/22/2021] [Accepted: 05/23/2021] [Indexed: 12/12/2022] Open
Abstract
The differentiation of pluripotent stem cells can be accomplished by sequential activation of signaling pathways or through transcription factor programming. Multistep differentiation imitates embryonic development to obtain authentic cell types, but it suffers from asynchronous differentiation with variable efficiency. Transcription factor programming induces synchronous and efficient differentiation with higher reproducibility but may not always yield authentic cell types. We systematically explored the generation of dopaminergic induced neuronal cells from mouse and human pluripotent stem cells. We found that the proneural factor Ascl1 in combination with mesencephalic factors Lmx1a and Nurr1 induce peripheral dopaminergic neurons. Co-delivery of additional midbrain transcription factors En1, FoxA2, and Pitx3 resulted in facile and robust generation of functional dopaminergic neurons of midbrain character. Our results suggest that more complex combinations of transcription factors may be needed for proper regional specification of induced neuronal cells generated by direct lineage induction. Ascl1 alone can induce tyrosine hydroxylase (TH)-positive ES-iN cells Ascl1 alone, or with Nurr1 and Lmx1a, induce peripheral TH-positive cells WNT1 and neurotrophic factors increase TH-positive iN cells in culture A 6-factor combination induces TH-positive dopamine iN cells of central identity
Collapse
Affiliation(s)
- Yi Han Ng
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Soham Chanda
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Justyna A Janas
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nan Yang
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yuko Kokubu
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
Strano A, Tuck E, Stubbs VE, Livesey FJ. Variable Outcomes in Neural Differentiation of Human PSCs Arise from Intrinsic Differences in Developmental Signaling Pathways. Cell Rep 2021; 31:107732. [PMID: 32521257 PMCID: PMC7296348 DOI: 10.1016/j.celrep.2020.107732] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 03/09/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
Directed differentiation of human pluripotent stem cells varies in specificity and efficiency. Stochastic, genetic, intracellular, and environmental factors affect maintenance of pluripotency and differentiation into early embryonic lineages. However, factors affecting variation in in vitro differentiation to defined cell types are not well understood. To address this, we focused on a well-established differentiation process to cerebral cortex neural progenitor cells and their neuronal progeny from human pluripotent stem cells. Analysis of 162 differentiation outcomes of 61 stem cell lines derived from 37 individuals showed that most variation occurs along gene expression axes reflecting dorsoventral and rostrocaudal spatial expression during in vivo brain development. Line-independent and line-dependent variations occur, with the latter driven largely by differences in endogenous Wnt signaling activity. Tuning Wnt signaling during a specific phase early in the differentiation process reduces variability, demonstrating that cell-line/genome-specific differentiation outcome biases can be corrected by controlling extracellular signaling. Analysis of 162 iPSC neural differentiation quantifies sources of variability Variation in outcomes occurs along developmental brain spatial and regional axes Variation depends on iPSC-line-specific differences in Wnt/β-catenin signaling Effects of signaling differences can be rescued by exogenous pathway activation
Collapse
Affiliation(s)
- Alessio Strano
- The Wellcome Trust/Cancer Research UK Gurdon Institute & Department of Biochemistry, University of Cambridge, Cambridge, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, 20 Guilford Street, London WC1N 1DZ, UK
| | - Eleanor Tuck
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, 20 Guilford Street, London WC1N 1DZ, UK
| | - Victoria E Stubbs
- The Wellcome Trust/Cancer Research UK Gurdon Institute & Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Frederick J Livesey
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, 20 Guilford Street, London WC1N 1DZ, UK.
| |
Collapse
|
32
|
Coles C, Lasek AW. Binge-Like Ethanol Drinking Increases Otx2, Wnt1, and Mdk Gene Expression in the Ventral Tegmental Area of Adult Mice. Neurosci Insights 2021; 16:26331055211009850. [PMID: 33954290 PMCID: PMC8058803 DOI: 10.1177/26331055211009850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/25/2021] [Indexed: 12/23/2022] Open
Abstract
Alcohol use disorder is associated with pathophysiological changes in the dopaminergic system. Orthodenticle homeobox 2 (OTX2) is a transcription factor important for the development of dopaminergic neurons residing in the ventral tegmental area (VTA), a critical region of the brain involved in drug reinforcement. Previous studies have demonstrated that ethanol exposure during embryonic development reduces Otx2 mRNA levels in the central nervous system. We hypothesized that levels of OTX2 would be altered by binge-like ethanol consumption in adult animals. To test this, Otx2 mRNA and protein levels in the mouse VTA were measured by quantitative real-time PCR and western blotting, respectively, after mice drank ethanol for 4 days in a procedure that elicits binge levels of ethanol consumption (drinking in the dark). Expression of known and putative OTX2 transcriptional target genes (Sema3c, Wnt1, and Mdk) were also measured in the VTA after ethanol drinking. Otx2 mRNA and protein levels were elevated in the VTA 24 hours after the fourth drinking session and there was a corresponding increase in the expression of Mdk transcript. Interestingly, Wnt1 transcript was elevated in the VTA immediately after the fourth drinking session but returned to control levels 24 hours later. We next investigated if viral-mediated reduction of Otx2 in the mouse VTA would alter ethanol or sucrose intake. Lentiviral vectors expressing a shRNA targeting Otx2 or a control shRNA were injected into the VTA and mice were tested in the drinking in the dark protocol for ethanol and sucrose drinking. Reducing levels of OTX2 in the VTA did not alter ethanol or sucrose consumption. One limitation is that the extent of OTX2 reduction may not have been sufficient. Although OTX2 in the VTA may not play a role in binge-like drinking in adult mice, OTX2 could contribute to ethanol-induced transcriptional changes in this region.
Collapse
Affiliation(s)
- Cassandre Coles
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA.,Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
33
|
Wulansari N, Darsono WHW, Woo HJ, Chang MY, Kim J, Bae EJ, Sun W, Lee JH, Cho IJ, Shin H, Lee SJ, Lee SH. Neurodevelopmental defects and neurodegenerative phenotypes in human brain organoids carrying Parkinson's disease-linked DNAJC6 mutations. SCIENCE ADVANCES 2021; 7:eabb1540. [PMID: 33597231 PMCID: PMC7888924 DOI: 10.1126/sciadv.abb1540] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 12/28/2020] [Indexed: 05/14/2023]
Abstract
Loss-of-function mutations of DNAJC6, encoding HSP40 auxilin, have recently been identified in patients with early-onset Parkinson's disease (PD). To study the roles of DNAJC6 in PD pathogenesis, we used human embryonic stem cells with CRISPR-Cas9-mediated gene editing. Here, we show that DNAJC6 mutations cause key PD pathologic features, i.e., midbrain-type dopamine (mDA) neuron degeneration, pathologic α-synuclein aggregation, increase of intrinsic neuronal firing frequency, and mitochondrial and lysosomal dysfunctions in human midbrain-like organoids (hMLOs). In addition, neurodevelopmental defects were also manifested in hMLOs carrying the mutations. Transcriptomic analyses followed by experimental validation revealed that defects in DNAJC6-mediated endocytosis impair the WNT-LMX1A signal during the mDA neuron development. Furthermore, reduced LMX1A expression during development caused the generation of vulnerable mDA neurons with the pathologic manifestations. These results suggest that the human model of DNAJC6-PD recapitulates disease phenotypes and reveals mechanisms underlying disease pathology, providing a platform for assessing therapeutic interventions.
Collapse
Affiliation(s)
- Noviana Wulansari
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Wahyu Handoko Wibowo Darsono
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hye-Ji Woo
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Mi-Yoon Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Jinil Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Eun-Jin Bae
- Department of Biomedical Sciences and Medicine, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ju-Hyun Lee
- Department of Anatomy, Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Daejeon, Republic of Korea
- School of Electrical and Electronics Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences and Medicine, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
34
|
Nouri P, Götz S, Rauser B, Irmler M, Peng C, Trümbach D, Kempny C, Lechermeier CG, Bryniok A, Dlugos A, Euchner E, Beckers J, Brodski C, Klümper C, Wurst W, Prakash N. Dose-Dependent and Subset-Specific Regulation of Midbrain Dopaminergic Neuron Differentiation by LEF1-Mediated WNT1/b-Catenin Signaling. Front Cell Dev Biol 2020; 8:587778. [PMID: 33195246 PMCID: PMC7649324 DOI: 10.3389/fcell.2020.587778] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/01/2020] [Indexed: 01/07/2023] Open
Abstract
The mesodiencephalic dopaminergic (mdDA) neurons, including the nigrostriatal subset that preferentially degenerates in Parkinson’s Disease (PD), strongly depend on an accurately balanced Wingless-type MMTV integration site family member 1 (WNT1)/beta-catenin signaling pathway during their development. Loss of this pathway abolishes the generation of these neurons, whereas excessive WNT1/b-catenin signaling prevents their correct differentiation. The identity of the cells responding to this pathway in the developing mammalian ventral midbrain (VM) as well as the precise progression of WNT/b-catenin action in these cells are still unknown. We show that strong WNT/b-catenin signaling inhibits the differentiation of WNT/b-catenin-responding mdDA progenitors into PITX3+ and TH+ mdDA neurons by repressing the Pitx3 gene in mice. This effect is mediated by RSPO2, a WNT/b-catenin agonist, and lymphoid enhancer binding factor 1 (LEF1), an essential nuclear effector of the WNT/b-catenin pathway, via conserved LEF1/T-cell factor binding sites in the Pitx3 promoter. LEF1 expression is restricted to a caudolateral mdDA progenitor subset that preferentially responds to WNT/b-catenin signaling and gives rise to a fraction of all mdDA neurons. Our data indicate that an attenuation of WNT/b-catenin signaling in mdDA progenitors is essential for their correct differentiation into specific mdDA neuron subsets. This is an important consideration for stem cell-based regenerative therapies and in vitro models of neuropsychiatric diseases.
Collapse
Affiliation(s)
- Parivash Nouri
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Sebastian Götz
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Benedict Rauser
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Changgeng Peng
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Advanced Institute of Translational Medicine, The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Kempny
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Carina G Lechermeier
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Agnes Bryniok
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Andrea Dlugos
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Ellen Euchner
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Experimental Genetics, Technical University of Munich, Munich, Germany.,German Center for Diabetes Research, Neuherberg, Germany
| | - Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Claudia Klümper
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Developmental Genetics, Helmholtz Zentrum München, Technical University of Munich/Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nilima Prakash
- Laboratory of Applied Genetics and Stem Cell Biology, Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, Hamm, Germany
| |
Collapse
|
35
|
Qin H, Zhao AD, Sun ML, Ma K, Fu XB. Direct conversion of human fibroblasts into dopaminergic neuron-like cells using small molecules and protein factors. Mil Med Res 2020; 7:52. [PMID: 33129359 PMCID: PMC7603706 DOI: 10.1186/s40779-020-00284-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/21/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Generation of neurons is essential in cell replacement therapy for neurodegenerative disorders like Parkinson's disease. Several studies have reported the generation of dopaminergic (DA) neurons from mouse and human fibroblasts by ectopic expression of transcription factors, in which genetic manipulation is associated with potential risks. METHODS The small molecules and protein factors were selected based on their function to directly induce human fetal lung IMR-90 fibroblasts into DA neuron-like cells. Microscopical, immunocytochemical, and RT-qPCR analyses were used to characterize the morphology, phenotype, and gene expression features of the induced cells. The whole-cell patch-clamp recordings were exploited to measure the electrophysiological properties. RESULTS Human IMR-90 fibroblasts were rapidly converted into DA neuron-like cells after the chemical induction using small molecules and protein factors, with a yield of approximately 95% positive TUJ1-positive cells. The induced DA neuron-like cells were immunopositive for pan-neuronal markers MAP2, NEUN, and Synapsin 1 and DA markers TH, DDC, DAT, and NURR1. The chemical induction process did not involve a neural progenitor/stem cell intermediate stage. The induced neurons could fire single action potentials, which reflected partially the electrophysiological properties of neurons. CONCLUSION We developed a chemical cocktail of small molecules and protein factors to convert human fibroblasts into DA neuron-like cells without passing through a neural progenitor/stem cell intermediate stage. The induced DA neuron-like cells from human fibroblasts might provide a cellular source for cell-based therapy of Parkinson's disease in the future.
Collapse
Affiliation(s)
- Hua Qin
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, China
| | - An-Dong Zhao
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, China.,Tianjin Medical University, Tianjin, 300070, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China
| | - Meng-Li Sun
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China
| | - Kui Ma
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China
| | - Xiao-Bing Fu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Haidian District, Beijing, 100853, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, 100048, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, Beijing, 100048, China.
| |
Collapse
|
36
|
Jang SE, Qiu L, Chan LL, Tan EK, Zeng L. Current Status of Stem Cell-Derived Therapies for Parkinson's Disease: From Cell Assessment and Imaging Modalities to Clinical Trials. Front Neurosci 2020; 14:558532. [PMID: 33177975 PMCID: PMC7596695 DOI: 10.3389/fnins.2020.558532] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/17/2020] [Indexed: 12/23/2022] Open
Abstract
Curative therapies or treatments reversing the progression of Parkinson’s disease (PD) have attracted considerable interest in the last few decades. PD is characterized by the gradual loss of dopaminergic (DA) neurons and decreased striatal dopamine levels. Current challenges include optimizing neuroprotective strategies, developing personalized drug therapy, and minimizing side effects from the long-term prescription of pharmacological drugs used to relieve short-term motor symptoms. Transplantation of DA cells into PD patients’ brains to replace degenerated DA has the potential to change the treatment paradigm. Herein, we provide updates on current progress in stem cell-derived DA neuron transplantation as a therapeutic alternative for PD. We briefly highlight cell sources for transplantation and focus on cell assessment methods such as identification of genetic markers, single-cell sequencing, and imaging modalities used to access cell survival and function. More importantly, we summarize clinical reports of patients who have undergone cell-derived transplantation in PD to better perceive lessons that can be drawn from past and present clinical outcomes. Modifying factors include (1) source of the stem cells, (2) quality of the stem cells, (3) age of the patient, (4) stage of disease progression at the time of cell therapy, (5) surgical technique/practices, and (6) the use of immunosuppression. We await the outcomes of joint efforts in clinical trials around the world such as NYSTEM and CiRA to further guide us in the selection of the most suitable parameters for cell-based neurotransplantation in PD.
Collapse
Affiliation(s)
- Se Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Ling Ling Chan
- Department of Diagnostic Radiology, Singapore General Hospital, Singapore, Singapore.,Neuroscience & Behavioral Disorders Program, Duke University and National University of Singapore (DUKE-NUS), Graduate Medical School, Singapore, Singapore
| | - Eng-King Tan
- Neuroscience & Behavioral Disorders Program, Duke University and National University of Singapore (DUKE-NUS), Graduate Medical School, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore General Hospital Campus, Singapore, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore.,Neuroscience & Behavioral Disorders Program, Duke University and National University of Singapore (DUKE-NUS), Graduate Medical School, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Novena Campus, Singapore, Singapore
| |
Collapse
|
37
|
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: 1.3] [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
|
38
|
Song B, Cha Y, Ko S, Jeon J, Lee N, Seo H, Park KJ, Lee IH, Lopes C, Feitosa M, Luna MJ, Jung JH, Kim J, Hwang D, Cohen BM, Teicher MH, Leblanc P, Carter BS, Kordower JH, Bolshakov VY, Kong SW, Schweitzer JS, Kim KS. Human autologous iPSC-derived dopaminergic progenitors restore motor function in Parkinson's disease models. J Clin Invest 2020; 130:904-920. [PMID: 31714896 DOI: 10.1172/jci130767] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder associated with loss of striatal dopamine, secondary to degeneration of midbrain dopamine (mDA) neurons in the substantia nigra, rendering cell transplantation a promising therapeutic strategy. To establish human induced pluripotent stem cell-based (hiPSC-based) autologous cell therapy, we report a platform of core techniques for the production of mDA progenitors as a safe and effective therapeutic product. First, by combining metabolism-regulating microRNAs with reprogramming factors, we developed a method to more efficiently generate clinical-grade iPSCs, as evidenced by genomic integrity and unbiased pluripotent potential. Second, we established a "spotting"-based in vitro differentiation methodology to generate functional and healthy mDA cells in a scalable manner. Third, we developed a chemical method that safely eliminates undifferentiated cells from the final product. Dopaminergic cells thus express high levels of characteristic mDA markers, produce and secrete dopamine, and exhibit electrophysiological features typical of mDA cells. Transplantation of these cells into rodent models of PD robustly restores motor function and reinnervates host brain, while showing no evidence of tumor formation or redistribution of the implanted cells. We propose that this platform is suitable for the successful implementation of human personalized autologous cell therapy for PD.
Collapse
Affiliation(s)
- Bin Song
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Young Cha
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Sanghyeok Ko
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Jeha Jeon
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Nayeon Lee
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Hyemyung Seo
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Department of Molecular and Life Sciences, Hanyang University, Ansan, Korea
| | | | - In-Hee Lee
- Department of Pediatrics.,Computational Health Informatics Program, Boston Children's Hospital, and
| | - Claudia Lopes
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Melissa Feitosa
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - María José Luna
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Jin Hyuk Jung
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Jisun Kim
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA.,Department of Molecular and Life Sciences, Hanyang University, Ansan, Korea
| | - Dabin Hwang
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | | | | | - Pierre Leblanc
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | | | - Sek Won Kong
- Department of Pediatrics.,Computational Health Informatics Program, Boston Children's Hospital, and
| | - Jeffrey S Schweitzer
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kwang-Soo Kim
- Department of Psychiatry and.,Molecular Neurobiology Laboratory, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, USA
| |
Collapse
|
39
|
Li X, Li J, Li P, Jiang Y, Wu Y, Li B. Injury to dopaminergic neurons development via the Lmx1a/Wnt1 autoregulatory loop induced by simazine. Toxicol Lett 2020; 333:279-289. [PMID: 32822773 DOI: 10.1016/j.toxlet.2020.07.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/17/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022]
Abstract
Simazine is a kind of persistent organic pollutant that is detected in both ground and water and has several routes of exposure. Here, we explored the mechanisms underlying simazine-related effects on dopaminergic neurons via development-related factors using mouse embryos and embryonic mesencephalic hybrid cell line (MN9D cells). We treated pregnant mice with 50 μg/kg bw, 200 μg/kg bw simazine from the 0.5 day to the 10.5 day of embryonic phase and MN9D cells with 600 μM simazine for 24 h to research the mechanism of dopaminergic neurons acute respond to simazine through preliminary experiments. Protein expressions of LIM homeobox transcription factor 1-alpha (Lmx1a) and LIM homeobox transcription factor 1-beta (Lmx1b) displayed a dose- and time-dependent increase after the exposure to simazine. In the 200 μg/kg bw of embryos and the 24h-600 μM of MN9D cells, protein levels of dopaminergic developmental factors were significantly upregulated, and dopaminergic function was significantly damaged for the abnormal expression of Dyt5b. We demonstrated simazine induced the injury to dopaminergic neurons via the Lmx1a/wingless-related integration site 1 (Wnt1) and Lmx1b pathways. In the transfection experiments, we knocked down Lmx1a and Lmx1b of cells to verify the potential target of simazine-induced injury to dopaminergic neurons, respectively. We detected the protein and mRNA levels of development-related genes of dopaminergic neurons and intracellular dopamine levels in different treatment groups. Based on our experiments' results, we demonstrated an acute response to 24 h-600 μM simazine treatment, the simazine-induced injury to dopaminergic neuronal which leads to abnormal dopamine levels and dopaminergic impairment is via the activation of the Lmx1a/Wnt1 autoregulatory loop. Lmx1a is a promising target in the search for the mechanisms underlying simazine-induced dopaminergic injury.
Collapse
Affiliation(s)
- Xueting Li
- Department of Toxicology, College of Public Health, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang Province, 150081, PR China
| | - Jianan Li
- Department of Toxicology, College of Public Health, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang Province, 150081, PR China
| | - Peng Li
- Department of Toxicology, College of Public Health, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang Province, 150081, PR China
| | - Yujia Jiang
- Department of Toxicology, College of Public Health, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang Province, 150081, PR China
| | - Yanping Wu
- Department of Toxicology, College of Public Health, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang Province, 150081, PR China
| | - Baixiang Li
- Department of Toxicology, College of Public Health, Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang Province, 150081, PR China.
| |
Collapse
|
40
|
Integrative Analysis of Gene Expression and Regulatory Network Interaction Data Reveals the Protein Kinase C Family of Serine/Threonine Receptors as a Significant Druggable Target for Parkinson's Disease. J Mol Neurosci 2020; 71:466-480. [PMID: 32728898 DOI: 10.1007/s12031-020-01669-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/14/2020] [Indexed: 10/23/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease affecting the ventral midbrain dopaminergic neurons, resulting in motor defects mainly tremor, rigidity, and bradykinesia along with a wide array of non-motor symptoms. The current study is focused on determining the potential druggable targets of PD by consolidating gene expression profiling and network methodology. Initially, the differentially expressed genes were established from which the central network was constructed by assimilating the interacting partners. Investigating the topological parameters of the network, the genes SYT1, CXCR4, CDC42, KIT, RET, DRD2, NTN1, PRKACB, KDR, NR4A2, SLC18A2, CCK, TH, KCNJ6, and TAC1 were identified as the hub genes and can be explored as potential candidate genes for PD therapeutics. Gene ontology and cluster analysis of the hub genes has provided further insights about the pathophysiology of the disease. Among the hub genes, PRKACB is observed in relatively all the enriched pathways which are modulated by G protein-coupled receptors through protein kinases. Further, we noticed SYT1 as a novel biomarker for PD. Moreover, the regulatory network was constructed with the hub genes as seed nodes with associated transcription factors (TFs) and microRNA (miRNAs). In this analysis, we identified MYC as the major TF and the miRNAs miR-21, miR-155, miR-7, and miR26A1 have a significant role in modulating the hub genes. Briefly, these significant hub genes and their enriched pathways, TFs, and miRNAs have aided in the better understanding of molecular mechanisms underlying PD and its potential core target genes.
Collapse
|
41
|
Development and Differentiation of Midbrain Dopaminergic Neuron: From Bench to Bedside. Cells 2020; 9:cells9061489. [PMID: 32570916 PMCID: PMC7349799 DOI: 10.3390/cells9061489] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/29/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s Disease (PD) is a neurodegenerative disorder affecting the motor system. It is primarily due to substantial loss of midbrain dopamine (mDA) neurons in the substantia nigra pars compacta and to decreased innervation to the striatum. Although existing drug therapy available can relieve the symptoms in early-stage PD patients, it cannot reverse the pathogenic progression of PD. Thus, regenerating functional mDA neurons in PD patients may be a cure to the disease. The proof-of-principle clinical trials showed that human fetal graft-derived mDA neurons could restore the release of dopamine neurotransmitters, could reinnervate the striatum, and could alleviate clinical symptoms in PD patients. The invention of human-induced pluripotent stem cells (hiPSCs), autologous source of neural progenitors with less ethical consideration, and risk of graft rejection can now be generated in vitro. This advancement also prompts extensive research to decipher important developmental signaling in differentiation, which is key to successful in vitro production of functional mDA neurons and the enabler of mass manufacturing of the cells required for clinical applications. In this review, we summarize the biology and signaling involved in the development of mDA neurons and the current progress and methodology in driving efficient mDA neuron differentiation from pluripotent stem cells.
Collapse
|
42
|
Stathakos P, Jiménez-Moreno N, Crompton LA, Nistor PA, Badger JL, Barbuti PA, Kerrigan TL, Randall AD, Caldwell MA, Lane JD. A monolayer hiPSC culture system for autophagy/mitophagy studies in human dopaminergic neurons. Autophagy 2020; 17:855-871. [PMID: 32286126 PMCID: PMC8078667 DOI: 10.1080/15548627.2020.1739441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Macroautophagy/autophagy cytoplasmic quality control pathways are required during neural development and are critical for the maintenance of functional neuronal populations in the adult brain. Robust evidence now exists that declining neuronal autophagy pathways contribute to human neurodegenerative diseases, including Parkinson disease (PD). Reliable and relevant human neuronal model systems are therefore needed to understand the biology of disease-vulnerable neural populations, to decipher the underlying causes of neurodegenerative disease, and to develop assays to test therapeutic interventions in vitro. Human induced pluripotent stem cell (hiPSC) neural model systems can meet this demand: they provide a renewable source of material for differentiation into regional neuronal sub-types for functional assays; they can be expanded to provide a platform for screening, and they can potentially be optimized for transplantation/neurorestorative therapy. So far, however, hiPSC differentiation protocols for the generation of ventral midbrain dopaminergic neurons (mDANs) – the predominant neuronal sub-type afflicted in PD – have been somewhat restricted by poor efficiency and/or suitability for functional and/or imaging-based in vitro assays. Here, we describe a reliable, monolayer differentiation protocol for the rapid and reproducible production of high numbers of mDANs from hiPSC in a format that is amenable for autophagy/mitophagy research. We characterize these cells with respect to neuronal differentiation and macroautophagy capability and describe qualitative and quantitative assays for the study of autophagy and mitophagy in these important cells. Abbreviations: AA: ascorbic acid; ATG: autophagy-related; BDNF: brain derived neurotrophic factor; CCCP: carbonyl cyanide m-chlorophenylhydrazone; dbcAMP: dibutyryl cAMP; DAN: dopaminergic neuron; DAPI: 4ʹ,6-diamidino-2-phenylindole; DAPT: N-[N-(3,5-difluorophenacetyl)-L-alanyl]-sphenylglycine; DLG4/PSD95: discs large MAGUK scaffold protein 4; DMEM: Dulbecco’s modified eagle’s medium; EB: embryoid body; ECAR: extracellular acidification rate; EGF: epidermal growth factor; FACS: fluorescence-activated cell sorting; FCCP: arbonyl cyanide p-triflouromethoxyphenylhydrazone; FGF: fibroblast growth factor; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GDNF: glia cell derived neurotrophic factor; hiPSC: human induced pluripotent stem cell; LAMP2A: lysosomal associated membrane protein 2A; LT-R: LysoTracker Red; MAP1LC3: microtubule associated protein 1 light chain 3; mDAN: midbrain dopaminergic neuron; MEF: mouse embryonic fibroblast; MT-GR: MitoTracker Green; MT-R: MitoTracker Red; NAS2: normal SNCA2; NEM: neuroprogenitor expansion media; NR4A2/NURR1: nuclear receptor subfamily group A member 2; OA: oligomycin and antimycin A; OCR: oxygen consumption rate; PD: Parkinson disease; SHH: sonic hedgehog signaling molecule; SNCA/α-synuclein: synuclein alpha; TH: tyrosine hydroxylase; VTN: vitronectin.
Collapse
Affiliation(s)
- Petros Stathakos
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK.,Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | | | - Lucy A Crompton
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Paul A Nistor
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Jennifer L Badger
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peter A Barbuti
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Talitha L Kerrigan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, UK.,Dementia Research Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Andrew D Randall
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, UK
| | - Maeve A Caldwell
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK.,Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Jon D Lane
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| |
Collapse
|
43
|
Homogenous generation of dopaminergic neurons from multiple hiPSC lines by transient expression of transcription factors. Cell Death Dis 2019; 10:898. [PMID: 31776327 PMCID: PMC6881336 DOI: 10.1038/s41419-019-2133-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/29/2022]
Abstract
A major hallmark of Parkinson's disease is loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). The pathophysiological mechanisms causing this relatively selective neurodegeneration are poorly understood, and thus experimental systems allowing to study dopaminergic neuron dysfunction are needed. Induced pluripotent stem cells (iPSCs) differentiated toward a dopaminergic neuronal phenotype offer a valuable source to generate human dopaminergic neurons. However, currently available protocols result in a highly variable yield of dopaminergic neurons depending on the source of hiPSCs. We have now developed a protocol based on HBA promoter-driven transient expression of transcription factors by means of adeno-associated viral (AAV) vectors, that allowed to generate very consistent numbers of dopaminergic neurons from four different human iPSC lines. We also demonstrate that AAV vectors expressing reporter genes from a neuron-specific hSyn1 promoter can serve as surrogate markers for maturation of hiPSC-derived dopaminergic neurons. Dopaminergic neurons differentiated by transcription factor expression showed aggravated neurodegeneration through α-synuclein overexpression, but were not sensitive to γ-synuclein overexpression, suggesting that these neurons are well suited to study neurodegeneration in the context of Parkinson’s disease.
Collapse
|
44
|
Peterson DJ, Marckini DN, Straight JL, King EM, Johnson W, Sarah SS, Chowdhary PK, DeLano-Taylor MK. The Basic Helix-Loop-Helix Gene Nato3 Drives Expression of Dopaminergic Neuron Transcription Factors in Neural Progenitors. Neuroscience 2019; 421:176-191. [PMID: 31672641 DOI: 10.1016/j.neuroscience.2019.09.003] [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] [Received: 01/03/2019] [Revised: 08/17/2019] [Accepted: 09/04/2019] [Indexed: 12/17/2022]
Abstract
The floor plate of the developing midbrain gives rise to dopaminergic (DA) neurons, an important class of cells involved in Parkinson's disease (PD). Neural progenitors of the midbrain floor plate utilize key genes in transcriptional networks to drive dopamine neurogenesis. Identifying factors that promote dopaminergic neuron transcriptional networks can provide insight into strategies for therapies in PD. Using the chick embryo, we developed a quantitative PCR (qPCR) based method to assess the potential of a candidate factor to drive DA neuron gene expression, including the basic helix-loop-helix transcription factor Nato3 (Ferd3l). We then showed that overexpression of Nato3 in the developing chick mesencephalon produces a regionally dependent increase in genes associated with the DA neurogenesis, (such as Foxa2, Lmx1b and Shh) as well as DA neuron genes Nurr1 (an immature DA neuron marker) and mRNA expression of tyrosine hydroxylase (TH, a mature DA neuron marker). Interestingly, our data also showed that Nato3 is a potent regulator of Lmx1b by its broad induction of Lmx1b expression in neural progenitors of multiple regions of the CNS, including the midbrain and spinal cord. These data introduce a new, in vivo approach to identifying a gene that can drive DA transcriptional networks and provide the new insight that Nato3 can drive expression of key DA neuron genes, including Lmx1b, in neural progenitors.
Collapse
Affiliation(s)
- Doug J Peterson
- Department of Biomedical Sciences, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA; Department of Cell and Molecular Biology, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA
| | - Darcy N Marckini
- Department of Cell and Molecular Biology, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA
| | - Jordan L Straight
- Department of Cell and Molecular Biology, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA
| | - Elizabeth M King
- Department of Biomedical Sciences, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA
| | - William Johnson
- Department of Cell and Molecular Biology, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA
| | - Sarala S Sarah
- Department of Biomedical Sciences, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA
| | - Puneet K Chowdhary
- Department of Biomedical Sciences, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA; Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave., Grand Rapids MI 49506, USA
| | - Merritt K DeLano-Taylor
- Department of Biomedical Sciences, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA; Department of Cell and Molecular Biology, Grand Valley State University, 1 Campus Drive, Allendale MI 49401, USA; Center for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Ave., Grand Rapids MI 49506, USA.
| |
Collapse
|
45
|
Okawa S, Del Sol A. A general computational approach to predicting synergistic transcriptional cores that determine cell subpopulation identities. Nucleic Acids Res 2019; 47:3333-3343. [PMID: 30820550 PMCID: PMC6468312 DOI: 10.1093/nar/gkz147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 12/21/2022] Open
Abstract
Advances in single-cell RNA-sequencing techniques reveal the existence of distinct cell subpopulations. Identification of transcription factors (TFs) that define the identity of these subpopulations poses a challenge. Here, we postulate that identity depends on background subpopulations, and is determined by a synergistic core combination of TFs mainly uniquely expressed in each subpopulation, but also TFs more broadly expressed across background subpopulations. Building on this view, we develop a new computational method for determining such synergistic identity cores of subpopulations within a given cell population. Our method utilizes an information-theoretic measure for quantifying transcriptional synergy, and implements a novel algorithm for searching for optimal synergistic cores. It requires only single-cell RNA-seq data as input, and does not rely on any prior knowledge of candidate genes or gene regulatory networks. Hence, it can be directly applied to any cellular systems, including those containing novel subpopulations. The method is capable of recapitulating known experimentally validated identity TFs in eight published single-cell RNA-seq datasets. Furthermore, some of these identity TFs are known to trigger cell conversions between subpopulations. Thus, this methodology can help design strategies for cell conversion within a cell population, guiding experimentalists in the field of stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Satoshi Okawa
- Integrated BioBank of Luxembourg, Dudelange L-3555, Luxembourg.,Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, L-4367 Belvaux, Luxembourg.,CIC bioGUNE, Bizkaia Technology Park, 801 building, Derio 48160, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain.,Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| |
Collapse
|
46
|
Cicadidae Periostracum, the Cast-Off Skin of Cicada, Protects Dopaminergic Neurons in a Model of Parkinson's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5797512. [PMID: 31772707 PMCID: PMC6854990 DOI: 10.1155/2019/5797512] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/30/2019] [Accepted: 08/10/2019] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is characterized by dopaminergic neuronal loss in the substantia nigra pars compacta (SNPC) and the striatum. Nuclear receptor-related 1 protein (Nurr1) is a nuclear hormone receptor implicated in limiting mitochondrial dysfunction, apoptosis, and inflammation in the central nervous system and protecting dopaminergic neurons and a promising therapeutic target for PD. Cicadidae Periostracum (CP), the cast-off skin of Cryptotympana pustulata Fabricius, has been used in traditional medicine for its many clinical pharmacological effects, including the treatment of psychological symptoms in PD. However, scientific evidence for the use of CP in neurodegenerative diseases, including PD, is lacking. Here, we investigated the protective effects of CP on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- (MPTP-) induced PD in mice and explored the underlying mechanisms of action, focusing on Nurr1. CP increased the expression levels of Nurr1, tyrosine hydroxylase, DOPA decarboxylase, dopamine transporter, and vesicular monoamine transporter 2 via extracellular signal-regulated kinase phosphorylation in differentiated PC12 cells and the mouse SNPC. In MPTP-induced PD, CP promoted recovery from movement impairments. CP prevented dopamine depletion and protected against dopaminergic neuronal degradation via mitochondria-mediated apoptotic proteins such as B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X, cytochrome c, and cleaved caspase-9 and caspase-3 by inhibiting MPTP-induced neuroinflammatory cytokines, inducible nitric oxide synthase, cyclooxygenase 2, and glial/microglial activation. Moreover, CP inhibited lipopolysaccharide-induced neuroinflammatory cytokines and response levels and glial/microglial activation in BV2 microglia and the mouse brain. Our findings suggest that CP might contribute to neuroprotective signaling by regulating neurotrophic factors primarily via Nurr1 signaling, neuroinflammation, and mitochondria-mediated apoptosis.
Collapse
|
47
|
Serotonin is elevated in risk-genotype carriers of TCF7L2 - rs7903146. Sci Rep 2019; 9:12863. [PMID: 31492908 PMCID: PMC6731216 DOI: 10.1038/s41598-019-49347-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/12/2019] [Indexed: 01/21/2023] Open
Abstract
The transcription factor 7-like 2 (TCF7L2) polymorphism rs7903146 is known to be tightly associated with an elevated risk for type 2 diabetes, whereas the molecular mechanisms remain elusive. We evaluated the metabolic profile of a total of 394 patients' serum samples with respect to their rs7903146 genotype using targeted metabolomics in a discovery (n = 154) and a validation (n = 240) study. We have identified serotonin as the top metabolite being increased in carriers of the risk allele. Serotonin was significantly associated with the rs7903146 genotype after full adjustment including type 2 diabetes and further top ranked metabolites. Given the role of peripheral serotonin in metabolic homeostasis and type 2 diabetes, this finding provides a first hint that the well-known impact of the TCF7L2 polymorphism on type 2 diabetes risk may involve a serotonin-dependent pathway.
Collapse
|
48
|
Raina A, Mahajani S, Bähr M, Kügler S. Neuronal Trans-differentiation by Transcription Factors Ascl1 and Nurr1: Induction of a Dopaminergic Neurotransmitter Phenotype in Cortical GABAergic Neurons. Mol Neurobiol 2019; 57:249-260. [PMID: 31317490 DOI: 10.1007/s12035-019-01701-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022]
Abstract
Neurons with a desired neurotransmitter phenotype can be differentiated from induced pluripotent stem cells or from somatic cells only through tedious protocols with relatively low yield. Readily available cortical neurons isolated from embryonic rat brain, which have already undergone a complete neuronal differentiation process, might serve as alternative template source. These cultures consist of 85% glutamatergic and 15% GABAergic neurons, and we attempted to trans-differentiate them into dopaminergic neurons. Transcription factors Nurr1, Lmx1A and Pitx3, essential determinants of a dopaminergic cell fate during CNS development, were not sufficient to induce tyrosine hydroxylase expression in a significant number of cells. Combining Nurr1 with the generic neuronal differentiator and re-programming factor Ascl1, however, resulted in generation of neurons which express dopaminergic markers TH, AADC, VMAT2 and DAT. Only neurons of GABAergic phenotype could be trans-differentiated towards a dopaminergic neurotransmitter phenotype, while for glutamatergic neurons, this process proved to be neurotoxic. Intriguingly, GABAergic neurons isolated from embryonal midbrain could not be trans-differentiated into dopaminergic neurons by Ascl1 and Nurr1. Thus, in principle, post-mitotic embryonal neurons can serve as templates for neurons with a desired neurotransmitter phenotype. However, neurotransmitter phenotype plasticity critically depends on the differentiation history of the template neurons, which can result in relatively low yields of dopaminergic neurons.
Collapse
Affiliation(s)
- Anupam Raina
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany.,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Sameehan Mahajani
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany.,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany.,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Sebastian Kügler
- Department of Neurology, University Medicine Göttingen, Waldweg 33, 37073, Göttingen, Germany. .,Center Nanoscale Microscopy and Physiology of the Brain (CNMPB), Göttingen, Germany.
| |
Collapse
|
49
|
Ma L, Yang Z, Ding J, Liu S, Guo B, Yue Z. Function and regulation of transforming growth factor β1 signalling in antler chondrocyte proliferation and differentiation. Cell Prolif 2019; 52:e12637. [PMID: 31168899 PMCID: PMC6668978 DOI: 10.1111/cpr.12637] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Chondrocyte proliferation and differentiation are crucial for endochondral ossification, but their regulatory mechanism remains unclear. The present study aimed to determine the physiological function of TGFβ1 signalling in the proliferation and differentiation of antler chondrocytes and explore its relationship with Notch, Shh signalling and Foxa. MATERIALS AND METHODS Immunofluorescence, Western blot, MTS assay, flow cytometry, RNA interference and real-time PCR were used to analyse the function and regulatory mechanisms of TGFβ1 signalling in antler chondrocyte proliferation and differentiation. RESULTS TGFβ1, TGFBR1 and TGFBR2 were highly expressed in antler cartilage. TGFβ1 promoted chondrocyte proliferation, increased the proportion of S-phase cells and induced the expression of hypertrophic chondrocyte markers Col X, Runx2 and Alpl. However, this induction was weakened by TGFβ receptor inhibitor SB431542 and Smad3 inhibitor SIS3. Simultaneously, TGFβ1 activated Notch and Shh signalling whose blockage attenuated the above effects of rTGFβ1, whereas addition of rShh rescued the defects in chondrocyte proliferation and differentiation elicited by SB431542 and SIS3. Further analysis revealed that inhibition of Notch signalling impeded TGFβ1 activation of the Shh pathway. Knockdown of Foxa1, Foxa2 and Foxa3 abrogated the effects of TGFβ1 on chondrocyte differentiation. Notch and Shh signalling mediated the regulation of Foxa transcription factors by TGFβ1. CONCLUSIONS TGFβ1 signalling could induce the proliferation and differentiation of antler chondrocytes through Notch-Shh-Foxa pathway.
Collapse
Affiliation(s)
- Li Ma
- College of Veterinary MedicineJilin UniversityChangchunChina
| | - Zhan‐Qing Yang
- College of Veterinary MedicineJilin UniversityChangchunChina
| | - Jun‐Li Ding
- College of Veterinary MedicineJilin UniversityChangchunChina
| | - Shu Liu
- College of Veterinary MedicineJilin UniversityChangchunChina
| | - Bin Guo
- College of Veterinary MedicineJilin UniversityChangchunChina
| | - Zhan‐Peng Yue
- College of Veterinary MedicineJilin UniversityChangchunChina
| |
Collapse
|
50
|
Brodski C, Blaess S, Partanen J, Prakash N. Crosstalk of Intercellular Signaling Pathways in the Generation of Midbrain Dopaminergic Neurons In Vivo and from Stem Cells. J Dev Biol 2019; 7:jdb7010003. [PMID: 30650592 PMCID: PMC6473842 DOI: 10.3390/jdb7010003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Dopamine-synthesizing neurons located in the mammalian ventral midbrain are at the center stage of biomedical research due to their involvement in severe human neuropsychiatric and neurodegenerative disorders, most prominently Parkinson’s Disease (PD). The induction of midbrain dopaminergic (mDA) neurons depends on two important signaling centers of the mammalian embryo: the ventral midline or floor plate (FP) of the neural tube, and the isthmic organizer (IsO) at the mid-/hindbrain boundary (MHB). Cells located within and close to the FP secrete sonic hedgehog (SHH), and members of the wingless-type MMTV integration site family (WNT1/5A), as well as bone morphogenetic protein (BMP) family. The IsO cells secrete WNT1 and the fibroblast growth factor 8 (FGF8). Accordingly, the FGF8, SHH, WNT, and BMP signaling pathways play crucial roles during the development of the mDA neurons in the mammalian embryo. Moreover, these morphogens are essential for the generation of stem cell-derived mDA neurons, which are critical for the modeling, drug screening, and cell replacement therapy of PD. This review summarizes our current knowledge about the functions and crosstalk of these signaling pathways in mammalian mDA neuron development in vivo and their applications in stem cell-based paradigms for the efficient derivation of these neurons in vitro.
Collapse
Affiliation(s)
- Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel.
| | - Sandra Blaess
- Institute of Reconstructive Neurobiology, University of Bonn Medical Center, 53127 Bonn, Germany.
| | - Juha Partanen
- Faculty of Biological and Environmental Sciences, FIN00014-University of Helsinki, P.O. Box 56, Viikinkaari 9, FIN-00014 Helsinki, Finland.
| | - Nilima Prakash
- Department Hamm 2, Hamm-Lippstadt University of Applied Sciences, 59063 Hamm, Germany.
| |
Collapse
|