1
|
Molina-Ruiz FJ, Sanders P, Gomis C, Abante J, Londoño F, Bombau G, Galofré M, Vinyes-Bassols GL, Monforte V, Canals JM. CD200-based cell sorting results in homogeneous transplantable striatal neuroblasts for human cell therapy for Huntington's disease. Neurobiol Dis 2025; 209:106905. [PMID: 40220917 DOI: 10.1016/j.nbd.2025.106905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
Neurodegenerative diseases are characterized by selective loss of neurons. Cell replacement therapies are the most promising therapeutic strategies to restore the neuronal functions lost during these neurodegenerative processes. However, cell replacement-based clinical trials for Huntington's (HD) and Parkinson's diseases (PD) failed due to the large heterogeneity of the samples. Here, we identify CD200 as a cell surface marker for human striatal neuroblasts (NBs) using massively parallel single-cell RNA sequencing. Next, we set up a CD200-based immunomagnetic sorting pipeline that allows high-yield enrichment of human striatal NBs from in vitro differentiation of human pluripotent stem cells (hPSCs). We also show that sorted CD200-positive cells are striatal projection neuron (SPN)-committed NBs which survive upon intra-striatal transplantation in adult mice with no evidence of graft overgrowth in vivo. In conclusion, we implemented a new CD200 cell selection strategy that reduces the heterogeneity and batch-to-batch variation and potentially decreases the teratogenic risk of hPSC-based cell therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Francisco J Molina-Ruiz
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Phil Sanders
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Cinta Gomis
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Jordi Abante
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Department of Biomedical Data Science, Stanford University, Stanford, CA, United States of America
| | - Francisco Londoño
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Georgina Bombau
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Mireia Galofré
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Gal la Vinyes-Bassols
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Veronica Monforte
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Josep M Canals
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Institute of Neurosciences; and Creatio, Production and Validation Center of Advanced Therapies, University of Barcelona, 08036 Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain.
| |
Collapse
|
2
|
Mishra AK, Dixit S, Singh A, Shukla T, Rizvi SI. Molecular Determinants of A9 Dopaminergic Neurons. Neuromolecular Med 2025; 27:43. [PMID: 40397062 DOI: 10.1007/s12017-025-08861-1] [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: 03/11/2025] [Accepted: 05/02/2025] [Indexed: 05/22/2025]
Abstract
In the human brain, the nigrostriatal pathway regulates motor functions, and its selective deterioration leads to the onset of Parkinson's disease (PD), a neurodegenerative disorder characterized by motor dysfunction and significant disability. The A9 neurons, a subgroup of ventral mesencephalic dopaminergic (DA) neurons, form the nigrostriatal pathway that emerges from the nigral region and innervates into the striatum. These DA neurons exhibit extensive and arborized axonal terminals projecting into the dorsal striatum. This review examines the distinct molecular determinants underlying the development, projection pattern, survival, maintenance, and vulnerability of A9 neurons, distinguishing them from other ventral midbrain DA subgroups such as A8 and A10. Key transcription factors (e.g., Lmx1a/b, FoxA2, Pitx3), signaling cascade pathways (e.g., Sonic Hedgehog, Wnt/β-catenin), and molecular markers (e.g., Aldh1a1, GIRK2, ANT2) are discussed in detail. A comparative assessment of the electrophysiology, cytoarchitecture, energy demand, and antioxidant reserves of A9 DA neurons versus the neighboring ventral mesencephalic DA subgroups elucidates the role of intrinsic determinants in susceptibility and selective degeneration in PD. The unique susceptibility of A9 cells to redox imbalance, neuronal inflammation, and mitochondrial dysfunction is also explored. Furthermore, recent advancements in stem cell-based approaches for generating A9-like neurons and their application in cell transplantation therapies for PD are discussed. Current challenges, including integration and long-term survival of transplanted neurons, are highlighted alongside prospects of cell replacement therapy. By evaluating the molecular biology of A9 neurons, this review aims to understand PD pathology and develop strategies for novel therapeutic approaches.
Collapse
Affiliation(s)
- Abhishek Kumar Mishra
- Department of Zoology, Government Shaheed Gendsingh College, Charama, Uttar Bastar Kanker, Chhattisgarh, 494 337, India.
| | - Shreya Dixit
- Department of Neurology, University of California, Irvine, USA
| | - Akanksha Singh
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| | - Toyaj Shukla
- Government Rani Durgawati College, Wadrafnagar, Balrampur, Chhattisgarh, India
| | - Syed Ibrahim Rizvi
- Department of Biochemistry, University of Allahabad, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
3
|
Ballion B, Bonnet ML, Brot S, Gaillard A. Electrophysiological characterisation of intranigral-grafted hiPSC-derived dopaminergic neurons in a mouse model of Parkinson's disease. Stem Cell Res Ther 2025; 16:232. [PMID: 40346597 PMCID: PMC12065326 DOI: 10.1186/s13287-025-04344-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a complex neurological disorder characterized by the progressive degeneration of midbrain dopaminergic (mDA) neurons in the substantia nigra (SN). This degeneration disrupts the basal ganglia loops, leading to both motor and non-motor dysfunctions. Cell therapy for PD aims to replace lost mDA neurons to restore the DA neurotransmission in the denervated forebrain targets. In clinical trials for PD, mDA neurons are implanted into the target area, the striatum, and not in the SN where they are normally located. This ectopic localisation of cells may affect the functionality of transplanted neurons due to the absence of appropriate host afferent regulation. We recently demonstrated that human induced pluripotent stem cells (hiPSCs) derived mDA progenitors grafted into the substantia nigra pars compacta (SNpc) in a mouse model of PD, differentiated into mature mDA neurons, restored the degenerated nigrostriatal pathway, and induced motor recovery. The objective of the present study was to evaluate the long-term functionality of these intranigral-grafted mDA neurons by assessing their electrophysiological properties. METHODS We performed intranigral transplantation of hiPSC-derived mDA progenitors in a 6-hydroxydopamine RAG2-KO mouse model of PD. We recorded in vivo unit extracellular activity of grafted mDA neurons in anesthetised mice from 9 to 12 months post-transplantation. Their electrophysiological properties, including firing rates, patterns and spike characteristics, were analysed and compared with those of native nigral dopaminergic neurons from control mice. RESULTS We demonstrated that these grafted mDA neurons exhibited functional characteristics similar to those of native nigral dopaminergic neurons, such as large bi- or triphasic spike waveforms, low firing rates, pacemaker-like properties, and two single-spike firing patterns. Although grafted mDA neurons also displayed low discharge frequencies below 10 Hz, their mean frequency was significantly lower than that of nigral mDA neurons, with a differential pattern distribution. CONCLUSIONS Our findings indicate that grafted mDA neurons exhibit dopaminergic-like functional properties, including intrinsic membrane potential oscillations leading to regular firing patterns. Additionally, they demonstrated irregular and burst firing patterns, suggesting they receive modulatory inputs. However, grafted mDA neurons displayed distinct properties, potentially related to their human origin or the incomplete maturation one year after transplantation.
Collapse
Affiliation(s)
- Bérengère Ballion
- Laboratoire des neurosciences expérimentales et cliniques (LNEC), Université de Poitiers- INSERM 1084, Poitiers Cedex 9, 86073, France.
| | - Marie-Laure Bonnet
- Laboratoire des neurosciences expérimentales et cliniques (LNEC), Université de Poitiers- INSERM 1084, Poitiers Cedex 9, 86073, France
- Centre hospitalier universitaire (CHU) de Poitiers, Poitiers, 86021, France
| | - Sébastien Brot
- Laboratoire des neurosciences expérimentales et cliniques (LNEC), Université de Poitiers- INSERM 1084, Poitiers Cedex 9, 86073, France
| | - Afsaneh Gaillard
- Laboratoire des neurosciences expérimentales et cliniques (LNEC), Université de Poitiers- INSERM 1084, Poitiers Cedex 9, 86073, France.
| |
Collapse
|
4
|
Matiukhova M, Ryapolova A, Andriianov V, Reshetnikov V, Zhuravleva S, Ivanov R, Karabelsky A, Minskaia E. A comprehensive analysis of induced pluripotent stem cell (iPSC) production and applications. Front Cell Dev Biol 2025; 13:1593207. [PMID: 40406420 PMCID: PMC12095295 DOI: 10.3389/fcell.2025.1593207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
The ability to reprogram mature, differentiated cells into induced pluripotent stem cells (iPSCs) using exogenous pluripotency factors opened up unprecedented opportunities for their application in biomedicine. iPSCs are already successfully used in cell and regenerative therapy, as various drug discovery platforms and for in vitro disease modeling. However, even though already 20 years have passed since their discovery, the production of iPSC-based therapies is still associated with a number of hurdles due to low reprogramming efficiency, the complexity of accurate characterization of the resulting colonies, and the concerns associated with the safety of this approach. However, significant progress in many areas of molecular biology facilitated the production, characterization, and thorough assessment of the safety profile of iPSCs. The number of iPSC-based studies has been steadily increasing in recent years, leading to the accumulation of significant knowledge in this area. In this review, we aimed to provide a comprehensive analysis of methods used for reprogramming and subsequent characterization of iPSCs, discussed barriers towards achieving these goals, and various approaches to improve the efficiency of reprogramming of different cell populations. In addition, we focused on the analysis of iPSC application in preclinical and clinical studies. The accumulated breadth of data helps to draw conclusions about the future of this technology in biomedicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ekaterina Minskaia
- Translational Medicine Research Center, Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
5
|
Barker RA, Lao-Kaim NP, Guzman NV, Athauda D, Bjartmarz H, Björklund A, Church A, Cutting E, Daft D, Dayal V, Dunnett S, Evans A, Grealish S, Hannaway N, He X, Hewitt S, Kefalopoulou Z, Mahlknecht P, Martín-Bastida A, Farrell K, Moore S, Bulstrode H, Nakornchai T, Nelander-Wahlestedt J, Roupé L, Paul G, Peall K, Rosser A, Roca-Fernández A, Rowlands S, McGorrian AM, Scherf C, Vinh NN, Roberton V, Kelly C, Lelos M, Torres E, Shires K, Hills R, Williams D, Roussakis AA, Sibley K, Tyers P, Wijeyekoon R, Williams-Gray C, Foltynie T, Piccini P, Morris R, Lazic SE, Lindvall O, Parmar M, Widner H. The TransEuro open-label trial of human fetal ventral mesencephalic transplantation in patients with moderate Parkinson's disease. Nat Biotechnol 2025:10.1038/s41587-025-02567-2. [PMID: 40316701 DOI: 10.1038/s41587-025-02567-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/17/2025] [Indexed: 05/04/2025]
Abstract
Transplantation of human fetal ventral mesencephalic tissue in individuals with Parkinson's disease has yielded clinical benefits but also side effects, such as graft-induced dyskinesias. The open-label TransEuro trial ( NCT01898390 ) was designed to determine whether this approach could be further developed into a clinically useful treatment. Owing to poor availability of human fetal ventral mesencephalic tissue, only 11 individuals were grafted at two centers using the same tissue preparation protocol but different implantation devices. No overall clinical effect was seen for the primary endpoint 3 years after grafting. No major graft-induced dyskinesias were seen, but we observed differences in outcome related to transplant device and/or site. Mean dopamine uptake improved at 18 months in seven individuals according to [18F]fluorodopa positron emission tomography imaging but was restored to near-normal levels in only one individual. Our findings highlight the need for a stem cell source of dopamine neurons for potential Parkinson's disease cell therapy and provide critical insights into how such clinical studies should be approached.
Collapse
Affiliation(s)
- Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - Nicholas P Lao-Kaim
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Natalie Valle Guzman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Dilan Athauda
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | - Hjalmar Bjartmarz
- Department of Neurosurgery, Skånes University Hospital and Lund University, Lund, Sweden
| | - Anders Björklund
- Department of Neurology, Skånes University Hospital and Lund University, Lund, Sweden
| | - Alistair Church
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Emma Cutting
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Danielle Daft
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Viswas Dayal
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | - Stephen Dunnett
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Amy Evans
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Shane Grealish
- Department of Neurology, Skånes University Hospital and Lund University, Lund, Sweden
| | - Naomi Hannaway
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Xiaoling He
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Sam Hewitt
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Zinovia Kefalopoulou
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | - Philipp Mahlknecht
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | | | - Krista Farrell
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Sarah Moore
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Harry Bulstrode
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Tagore Nakornchai
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | | - Linnea Roupé
- Department of Neurology, Skånes University Hospital and Lund University, Lund, Sweden
| | - Gesine Paul
- Department of Neurology, Skånes University Hospital and Lund University, Lund, Sweden
| | - Kathryn Peall
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Anne Rosser
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | | | - Sophie Rowlands
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Anne-Marie McGorrian
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Caroline Scherf
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Ngoc Nga Vinh
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Victoria Roberton
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Claire Kelly
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Mariah Lelos
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Eduardo Torres
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Kate Shires
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Rachel Hills
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Debbie Williams
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | | | - Krista Sibley
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | - Pamela Tyers
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Ruwani Wijeyekoon
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Caroline Williams-Gray
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Thomas Foltynie
- Department of Clinical & Movement Neurosciences, UCL Institute of Neurology, London, UK
| | - Paola Piccini
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Robert Morris
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Stanley E Lazic
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Olle Lindvall
- Lund Stem Cell Center and Division of Neurology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Malin Parmar
- Department of Neurosurgery, Skånes University Hospital and Lund University, Lund, Sweden
- Lund Stem Cell Center and Division of Neurology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Hakan Widner
- Department of Neurosurgery, Skånes University Hospital and Lund University, Lund, Sweden
| |
Collapse
|
6
|
Verma I, Seshagiri PB. Current Applications of Human Pluripotent Stem Cells in Neuroscience Research and Cell Transplantation Therapy for Neurological Disorders. Stem Cell Rev Rep 2025; 21:964-987. [PMID: 40186708 DOI: 10.1007/s12015-025-10851-6] [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] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Many neurological diseases involving tissue damage cannot be treated with drug-based approaches, and the inaccessibility of human brain samples further hampers the study of these diseases. Human pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), provide an excellent model for studying neural development and function. PSCs can be differentiated into various neural cell types, providing a renewal source of functional human brain cells. Therefore, PSC-derived neural cells are increasingly used for multiple applications, including neurodevelopmental and neurotoxicological studies, neurological disease modeling, drug screening, and regenerative medicine. In addition, the neural cells generated from patient iPSCs can be used to study patient-specific disease signatures and progression. With the recent advances in genome editing technologies, it is possible to remove the disease-related mutations in the patient iPSCs to generate corrected iPSCs. The corrected iPSCs can differentiate into neural cells with normal physiological functions, which can be used for autologous transplantation. This review highlights the current progress in using PSCs to understand the fundamental principles of human neurodevelopment and dissect the molecular mechanisms of neurological diseases. This knowledge can be applied to develop better drugs and explore cell therapy options. We also discuss the basic requirements for developing cell transplantation therapies for neurological disorders and the current status of the ongoing clinical trials.
Collapse
Affiliation(s)
- Isha Verma
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Neurology, University of Michigan, Ann Arbor, 48109, USA.
| | - Polani B Seshagiri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
7
|
Sawamoto N, Doi D, Nakanishi E, Sawamura M, Kikuchi T, Yamakado H, Taruno Y, Shima A, Fushimi Y, Okada T, Kikuchi T, Morizane A, Hiramatsu S, Anazawa T, Shindo T, Ueno K, Morita S, Arakawa Y, Nakamoto Y, Miyamoto S, Takahashi R, Takahashi J. Phase I/II trial of iPS-cell-derived dopaminergic cells for Parkinson's disease. Nature 2025; 641:971-977. [PMID: 40240591 PMCID: PMC12095070 DOI: 10.1038/s41586-025-08700-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/24/2025] [Indexed: 04/18/2025]
Abstract
Parkinson's disease is caused by the loss of dopamine neurons, causing motor symptoms. Initial cell therapies using fetal tissues showed promise but had complications and ethical concerns1-5. Pluripotent stem (PS) cells emerged as a promising alternative for developing safe and effective treatments6. In this phase I/II trial at Kyoto University Hospital, seven patients (ages 50-69) received bilateral transplantation of dopaminergic progenitors derived from induced PS (iPS) cells. Primary outcomes focused on safety and adverse events, while secondary outcomes assessed motor symptom changes and dopamine production for 24 months. There were no serious adverse events, with 73 mild to moderate events. Patients' anti-parkinsonian medication doses were maintained unless therapeutic adjustments were required, resulting in increased dyskinesia. Magnetic resonance imaging showed no graft overgrowth. Among six patients subjected to efficacy evaluation, four showed improvements in the Movement Disorder Society Unified Parkinson's Disease Rating Scale part III OFF score, and five showed improvements in the ON scores. The average changes of all six patients were 9.5 (20.4%) and 4.3 points (35.7%) for the OFF and ON scores, respectively. Hoehn-Yahr stages improved in four patients. Fluorine-18-L-dihydroxyphenylalanine (18F-DOPA) influx rate constant (Ki) values in the putamen increased by 44.7%, with higher increases in the high-dose group. Other measures showed minimal changes. This trial (jRCT2090220384) demonstrated that allogeneic iPS-cell-derived dopaminergic progenitors survived, produced dopamine and did not form tumours, therefore suggesting safety and potential clinical benefits for Parkinson's disease.
Collapse
Affiliation(s)
- Nobukatsu Sawamoto
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Doi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Etsuro Nakanishi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masanori Sawamura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayuki Kikuchi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hodaka Yamakado
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yosuke Taruno
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Atsushi Shima
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasutaka Fushimi
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomohisa Okada
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tetsuhiro Kikuchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Satoe Hiramatsu
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Takayuki Anazawa
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takero Shindo
- Department of Hematology/Oncology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kentaro Ueno
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.
| |
Collapse
|
8
|
Emborg ME, Metzger JM, D'Amour K, Colwell JC, Neumann LC, Zhang A, Federoff HJ. Advantages and challenges of using allogeneic vs. autologous sources for neuronal cell replacement in Parkinson's disease: Insights from non-human primate studies. Brain Res Bull 2025; 224:111297. [PMID: 40086764 PMCID: PMC12036832 DOI: 10.1016/j.brainresbull.2025.111297] [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: 09/25/2024] [Revised: 02/25/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Intracerebral grafting of dopamine-producing cells is proposed as a strategy to replace the typical neurons lost to Parkinson's disease (PD) and improve PD motor symptoms. Non-human primate studies have provided clues on the relationship between the host's immune response and grafting success. Herein, we discuss how the host's immune system differentially affects the graft depending on the origin of the cells and reflect on the advantages and limitations of the immune paradigms utilized to assess graft-related outcomes. We also consider new strategies to minimize or circumvent the host's immunological response and related preclinical research needed to identify the most promising new approaches to be translated into the clinic.
Collapse
Affiliation(s)
- Marina E Emborg
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Department of Medical Physics, University of Wisconsin-Madison, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, USA.
| | - Jeanette M Metzger
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | | | - Julia C Colwell
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, USA
| | - Lindsey C Neumann
- Preclinical Parkinson's Research Program, Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | - Ai Zhang
- Genentech, South San Francisco, CA, USA
| | - Howard J Federoff
- Kenai Therapeutics, San Diego, CA, USA; Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
9
|
Boschen SL, A Mukerjee A, H Faroqi A, E Rabichow B, Fryer J. Research models to study lewy body dementia. Mol Neurodegener 2025; 20:46. [PMID: 40269912 PMCID: PMC12020038 DOI: 10.1186/s13024-025-00837-w] [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: 12/18/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Lewy body dementia (LBD) encompasses neurodegenerative dementias characterized by cognitive fluctuations, visual hallucinations, and parkinsonism. Clinical differentiation of LBD from Alzheimer's disease (AD) remains complex due to symptom overlap, yet approximately 25% of dementia cases are diagnosed as LBD postmortem, primarily identified by the presence of α-synuclein aggregates, tau tangles, and amyloid plaques. These pathological features position LBD as a comorbid condition of both Parkinson's disease (PD) and AD, with over 50% of LBD cases exhibiting co-pathologies. LBD's mixed pathology complicates the development of comprehensive models that reflect the full spectrum of LBD's etiological, clinical, and pathological features. While existing animal and cellular models have facilitated significant discoveries in PD and AD research, they lack specificity in capturing LBD's unique pathogenic mechanisms, limiting the exploration of therapeutic avenues for LBD specifically. This review assesses widely used PD and AD models in terms of their relevance to LBD, particularly focusing on their ability to replicate human disease pathology and assess treatment efficacy. Furthermore, we discuss potential modifications to these models to advance the understanding of LBD mechanisms and propose innovative research directions aimed at developing models with enhanced etiological, face, predictive, and construct validity.
Collapse
Affiliation(s)
- Suelen Lucio Boschen
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
- Department of Neurosurgery, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| | - Aarushi A Mukerjee
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Ayman H Faroqi
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ben E Rabichow
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - John Fryer
- Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 850054, USA
| |
Collapse
|
10
|
Watanabe T, La Shu S, Rio-Espinola AD, Ferreira JR, Bando K, Lemmens M, Pande P, de Wolf C, Chen CL, Elke E, Rao GK, van den Hoorn T, Mouriès LP, Myers MB, Yasuda S. Evaluating teratoma formation risk of pluripotent stem cell-derived cell therapy products: a consensus recommendation from the Health and Environmental Sciences Institute's International Cell Therapy Committee. Cytotherapy 2025:S1465-3249(25)00684-X. [PMID: 40392167 DOI: 10.1016/j.jcyt.2025.04.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 05/22/2025]
Abstract
Human pluripotent stem cells (hPSCs) can differentiate into any cell of choice and hold significant promise in regenerative medicine and for treating diseases that currently lack adequate therapies. However, hPSCs are intrinsically tumorigenic and can form teratomas. Therefore, the presence of residual undifferentiated hPSCs must be rigorously assessed using sensitive methodologies to mitigate the potential tumorigenicity risks of hPSC-derived cell therapy products (CTPs). In this comprehensive review, we describe methods for detecting residual undifferentiated hPSCs and discuss the relative value of current in vitro assays versus conventional in vivo assays. We highlight that in vitro assays such as digital PCR detection of hPSC-specific RNA and the highly efficient culture assay, have superior detection sensitivity. Additionally, we outline important considerations for validating in vitro assays when applying them to assess each product. This article lays the groundwork for guiding internationally harmonized procedures for evaluating the potential teratoma formation risk of hPSC-derived CTPs and increasing confidence in the safety of these products.
Collapse
Affiliation(s)
- Takeshi Watanabe
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan.
| | - Shin La Shu
- Frederick National Laboratories for Cancer Research, Maryland, USA
| | | | - Joana Rita Ferreira
- Safety Sciences, Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kiyoko Bando
- Regenerative & Cellular Medicine Office, Sumitomo Pharma Co., Ltd., Kobe, Japan
| | - Myriam Lemmens
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Parimal Pande
- Johnson & Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | | | - Connie L Chen
- Health & Environmental Sciences Institute (HESI), Washington DC, USA
| | - Ericson Elke
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Gautham K Rao
- Department of Translational Safety, Genentech Inc., South San Francisco, California, USA
| | | | | | - Meagan B Myers
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arizona, USA
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
11
|
Atkinson EA, Gregory HN, Carter LN, Evans RE, Roberton VH, Dickman R, Phillips JB. An immunomodulatory encapsulation system to deliver human iPSC-derived dopaminergic neuron progenitors for Parkinson's disease treatment. Biomater Sci 2025; 13:2012-2025. [PMID: 40013398 DOI: 10.1039/d4bm01566e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Parkinson's disease is a neurodegenerative condition associated with the progressive loss of dopaminergic neurons. This leads to neurological impairments with heightening severity and is globally increasing in prevalence due to population ageing. Cell transplantation has demonstrated significant promise in altering the disease course in the clinic, and stem cell-derived grafts are being investigated. Current clinical protocols involve systemic immunosuppression to prevent graft rejection, which could potentially be avoided by encapsulating the therapeutic cells in a locally immunosuppressive biomaterial matrix before delivery. Here we report the progression of an immunomodulatory encapsulation system employing ultrapure alginate hydrogel beads alongside tacrolimus-loaded microparticles in the encapsulation of dopaminergic neuron progenitors derived from human induced pluripotent stem cells (hiPSCs). The hiPSC-derived progenitors were characterised and displayed robust viability after encapsulation within alginate beads, producing dopamine as they matured in vitro. The encapsulation system effectively reduced T cell activation (3-fold) and protected progenitors from cytotoxicity in vitro. The alginate bead diameter was optimised using microfluidics to yield spherical and monodisperse hydrogels with a median size of 215.6 ± 0.5 μm, suitable for delivery to the brain through a surgical cannula. This technology has the potential to advance cell transplantation by locally protecting grafts from the host immune system.
Collapse
Affiliation(s)
- Emily A Atkinson
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Holly N Gregory
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Lara N Carter
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Rachael E Evans
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Victoria H Roberton
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Rachael Dickman
- UCL School of Pharmacy, University College London, London, UK.
| | - James B Phillips
- UCL School of Pharmacy, University College London, London, UK.
- UCL Centre for Nerve Engineering, University College London, London, UK
| |
Collapse
|
12
|
Deng S, Xie H, Xie B. Cell-based regenerative and rejuvenation strategies for treating neurodegenerative diseases. Stem Cell Res Ther 2025; 16:167. [PMID: 40189500 PMCID: PMC11974143 DOI: 10.1186/s13287-025-04285-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/19/2025] [Indexed: 04/09/2025] Open
Abstract
Neurodegenerative diseases including Alzheimer's and Parkinson's disease are age-related disorders which severely impact quality of life and impose significant societal burdens. Cellular senescence is a critical factor in these disorders, contributing to their onset and progression by promoting permanent cell cycle arrest and reducing cellular function, affecting various types of cells in brain. Recent advancements in regenerative medicine have highlighted "R3" strategies-rejuvenation, regeneration, and replacement-as promising therapeutic approaches for neurodegeneration. This review aims to critically analyze the role of cellular senescence in neurodegenerative diseases and organizes therapeutic approaches within the R3 regenerative medicine paradigm. Specifically, we examine stem cell therapy, direct lineage reprogramming, and partial reprogramming in the context of R3, emphasizing how these interventions mitigate cellular senescence and counteracting aging-related neurodegeneration. Ultimately, this review seeks to provide insights into the complex interplay between cellular senescence and neurodegeneration while highlighting the promise of cell-based regenerative strategies to address these debilitating conditions.
Collapse
Affiliation(s)
- Sixiu Deng
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, China
- Department of Gastroenterology, The Shapingba Hospital, Chongqing University( People's Hospital of Shapingba District), Chongqing, China
| | - Huangfan Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, China.
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
13
|
Strell P, Waldron MA, Johnson S, Shetty A, Crane AT, Steer CJ, Low WC. Characterization of the intraspecies chimeric mouse brain at embryonic day 12.5. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646380. [PMID: 40236149 PMCID: PMC11996362 DOI: 10.1101/2025.03.31.646380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Incidence of neurodegenerative diseases such as Alzheimer's, Parkinson's, Huntington's, and amyotrophic lateral sclerosis have increased dramatically as life expectancy at birth has risen year-over-year and the population ages. Neurological changes within the central nervous system, specifically the brain, include cell loss and deterioration that impact motor function, memory, executive function, and mood. Available treatments are limited and often only address symptomatic manifestations of the disease rather than disease progression. Cell transplantation therapy has shown promise for treating neurodegenerative diseases, but a source of autologous cells is required. Blastocyst complementation provides an innovative method for generating those autologous neural cells. By injecting mouse induced pluripotent stem cells (iPSCs) into a wild type (WT) mouse blastocyst, we generated a chimeric mouse brain derived of both donor and host neuronal and non-neuronal cells. An embryonic day 12.5 (E12.5), automated image analysis of mouse-mouse chimeric brains showed the presence of GFP-labeled donor-derived dopaminergic and serotonergic neuronal precursors. GFP-labeled donor-derived cholinergic precursor neurons and non-neuronal microglia-like and macrophage-like cells were also observed using more conventional imaging analysis software. This work demonstrates that the generation of mouse-mouse chimeric neural cells is possible; and that characterization of early neuronal and non-neuronal precursors provides a first step towards utilizing these cells for cell transplantation therapies for neurodegenerative diseases.
Collapse
|
14
|
Zhu F, Nie G. Cell reprogramming: methods, mechanisms and applications. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:12. [PMID: 40140235 PMCID: PMC11947411 DOI: 10.1186/s13619-025-00229-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/05/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025]
Abstract
Cell reprogramming represents a powerful approach to achieve the conversion cells of one type into cells of another type of interest, which has substantially changed the landscape in the field of developmental biology, regenerative medicine, disease modeling, drug discovery and cancer immunotherapy. Cell reprogramming is a complex and ordered process that involves the coordination of transcriptional, epigenetic, translational and metabolic changes. Over the past two decades, a range of questions regarding the facilitators/barriers, the trajectories, and the mechanisms of cell reprogramming have been extensively investigated. This review summarizes the recent advances in cell reprogramming mediated by transcription factors or chemical molecules, followed by elaborating on the important roles of biophysical cues in cell reprogramming. Additionally, this review will detail our current understanding of the mechanisms that govern cell reprogramming, including the involvement of the recently discovered biomolecular condensates. Finally, the review discusses the broad applications and future directions of cell reprogramming in developmental biology, disease modeling, drug development, regenerative/rejuvenation therapy, and cancer immunotherapy.
Collapse
Affiliation(s)
- Fei Zhu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing, 100190, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
15
|
Lahti L, Volakakis N, Gillberg L, Yaghmaeian Salmani B, Tiklová K, Kee N, Lundén-Miguel H, Werkman M, Piper M, Gronostajski R, Perlmann T. Sox9 and nuclear factor I transcription factors regulate the timing of neurogenesis and ependymal maturation in dopamine progenitors. Development 2025; 152:dev204421. [PMID: 39995267 DOI: 10.1242/dev.204421] [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: 09/26/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
Correct timing of neurogenesis is crucial for generating the correct number and subtypes of glia and neurons in the embryo, and for preventing tumours and stem cell depletion in the adults. Here, we analyse how the midbrain dopamine (mDA) neuron progenitors transition into cell cycle arrest (G0) and begin to mature into ependymal cells. Comparison of mDA progenitors from different embryonic stages revealed upregulation of the genes encoding Sox9 and nuclear factor I transcription factors during development. Their conditional inactivation in the early embryonic midbrain led to delayed G0 entry and ependymal maturation in the entire midbrain ventricular zone, reduced gliogenesis and increased generation of neurons, including mDA neurons. In contrast, their inactivation in late embryogenesis did not result in mitotic re-entry, suggesting that these factors are necessary for G0 induction, but not for its maintenance. Our characterisation of adult ependymal cells by single-cell RNA sequencing and histology show that mDA-progenitor-derived cells retain several progenitor features but also secrete neuropeptides and contact neighbouring cells and blood vessels, indicating that these cells may form part of the circumventricular organ system.
Collapse
Affiliation(s)
- Laura Lahti
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Linda Gillberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Katarína Tiklová
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Nigel Kee
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Maarten Werkman
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Michael Piper
- The School of Biomedical Sciences and The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Richard Gronostajski
- Genetics, Genomics & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Ludwig Institute for Cancer Research, 171 77 Stockholm, Sweden
| |
Collapse
|
16
|
Holm Nygaard A, Schörling AL, Abay-Nørgaard Z, Hänninen E, Li Y, Ramón Santonja A, Rathore GS, Salvador A, Rusimbi C, Lauritzen KB, Zhang Y, Kirkeby A. Patterning effects of FGF17 and cAMP on generation of dopaminergic progenitors for cell replacement therapy in Parkinson's disease. Stem Cells 2025; 43:sxaf004. [PMID: 40071608 PMCID: PMC11976395 DOI: 10.1093/stmcls/sxaf004] [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: 07/08/2024] [Accepted: 01/06/2025] [Indexed: 04/09/2025]
Abstract
Cell replacement therapies using human pluripotent stem cell-derived ventral midbrain (VM) dopaminergic (DA) progenitors are currently in clinical trials for treatment of Parkinson's disease (PD). Recapitulating developmental patterning cues, such as fibroblast growth factor 8 (FGF8), secreted at the midbrain-hindbrain boundary (MHB), is critical for the in vitro production of authentic VM DA progenitors. Here, we explored the application of alternative MHB-secreted FGF-family members, FGF17 and FGF18, for VM DA progenitor patterning. We show that while FGF17 and FGF18 both recapitulated VM DA progenitor patterning events, FGF17 induced expression of key VM DA progenitor markers at higher levels than FGF8 and transplanted FGF17-patterned progenitors fully reversed motor deficits in a rat PD model. Early activation of the cAMP pathway mimicked FGF17-induced patterning, although strong cAMP activation came at the expense of EN1 expression. In summary, we identified FGF17 as a promising alternative FGF candidate for robust VM DA progenitor patterning.
Collapse
Affiliation(s)
- Amalie Holm Nygaard
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alrik L Schörling
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
| | - Zehra Abay-Nørgaard
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Erno Hänninen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yuan Li
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
| | - Adrian Ramón Santonja
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Gaurav Singh Rathore
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alison Salvador
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Charlotte Rusimbi
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Katrine Bech Lauritzen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yu Zhang
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
17
|
Jeon J, Cha Y, Hong YJ, Lee IH, Jang H, Ko S, Naumenko S, Kim M, Ryu HL, Shrestha Z, Lee N, Park TY, Park H, Kim SH, Yoon KJ, Song B, Schweitzer J, Herrington TM, Kong SW, Carter B, Leblanc P, Kim KS. Pre-clinical safety and efficacy of human induced pluripotent stem cell-derived products for autologous cell therapy in Parkinson's disease. Cell Stem Cell 2025; 32:343-360.e7. [PMID: 39952239 PMCID: PMC11980241 DOI: 10.1016/j.stem.2025.01.006] [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: 05/21/2024] [Revised: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 02/17/2025]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived midbrain dopaminergic cells (mDACs) represent a promising source for autologous cell therapy in Parkinson's disease (PD), but standardized regulatory criteria are essential for clinical translation. In this pre-clinical study, we generated multiple clinical-grade hiPSC lines from freshly biopsied fibroblasts of four sporadic PD patients using episomal reprogramming and differentiated them into mDACs using a refined 21-day protocol. Rigorous evaluations included whole-genome/exome sequencing, RNA sequencing, and in vivo studies, including a 39-week Good Laboratory Practice-compliant mouse safety study. While mDACs from all lines met safety criteria, mDACs from one patient failed to improve rodent behavioral outcomes, underscoring inter-individual variability. Importantly, in vitro assessments did not reliably predict in vivo efficacy, identifying dopaminergic fiber density as a key efficacy criterion. These findings support comprehensive quality control guidelines for autologous cell therapy and pave the way for a clinical trial with eight sporadic PD patients, scheduled to commence in 2025.
Collapse
Affiliation(s)
- Jeha Jeon
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Young Cha
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Yean Ju Hong
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - In-Hee Lee
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Heejin Jang
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Sanghyeok Ko
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Serhiy Naumenko
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA 02215, USA
| | - Minseon Kim
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Hannah L Ryu
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Zenith Shrestha
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Nayeon Lee
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - Tae-Yoon Park
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA
| | - HoeWon Park
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Seo-Hyun Kim
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Bin Song
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey Schweitzer
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Todd M Herrington
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sek Won Kong
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bob Carter
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Pierre Leblanc
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA.
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, McLean Hospital and Department of Psychiatry, Harvard Medical School, Belmont, MA 02478, USA; Program in Neuroscience, Harvard Medical School, Belmont, MA 02478, USA; Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Harvard Stem Cell Institute, Harvard Medical School, Cambridge, MA 02138, USA.
| |
Collapse
|
18
|
Izrael M, Chebath J, Molakandov K, Revel M. Clinical perspective on pluripotent stem cells derived cell therapies for the treatment of neurodegenerative diseases. Adv Drug Deliv Rev 2025; 218:115525. [PMID: 39880333 DOI: 10.1016/j.addr.2025.115525] [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: 09/24/2024] [Revised: 01/09/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Self-renewal capacity and potential to differentiate into almost any cell type of the human body makes pluripotent stem cells a valuable starting material for manufacturing of clinical grade cell therapies. Neurodegenerative diseases are characterized by gradual loss of structure or function of neurons, often leading to neuronal death. This results in gradual decline of cognitive, motor, and physiological functions due to the degeneration of the central nervous systems. Over the past two decades, comprehensive preclinical efficacy (proof-of-concept) and safety studies have led to the initiation of First-in-Human phase I-II clinical trials for a range of neurodegenerative diseases. In this review, we explore the fundamentals and challenges of neural-cell therapies derived from pluripotent stem cells for treating neurodegenerative diseases. Additionally, we highlight key preclinical investigations that paved the way for regulatory approvals of these trials. Furthermore, we provide an overview on progress and status of clinical trials done so far in treating neurodegenerative diseases such as spinal cord injury (SCI), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), as well as advances in retina diseases such as Stargardt disease (a.k.a fundus flavimaculatus), retinitis pigmentosa (RP) and age-related macular degeneration (AMD). These trials will pave the way for the development of new cell-based therapies targeting additional neurological conditions, including Alzheimer's disease and epilepsy.
Collapse
Affiliation(s)
- Michal Izrael
- Neurodegenerative Diseases Department, Kadimastem Ltd, Pinchas Sapir 7, Weizmann Science Park, Ness-Ziona, Israel.
| | - Judith Chebath
- Neurodegenerative Diseases Department, Kadimastem Ltd, Pinchas Sapir 7, Weizmann Science Park, Ness-Ziona, Israel
| | - Kfir Molakandov
- Neurodegenerative Diseases Department, Kadimastem Ltd, Pinchas Sapir 7, Weizmann Science Park, Ness-Ziona, Israel
| | - Michel Revel
- Neurodegenerative Diseases Department, Kadimastem Ltd, Pinchas Sapir 7, Weizmann Science Park, Ness-Ziona, Israel; Department of Molecular Genetics, Weizmann Institute of Science, 76100, Rehovot, Israel
| |
Collapse
|
19
|
Calvo B, Schembri-Wismayer P, Durán-Alonso MB. Age-Related Neurodegenerative Diseases: A Stem Cell's Perspective. Cells 2025; 14:347. [PMID: 40072076 PMCID: PMC11898746 DOI: 10.3390/cells14050347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Neurodegenerative diseases encompass a number of very heterogeneous disorders, primarily characterized by neuronal loss and a concomitant decline in neurological function. Examples of this type of clinical condition are Alzheimer's Disease, Parkinson's Disease, Huntington's Disease and Amyotrophic Lateral Sclerosis. Age has been identified as a major risk in the etiology of these disorders, which explains their increased incidence in developed countries. Unfortunately, despite continued and intensive efforts, no cure has yet been found for any of these diseases; reliable markers that allow for an early diagnosis of the disease and the identification of key molecular events leading to disease onset and progression are lacking. Altered adult neurogenesis appears to precede the appearance of severe symptoms. Given the scarcity of human samples and the considerable differences with model species, increasingly complex human stem-cell-based models are being developed. These are shedding light on the molecular alterations that contribute to disease development, facilitating the identification of new clinical targets and providing a screening platform for the testing of candidate drugs. Moreover, the secretome and other promising features of these cell types are being explored, to use them as replacement cells of high plasticity or as co-adjuvant therapy in combinatorial treatments.
Collapse
Affiliation(s)
- Belén Calvo
- Faculty of Health Sciences, Catholic University of Ávila, 05005 Ávila, Spain;
| | - Pierre Schembri-Wismayer
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta;
| | - María Beatriz Durán-Alonso
- Department of Biochemistry and Molecular Biology and Physiology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
| |
Collapse
|
20
|
Takahashi J. iPSC-based cell replacement therapy: from basic research to clinical application. Cytotherapy 2025:S1465-3249(25)00053-2. [PMID: 39969437 DOI: 10.1016/j.jcyt.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
The advancement of induced pluripotent stem cell (iPSC) technology has revolutionized regenerative medicine, enabling breakthroughs in disease modeling, drug discovery, and cell replacement therapies. This review examines the progression of iPSC-based regenerative medicine, focusing on cell replacement therapy and mechanisms like the Replacement Effect, which is crucial for long-term tissue regeneration. Using Parkinson's disease as a key example, it discusses the induction of midbrain dopaminergic neurons from iPSCs and the importance of precise signaling for safety and efficacy. By demonstrating the integration and safety of these cells, animal studies have paved the way for clinical trials. This review highlights the need for strategic collaboration among stakeholders-regulatory authorities, research and medical staff, and industry-to ensure successful clinical applications. iPSC technology's ongoing evolution holds significant promise for broader therapeutic applications and improved patient outcomes.
Collapse
Affiliation(s)
- Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan.
| |
Collapse
|
21
|
Dou D, Lu J, Dou J, Huo Y, Gong X, Zhang X, Chen X. Global regulatory considerations and practices for tumorigenicity evaluation of cell-based therapy. Regul Toxicol Pharmacol 2025; 156:105769. [PMID: 39743127 DOI: 10.1016/j.yrtph.2024.105769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Cell-based therapy, as a "living drug", possesses inherent complexity and heterogeneity. Tumorigenicity evaluation is a crucial aspect of safety assessment for cell-based therapies. Stem cell-based therapies such as hESCs and hiPSCs, may contain residual undifferentiated cells in final product, which have a high potential for proliferation and differentiation, posing a risk of tumor formation in vivo. Additionally, the source, phenotype, differentiation status, proliferative capacity, ex vivo culture conditions, ex vivo processing methods, injection site, and route of administration also influence the tumorigenicity risk of the cells. Tumorigenicity evaluation needs to consider the complexity of design and multifactorial influences. Through the analysis and summary of partial existing marketed and under-development products, combined with practical experience, it is found that there are many differences in requirements and practices related to cell tumorigenicity globally. Regulatory requirements also vary, and guidance and support for applicants' declaration requirements in different regions need to be considered in conjunction with product characteristics and regulatory considerations. This article comprehensively summarizes the requirements of tumorigenicity from main regulatory agencies. Currently, there is no unified global regulatory consensus on technical implementation guide, measures for quantitation or standardization have not been established for evaluation systems. However, based on regulatory requirements and industry practice summaries, through literature research and analysis of tumorigenicity strategy of representative marketed products, the basic focus, and evaluation strategies for tumorigenicity assessment have been preliminarily clarified, providing a reference for the tumorigenicity design of variety of cell-based therapy products.
Collapse
Affiliation(s)
- Dehu Dou
- TriApex Laboratories Co., Ltd. Nanjing, Jiangsu, China; China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Jing Lu
- TriApex Laboratories Co., Ltd. Nanjing, Jiangsu, China
| | - Jinhui Dou
- TriApex Laboratories Co., Ltd. Nanjing, Jiangsu, China
| | - Yan Huo
- TriApex Laboratories Co., Ltd. Nanjing, Jiangsu, China
| | - Xinjiang Gong
- TriApex Laboratories Co., Ltd. Nanjing, Jiangsu, China
| | - Xuefeng Zhang
- TriApex Laboratories Co., Ltd. Nanjing, Jiangsu, China
| | - Xijing Chen
- China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
22
|
Wang L, Yao Y, Xie B, Lei M, Li Y, Shi J, Yu L, Zhou W, Sang Y, Kong L, Liu H, Qiu J. Nanoelectrode-Mediated Extracellular Electrical Stimulation Directing Dopaminergic Neuronal Differentiation of Stem Cells for Improved Parkinson's Disease Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2409745. [PMID: 39703114 DOI: 10.1002/adma.202409745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/18/2024] [Indexed: 12/21/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease caused by the dysfunction and death of dopaminergic neurons. Neural-stem-cell (NSC)-based therapy is a promising approach for the treatment of PD but its therapeutic performance is limited by low efficiency of differentiation of NSCs to dopaminergic neurons. Although electrical stimulation can promote neuronal differentiation, it is not verified whether it can induce the NSCs to specifically differentiate into dopaminergic neurons. Meanwhile, it is a great challenge to precisely apply electrical stimulation to dynamically migrating NSCs after transplantation. Here, electrochemically exfoliated graphene nanosheets are designed to anchor to the membrane of NSCs to serve as wireless nanoelectrodes. After anchoring to the cell membrane, these nanoelectrodes are able to migrate together with the cells and precisely apply extracellular electrical stimulation to the receptors or ion transport channels on the membrane of transplanted cells under alternating magnetic field. The nanoelectrode-mediated electrical stimulation induces 38.46% of the NSCs to specifically differentiate into dopaminergic neurons, while the percentage is only 5.82% for NSCs without the nanoelectrode stimulation. Transplantation of NSCs anchored with the nanoelectrodes effectively improves the recovery of the motor and memory ability of PD mice under alternating magnetic field within 2 weeks.
Collapse
Affiliation(s)
- Liang Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Yuan Yao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Bojun Xie
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Ming Lei
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Yiwei Li
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Jiaming Shi
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Liyang Yu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Wenjuan Zhou
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, China
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
- Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan, Shandong, 250022, P. R. China
| | - Jichuan Qiu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong, 250100, P. R. China
| |
Collapse
|
23
|
Kim TW, Piao J, Bocchi VD, Koo SY, Choi SJ, Chaudhry F, Yang D, Cho HS, Hergenreder E, Perera LR, Joshi S, Mrad ZA, Claros N, Donohue SA, Frank AK, Walsh R, Mosharov EV, Betel D, Tabar V, Studer L. Enhanced yield and subtype identity of hPSC-derived midbrain dopamine neuron by modulation of WNT and FGF18 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631400. [PMID: 39829874 PMCID: PMC11741396 DOI: 10.1101/2025.01.06.631400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
While clinical trials are ongoing using human pluripotent stem cell-derived midbrain dopamine (mDA) neuron precursor grafts in Parkinson's disease (PD), current protocols to derive mDA neurons remain suboptimal. In particular, the yield of TH+ mDA neurons after in vivo grafting and the expression of some mDA neuron and subtype-specific markers can be further improved. For example, characterization of mDA grafts by single cell transcriptomics has yielded only a small proportion of mDA neurons and a considerable fraction of contaminating cell populations. Here we present an optimized mDA neuron differentiation strategy that builds on our clinical grade ("Boost") protocol but includes the addition of FGF18 and IWP2 treatment ("Boost+") at the mDA neurogenesis stage. We demonstrate that Boost+ mDA neurons show higher expression of EN1, PITX3 and ALDH1A1. Improvements in both mDA neurons yield and transcriptional similarity to primary mDA neurons is observed both in vitro and in grafts. Furthermore, grafts are enriched in authentic A9 mDA neurons by single nucSeq. Functional studies in vitro demonstrate increased dopamine production and release and improved electrophysiological properties. In vivo analyses show increased percentages of TH+ mDA neurons resulting in efficient rescue of amphetamine induced rotation behavior in the 6-OHDA rat model and rescue of some motor deficits in non-drug induced assays, including the ladder rung assay that is not improved by Boost mDA neurons. The Boost+ conditions present an optimized protocol with advantages for disease modeling and mDA neuron grafting paradigms.
Collapse
|
24
|
Imani Farahani N, Lin L, Nazir S, Naderi A, Rokos L, McIntosh AR, Julian LM. Advances in physiological and clinical relevance of hiPSC-derived brain models for precision medicine pipelines. Front Cell Neurosci 2025; 18:1478572. [PMID: 39835290 PMCID: PMC11743572 DOI: 10.3389/fncel.2024.1478572] [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: 08/10/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Precision, or personalized, medicine aims to stratify patients based on variable pathogenic signatures to optimize the effectiveness of disease prevention and treatment. This approach is favorable in the context of brain disorders, which are often heterogeneous in their pathophysiological features, patterns of disease progression and treatment response, resulting in limited therapeutic standard-of-care. Here we highlight the transformative role that human induced pluripotent stem cell (hiPSC)-derived neural models are poised to play in advancing precision medicine for brain disorders, particularly emerging innovations that improve the relevance of hiPSC models to human physiology. hiPSCs derived from accessible patient somatic cells can produce various neural cell types and tissues; current efforts to increase the complexity of these models, incorporating region-specific neural tissues and non-neural cell types of the brain microenvironment, are providing increasingly relevant insights into human-specific neurobiology. Continued advances in tissue engineering combined with innovations in genomics, high-throughput screening and imaging strengthen the physiological relevance of hiPSC models and thus their ability to uncover disease mechanisms, therapeutic vulnerabilities, and tissue and fluid-based biomarkers that will have real impact on neurological disease treatment. True physiological understanding, however, necessitates integration of hiPSC-neural models with patient biophysical data, including quantitative neuroimaging representations. We discuss recent innovations in cellular neuroscience that can provide these direct connections through generative AI modeling. Our focus is to highlight the great potential of synergy between these emerging innovations to pave the way for personalized medicine becoming a viable option for patients suffering from neuropathologies, particularly rare epileptic and neurodegenerative disorders.
Collapse
Affiliation(s)
- Negin Imani Farahani
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa Lin
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Shama Nazir
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Alireza Naderi
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Leanne Rokos
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Rotman Research Institute, Baycrest Health Sciences, University of Toronto, Toronto, ON, Canada
| | - Anthony Randal McIntosh
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa M. Julian
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
25
|
Li X, Fang K, Wang F. Somatic cell reprogramming for Parkinson's disease treatment. IBRAIN 2025; 11:59-73. [PMID: 40103698 PMCID: PMC11911114 DOI: 10.1002/ibra.12189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 03/20/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by degeneration of dopamine neurons in the substantia nigra pars compacta. The patient exhibits a series of motor symptoms, such as static tremors, which impair their capacity to take care for themselves in daily life. In the late stage, the patient is unable to walk independently and is bedridden for an extended period of time, reducing their quality of life significantly. So far, treatment methods for PD mainly include drug therapy and deep brain stimulation. Pharmacotherapy is aimed at increasing dopamine (DA) levels; however, the treatment effect is more pronounced in the short term, and there is no benefit in improvement in the overall progression of the disease. In recent years, novel therapeutic strategies have been developed, such as cell reprogramming, trying to generate more DA in PD treatment. This review mainly discusses the advantages, methodology, cell origin, transformation efficiency, and practical application shortcomings of cell reprogramming therapy in PD strategy.
Collapse
Affiliation(s)
- Xiaozhuo Li
- School of Institute of Primate Translational Medicine Kunming University of Science and Technology Kunming China
| | - Kevin Fang
- Living Systems Institute University of Exeter Exeter UK
| | - Fengping Wang
- College of Traditional Chinese Medicine Shandong Second Medical University Weifang Shandong China
| |
Collapse
|
26
|
Jahan I, Harun-Ur-Rashid M, Islam MA, Sharmin F, Al Jaouni SK, Kaki AM, Selim S. Neuronal plasticity and its role in Alzheimer's disease and Parkinson's disease. Neural Regen Res 2024; 21:01300535-990000000-00637. [PMID: 39688547 PMCID: PMC12094540 DOI: 10.4103/nrr.nrr-d-24-01019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/09/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
ABSTRACT Neuronal plasticity, the brain's ability to adapt structurally and functionally, Is essential for learning, memory, and recovery from injuries. In neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, this plasticity is disrupted, leading to cognitive and motor deficits. This review explores the mechanisms of neuronal plasticity and its effect on Alzheimer's disease and Parkinson's disease. Alzheimer's disease features amyloid-beta plaques and tau tangles that impair synaptic function, while Parkinson's disease involves the loss of dopaminergic neurons affecting motor control. Enhancing neuronal plasticity offers therapeutic potential for these diseases. A systematic literature review was conducted using databases such as PubMed, Scopus, and Google Scholar, focusing on studies of neuronal plasticity in Alzheimer's disease and Parkinson's disease. Data synthesis identified key themes such as synaptic mechanisms, neurogenesis, and therapeutic strategies, linking molecular insights to clinical applications. Results highlight that targeting synaptic plasticity mechanisms, such as long-term potentiation and long-term depression, shows promise. Neurotrophic factors, advanced imaging techniques, and molecular tools (e.g., clustered regularly interspaced short palindromic repeats and optogenetics) are crucial in understanding and enhancing plasticity. Current therapies, including dopamine replacement, deep brain stimulation, and lifestyle interventions, demonstrate the potential to alleviate symptoms and improve outcomes. In conclusion, enhancing neuronal plasticity through targeted therapies holds significant promise for treating neurodegenerative diseases. Future research should integrate multidisciplinary approaches to fully harness the therapeutic potential of neuronal plasticity in Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Israt Jahan
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Mohammad Harun-Ur-Rashid
- Department of Chemistry, International University of Business Agriculture and Technology (IUBAT), Sector 10, Uttara Model Town, Dhaka, Bangladesh
| | - Md. Aminul Islam
- Genetic Engineering and Biotechnology Research Laboratory (GEBRL), Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka, Bangladesh
| | - Farhana Sharmin
- Department of Anatomy, Shaheed Suhrawardy Medical College, Dhaka, Bangladesh
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah M. Kaki
- Department of Anesthesia and Pain Medicine, Director of Pain Clinic, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
27
|
Zhang S, Jiang X, Yan M, Cheng Z, Bi J, Wang Q, Luo Y, Tian X. Revealing induced pluripotent stem cells' potential as a better alternative to embryonic stem cells for Parkinson's disease treatment based on single-cell RNA-seq. Braz J Med Biol Res 2024; 57:e13482. [PMID: 39699375 PMCID: PMC11653487 DOI: 10.1590/1414-431x2024e13482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 10/29/2024] [Indexed: 12/20/2024] Open
Abstract
Both embryonic stem cells (ESCs) and the successful reprogramming of induced pluripotent stem cells (iPSCs) offer an unprecedented therapeutic potential for Parkinson's disease (PD), allowing for the replacement of depleted neurons in PD-affected brain regions, thereby achieving therapeutic goals. This study explored the differences in cell types between iPSCs and ESCs in the PD brain to provide a feasible theoretical basis for the improved use of iPSCs as a replacement for ESCs in treating PD. Signal cell RNA sequencing data and microarray data of ESCs and iPSCs were collected from the GEO database. scRNA-seq data were subjected to quality control, clustering, and identification using the Seurat R package to determine cell types and proportions in ESCs and iPSCs. Differential expression analysis was performed to identify differentially expressed genes between ESCs and iPSCs, and PPI network analysis was conducted using String. Based on scRNA-seq data, we identified 13 cell clusters in ESCs and 13 cell clusters in iPSCs. iPSCs were predominantly composed of immune cells and lacked astrocytes, neurons, and dopamine neurons compared to ESCs. iPSCs also exhibited lower cell type diversity compared to ESCs. At the gene level, iPSCs lacked key genes, such as TH and GAP43 for nerve growth and development. At the metabolic level, the difference between ESCs and iPSC was mainly reflected in nerve cells and was closely related to the tumor-proliferation signature. iPSCs can be promoted to differentiate into cell types closer to or even replace ESCs, providing a better therapeutic option for PD treatment.
Collapse
Affiliation(s)
- Sen Zhang
- Shandong Sport University, Jinan, Shandong Province, China
| | - Xing Jiang
- Shandong Sport University, Jinan, Shandong Province, China
| | - Min Yan
- Shandong Sport University, Jinan, Shandong Province, China
| | - Zixiao Cheng
- Gdansk University of Physical Education and Sport, Gdansk, Poland
| | - Jun Bi
- Shandong Sport University, Jinan, Shandong Province, China
| | - Qinglu Wang
- Shandong Sport University, Jinan, Shandong Province, China
| | - Ying Luo
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Xuewen Tian
- Shandong Sport University, Jinan, Shandong Province, China
| |
Collapse
|
28
|
Göksu AY. A review article on the development of dopaminergic neurons and establishment of dopaminergic neuron-based in vitro models by using immortal cell lines or stem cells to study and treat Parkinson's disease. Int J Dev Neurosci 2024; 84:817-842. [PMID: 39379284 DOI: 10.1002/jdn.10383] [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: 11/13/2023] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
The primary pathological hallmark of Parkinson's disease (PD) is the degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta, a critical midbrain region. In vitro models based on DA neurons provide a powerful platform for investigating the cellular and molecular mechanisms of PD and testing novel therapeutic strategies. A deep understanding of DA neuron development, including the signalling pathways and transcription factors involved, is essential for advancing PD research. This article first explores the differentiation and maturation processes of DA neurons in the midbrain, detailing the relevant signalling pathways. It then compares various in vitro models, including primary cells, immortalized cell lines, and stem cell-based models, focusing on the advantages and limitations of each. Special attention is given to the role of immortalized and stem cell models in PD research. This review aims to guide researchers in selecting the most appropriate model for their specific research goals. Ethical considerations and clinical implications of using stem cells in PD research are also discussed.
Collapse
Affiliation(s)
- Azize Yasemin Göksu
- Department of Histology and Embryology, Department of Gene and Cell Therapy, Akdeniz University, School of Medicine, Antalya, Turkey
| |
Collapse
|
29
|
Lei T, Zhang X, Fu G, Luo S, Zhao Z, Deng S, Li C, Cui Z, Cao J, Chen P, Yang H. Advances in human cellular mechanistic understanding and drug discovery of brain organoids for neurodegenerative diseases. Ageing Res Rev 2024; 102:102517. [PMID: 39321879 DOI: 10.1016/j.arr.2024.102517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
The prevalence of neurodegenerative diseases (NDs) is increasing rapidly as the aging population accelerates, and there are still no treatments to halt or reverse the progression of these diseases. While traditional 2D cultures and animal models fail to translate into effective therapies benefit patients, 3D cultured human brain organoids (hBOs) facilitate the use of non-invasive methods to capture patient data. The purpose of this study was to review the research and application of hBO in disease models and drug screening in NDs. The pluripotent stem cells are induced in multiple stages to form cerebral organoids, brain region-specific organoids and their derived brain cells, which exhibit complex brain-like structures and perform electrophysiological activities. The brain region-specific organoids and their derived neurons or glial cells contribute to the understanding of the pathogenesis of NDs and the efficient development of drugs, including Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Glial-rich brain organoids facilitate the study of glial function and neuroinflammation, including astrocytes, microglia, and oligodendrocytes. Further research on the maturation enhancement, vascularization and multi-organoid assembly of hBO will help to enhance the research and application of NDs cellular models.
Collapse
Affiliation(s)
- Tong Lei
- Department of Disease and Syndromes Research, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Xiaoshuang Zhang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Gaoshuang Fu
- Department of Disease and Syndromes Research, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shaohan Luo
- Department of Disease and Syndromes Research, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ziwei Zhao
- Department of Disease and Syndromes Research, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; Hunan Provincial Key Laboratory of Complex Effects Analysis for Chinese Patent Medicine, Yongzhou, Hunan Province 425199, China.
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; Hunan Provincial Key Laboratory of Complex Effects Analysis for Chinese Patent Medicine, Yongzhou, Hunan Province 425199, China.
| |
Collapse
|
30
|
Barker RA, Björklund A, Parmar M. The history and status of dopamine cell therapies for Parkinson's disease. Bioessays 2024; 46:e2400118. [PMID: 39058892 PMCID: PMC11589688 DOI: 10.1002/bies.202400118] [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: 05/23/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
Parkinson's disease (PD) is characterized by the loss of the dopaminergic nigrostriatal pathway which has led to the successful development of drug therapies that replace or stimulate this network pharmacologically. Although these drugs work well in the early stages of the disease, over time they produce side effects along with less consistent clinical benefits to the person with Parkinson's (PwP). As such there has been much interest in repairing this pathway using transplants of dopamine neurons. This work which began 50 years ago this September is still ongoing and has now moved to first in human trials using human pluripotent stem cell-derived dopaminergic neurons. The results of these trials are eagerly awaited although proof of principle data has already come from trials using human fetal midbrain dopamine cell transplants. This data has shown that developing dopamine cells when transplanted in the brain of a PwP can survive long term with clinical benefits lasting decades and with restoration of normal dopaminergic innervation in the grafted striatum. In this article, we discuss the history of this field and how this has now led us to the recent stem cell trials for PwP.
Collapse
Affiliation(s)
- Roger A. Barker
- Department of Clinical Neurosciences and Cambridge Stem Cell InstituteJohn van Geest Centre for Brain RepairUniversity of CambridgeCambridgeUK
| | - Anders Björklund
- Department of Experimental Medical ScienceWallenberg Neuroscience CenterLund UniversityLundSweden
| | - Malin Parmar
- Department of Experimental Medical ScienceWallenberg Neuroscience CenterLund UniversityLundSweden
- Department of Clinical Sciences LundLund Stem Cell Center and Division of NeurologyLund UniversityLundSweden
| |
Collapse
|
31
|
Bamba K, Ozawa M, Daitoku H, Kohara A. Diverting the food-freezing technology improves the cryopreservation efficiency of induced pluripotent stem cells and derived neurospheres. Regen Ther 2024; 27:83-91. [PMID: 38525239 PMCID: PMC10957518 DOI: 10.1016/j.reth.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/19/2024] [Accepted: 03/09/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction Recent advances in induced pluripotent stem (iPS) technology and regenerative medicine require effective cryopreservation of iPSC-derived differentiated cells and three-dimensional cell aggregates (eg. Spheroids and organoids). Moreover, innovative freezing technologies for keeping food fresh over the long-term rapidly developed in the food industry. Therefore, we examined whether one of such freezing technologies, called "Dynamic Effect Powerful Antioxidation Keeping (DEPAK)," could be effective for the cryopreservation of biological materials. Methods We evaluated the efficiency of cryopreservation using DEPAK and Proton freezers, both of which are used in the food industry, compared with conventional slow-freezing methods using a programmable freezer and a cell-freezing vessel. As they are highly susceptible cells to freeze-thaw damage, we selected two suspension cell lines (KHYG-1 derived from human natural killer cell leukemia and THP-1 derived from human acute monocyte leukemia) and two adherent cell lines (OVMANA derived from human ovarian tumors and HuH-7 derived from human hepatocarcinoma). We used two human iPS cell lines, 201B7-Ff and 1231A3, which were either undifferentiated or differentiated into neurospheres. After freezing using the above methods, the frozen cells and neurospheres were immediately transferred to liquid nitrogen. After thawing, we assessed the cryopreservation efficiency of cell viability, proliferation, neurosphere formation, and neurite outgrowth after thawing. Results Among the four cryopreservation methods, DEPAK freezing resulted in the highest cell proliferation in suspension and adherent cell lines. Similar results were obtained for the cryopreservation of undifferentiated human iPS cells. In addition, we demonstrated that the DEPAK freezing method sustained the neurosphere formation capacity of differentiated iPS cells to the same extent as unfrozen controls. In addition, we observed that DEPAK-frozen neurospheres exhibited higher viability after thawing and underwent neural differentiation more efficiently than slow-freezing methods. Conclusions Our results suggest that diversifying food-freezing technologies can overcome the difficulties associated with the cryopreservation of various biological materials, including three-dimensional cell aggregates.
Collapse
Affiliation(s)
- Kenzo Bamba
- JCRB Cell Bank, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Midori Ozawa
- JCRB Cell Bank, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Hiroaki Daitoku
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Arihiro Kohara
- JCRB Cell Bank, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| |
Collapse
|
32
|
Fiorenzano A, Storm P, Sozzi E, Bruzelius A, Corsi S, Kajtez J, Mudannayake J, Nelander J, Mattsson B, Åkerblom M, Björklund T, Björklund A, Parmar M. TARGET-seq: Linking single-cell transcriptomics of human dopaminergic neurons with their target specificity. Proc Natl Acad Sci U S A 2024; 121:e2410331121. [PMID: 39541349 PMCID: PMC11588066 DOI: 10.1073/pnas.2410331121] [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: 05/27/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Dopaminergic (DA) neurons exhibit significant diversity characterized by differences in morphology, anatomical location, axonal projection pattern, and selective vulnerability to disease. More recently, scRNAseq has been used to map DA neuron diversity at the level of gene expression. These studies have revealed a higher than expected molecular diversity in both mouse and human DA neurons. However, whether different molecular expression profiles correlate with specific functions of different DA neurons or with their classical division into mesolimbic (A10) and nigrostriatal (A9) neurons, remains to be determined. To address this, we have developed an approach termed TARGET-seq (Tagging projections by AAV-mediated RetroGrade Enrichment of Transcriptomes) that links the transcriptional profile of the DA neurons with their innervation of specific target structures in the forebrain. Leveraging this technology, we identify molecularly distinct subclusters of human DA neurons with a clear link between transcriptome and axonal target-specificity, offering the possibility to infer neuroanatomical-based classification to molecular identity and target-specific connectivity. We subsequently used this dataset to identify candidate transcription factors along DA developmental trajectories that may control subtype identity, thus providing broad avenues that can be further explored in the design of next-generation A9 and A10 enriched DA-neurons for drug screening or A9 enriched DA cells for clinical stem cell-based therapies.
Collapse
Affiliation(s)
- Alessandro Fiorenzano
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Petter Storm
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Edoardo Sozzi
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Andreas Bruzelius
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Sara Corsi
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Janko Kajtez
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Janitha Mudannayake
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Jenny Nelander
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Bengt Mattsson
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Malin Åkerblom
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund Skåne223 62, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund Skåne223 62, Sweden
| | - Anders Björklund
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Skåne223 62, Sweden
| |
Collapse
|
33
|
Matsuo-Takasaki M, Kambayashi S, Hemmi Y, Wakabayashi T, Shimizu T, An Y, Ito H, Takeuchi K, Ibuki M, Kawashima T, Masayasu R, Suzuki M, Kawai Y, Umekage M, Kato TM, Noguchi M, Nakade K, Nakamura Y, Nakaishi T, Nishishita N, Tsukahara M, Hayashi Y. Complete suspension culture of human induced pluripotent stem cells supplemented with suppressors of spontaneous differentiation. eLife 2024; 12:RP89724. [PMID: 39529479 PMCID: PMC11556790 DOI: 10.7554/elife.89724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are promising resources for producing various types of tissues in regenerative medicine; however, the improvement in a scalable culture system that can precisely control the cellular status of hiPSCs is needed. Utilizing suspension culture without microcarriers or special materials allows for massive production, automation, cost-effectiveness, and safety assurance in industrialized regenerative medicine. Here, we found that hiPSCs cultured in suspension conditions with continuous agitation without microcarriers or extracellular matrix components were more prone to spontaneous differentiation than those cultured in conventional adherent conditions. Adding PKCβ and Wnt signaling pathway inhibitors in the suspension conditions suppressed the spontaneous differentiation of hiPSCs into ectoderm and mesendoderm, respectively. In these conditions, we successfully completed the culture processes of hiPSCs, including the generation of hiPSCs from peripheral blood mononuclear cells with the expansion of bulk population and single-cell sorted clones, long-term culture with robust self-renewal characteristics, single-cell cloning, direct cryopreservation from suspension culture and their successful recovery, and efficient mass production of a clinical-grade hiPSC line. Our results demonstrate that precise control of the cellular status in suspension culture conditions paves the way for their stable and automated clinical application.
Collapse
Affiliation(s)
- Mami Matsuo-Takasaki
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Sho Kambayashi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Yasuko Hemmi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Tamami Wakabayashi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Tomoya Shimizu
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Yuri An
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Hidenori Ito
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
| | - Kazuhiro Takeuchi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Masato Ibuki
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Terasu Kawashima
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Rio Masayasu
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Manami Suzuki
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Yoshikazu Kawai
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | | | - Tomoaki M Kato
- Research and Development Center, CiRA FoundationKyotoJapan
| | - Michiya Noguchi
- Cell Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Koji Nakade
- Gene Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research CenterIbarakiJapan
| | - Tomoyuki Nakaishi
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | - Naoki Nishishita
- Regenerative Medicine and Cell Therapy Laboratories, KANEKA CORPORATIONKobeJapan
| | | | - Yohei Hayashi
- iPS Cell Advanced Characterization and Development Team, RIKEN BioResource Research CenterIbarakiJapan
- Faculty of Medicine and School of Integrative and Global Majors, University of TsukubaIbarakiJapan
| |
Collapse
|
34
|
Xue J, Wu D, Bao Y, Wu Y, Zhang X, Chen L. The Abnormal Proliferation of Midbrain Dopamine Cells From Human Pluripotent Stem Cells Is Induced by Exposure to the Tumor Microenvironment. CNS Neurosci Ther 2024; 30:e70117. [PMID: 39563017 PMCID: PMC11576488 DOI: 10.1111/cns.70117] [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: 02/03/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
AIMS Tumorigenicity is a significant concern in stem cell-based therapies. However, traditional tumorigenicity tests using animal models often produce inaccurate results. Consequently, a more sensitive method for assessing tumorigenicity is required. This study aimed to enhance sensitivity by exposing functional progenitors derived from human pluripotent stem cells (hPSCs) to the tumor microenvironment (TME) in vitro before transplantation, potentially making them more prone to abnormal proliferation or tumorigenicity. METHODS Midbrain dopamine (mDA) cells derived from hPSCs were exposed to the TME by coculturing with medulloblastoma. The cellular characteristics of these cocultured mDA cells were evaluated both in vitro and in vivo, and the mechanisms underlying the observed alterations were investigated. RESULTS Our findings demonstrated increased proliferation of cocultured mDA cells both in vitro and in vivo. Moreover, these proliferating cells showed a higher expression of Ki67 and SOX1, suggesting abnormal proliferation. The observed abnormal proliferation in cocultured mDA cells was attributed to the hyperactivation of proliferation-related genes, the JAK/STAT3 pathway, and cytokine stimulation. CONCLUSION This study indicates that exposing functional progenitors to the TME in vitro before transplantation can induce abnormal proliferation, thereby increasing the sensitivity of tumorigenicity tests.
Collapse
Affiliation(s)
- Jun Xue
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Dongyan Wu
- Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Yuting Bao
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Yifan Wu
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Xin Zhang
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
35
|
Luciani M, Garsia C, Beretta S, Cifola I, Peano C, Merelli I, Petiti L, Miccio A, Meneghini V, Gritti A. Human iPSC-derived neural stem cells displaying radial glia signature exhibit long-term safety in mice. Nat Commun 2024; 15:9433. [PMID: 39487141 PMCID: PMC11530573 DOI: 10.1038/s41467-024-53613-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NSCs) hold promise for treating neurodegenerative and demyelinating disorders. However, comprehensive studies on their identity and safety remain limited. In this study, we demonstrate that hiPSC-NSCs adopt a radial glia-associated signature, sharing key epigenetic and transcriptional characteristics with human fetal neural stem cells (hfNSCs) while exhibiting divergent profiles from glioblastoma stem cells. Long-term transplantation studies in mice showed robust and stable engraftment of hiPSC-NSCs, with predominant differentiation into glial cells and no evidence of tumor formation. Additionally, we identified the Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) as a regulator of astroglial differentiation in hiPSC-NSCs. These findings provide valuable transcriptional and epigenetic reference datasets to prospectively define the maturation stage of NSCs derived from different hiPSC sources and demonstrate the long-term safety of hiPSC-NSCs, reinforcing their potential as a viable alternative to hfNSCs for clinical applications.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Garsia
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Beretta
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Ingrid Cifola
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, UoS of Milan, National Research Council, Rozzano, Milan, Italy
- Human Technopole, Via Rita Levi Montalcini 1, Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Luca Petiti
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), via F.lli Cervi 93, 20054 Segrate, Milan, Italy
| | - Annarita Miccio
- IMAGINE Institute, Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
36
|
Yang B, Hu S, Jiang Y, Xu L, Shu S, Zhang H. Advancements in Single-Cell RNA Sequencing Research for Neurological Diseases. Mol Neurobiol 2024; 61:8797-8819. [PMID: 38564138 DOI: 10.1007/s12035-024-04126-3] [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: 11/29/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Neurological diseases are a major cause of the global burden of disease. Although the mechanisms of the occurrence and development of neurological diseases are not fully clear, most of them are associated with cells mediating neuroinflammation. Yet medications and other therapeutic options to improve treatment are still very limited. Single-cell RNA sequencing (scRNA-seq), as a delightfully potent breakthrough technology, not only identifies various cell types and response states but also uncovers cell-specific gene expression changes, gene regulatory networks, intercellular communication, and cellular movement trajectories, among others, in different cell types. In this review, we describe the technology of scRNA-seq in detail and discuss and summarize the application of scRNA-seq in exploring neurological diseases, elaborating the corresponding specific mechanisms of the diseases as well as providing a reliable basis for new therapeutic approaches. Finally, we affirm that scRNA-seq promotes the development of the neuroscience field and enables us to have a deeper cellular understanding of neurological diseases in the future, which provides strong support for the treatment of neurological diseases and the improvement of patients' prognosis.
Collapse
Affiliation(s)
- Bingjie Yang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuqi Hu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yiru Jiang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lei Xu
- Department of Neurology, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Song Shu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Hao Zhang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
37
|
Dong W, Liu S, Li S, Wang Z. Cell reprogramming therapy for Parkinson's disease. Neural Regen Res 2024; 19:2444-2455. [PMID: 38526281 PMCID: PMC11090434 DOI: 10.4103/1673-5374.390965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/23/2023] [Accepted: 10/08/2023] [Indexed: 03/26/2024] Open
Abstract
Parkinson's disease is typically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Many studies have been performed based on the supplementation of lost dopaminergic neurons to treat Parkinson's disease. The initial strategy for cell replacement therapy used human fetal ventral midbrain and human embryonic stem cells to treat Parkinson's disease, which could substantially alleviate the symptoms of Parkinson's disease in clinical practice. However, ethical issues and tumor formation were limitations of its clinical application. Induced pluripotent stem cells can be acquired without sacrificing human embryos, which eliminates the huge ethical barriers of human stem cell therapy. Another widely considered neuronal regeneration strategy is to directly reprogram fibroblasts and astrocytes into neurons, without the need for intermediate proliferation states, thus avoiding issues of immune rejection and tumor formation. Both induced pluripotent stem cells and direct reprogramming of lineage cells have shown promising results in the treatment of Parkinson's disease. However, there are also ethical concerns and the risk of tumor formation that need to be addressed. This review highlights the current application status of cell reprogramming in the treatment of Parkinson's disease, focusing on the use of induced pluripotent stem cells in cell replacement therapy, including preclinical animal models and progress in clinical research. The review also discusses the advancements in direct reprogramming of lineage cells in the treatment of Parkinson's disease, as well as the controversy surrounding in vivo reprogramming. These findings suggest that cell reprogramming may hold great promise as a potential strategy for treating Parkinson's disease.
Collapse
Affiliation(s)
- Wenjing Dong
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shuyi Liu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| |
Collapse
|
38
|
Xue J, Chu Y, Huang Y, Chen M, Sun M, Fan Z, Wu Y, Chen L. A tumorigenicity evaluation platform for cell therapies based on brain organoids. Transl Neurodegener 2024; 13:53. [PMID: 39472972 PMCID: PMC11520457 DOI: 10.1186/s40035-024-00446-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Tumorigenicity represents a critical challenge in stem cell-based therapies requiring rigorous monitoring. Conventional approaches for tumorigenicity evaluation are based on animal models and have numerous limitations. Brain organoids, which recapitulate the structural and functional complexity of the human brain, have been widely used in neuroscience research. However, the capacity of brain organoids for tumorigenicity evaluation needs to be further elucidated. METHODS A cerebral organoid model produced from human pluripotent stem cells (hPSCs) was employed. Meanwhile, to enhance the detection sensitivity for potential tumorigenic cells, we created a glioblastoma-like organoid (GBM organoid) model from TP53-/-/PTEN-/- hPSCs to provide a tumor microenvironment for injected cells. Midbrain dopamine (mDA) cells from human embryonic stem cells were utilized as a cell therapy product. mDA cells, hPSCs, mDA cells spiked with hPSCs, and immature mDA cells were then injected into the brain organoids and NOD SCID mice. The injected cells within the brain organoids were characterized, and compared with those injected in vivo to evaluate the capability of the brain organoids for tumorigenicity evaluation. Single-cell RNA sequencing was performed to identify the differential gene expression between the cerebral organoids and the GBM organoids. RESULTS Both cerebral organoids and GBM organoids supported maturation of the injected mDA cells. The hPSCs and immature mDA cells injected in the GBM organoids showed a significantly higher proliferative capacity than those injected in the cerebral organoids and in NOD SCID mice. Furthermore, the spiked hPSCs were detectable in both the cerebral organoids and the GBM organoids. Notably, the GBM organoids demonstrated a superior capacity to enhance proliferation and pluripotency of spiked hPSCs compared to the cerebral organoids and the mouse model. Kyoto Encyclopedia of Genes and Genomes analysis revealed upregulation of tumor-related metabolic pathways and cytokines in the GBM organoids, suggesting that these factors underlie the high detection sensitivity for tumorigenicity evaluation. CONCLUSIONS Our findings suggest that brain organoids could represent a novel and effective platform for evaluating the tumorigenic risk in stem cell-based therapies. Notably, the GBM organoids offer a superior platform that could complement or potentially replace traditional animal-based models for tumorigenicity evaluation.
Collapse
Affiliation(s)
- Jun Xue
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yanwang Huang
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ming Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Meng Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Zhiqin Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China.
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
39
|
Storm P, Zhang Y, Nilsson F, Fiorenzano A, Krausse N, Åkerblom M, Davidsson M, Yuan J, Kirkeby A, Björklund T, Parmar M. Lineage tracing of stem cell-derived dopamine grafts in a Parkinson's model reveals shared origin of all graft-derived cells. SCIENCE ADVANCES 2024; 10:eadn3057. [PMID: 39423273 PMCID: PMC11488568 DOI: 10.1126/sciadv.adn3057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Stem cell therapies for Parkinson's disease are at an exciting time of development, and several clinical trials have recently been initiated. Human pluripotent stem cells are differentiated into transplantable dopamine (DA) progenitors which are proliferative at the time of grafting and undergo terminal differentiation and maturation in vivo. While the progenitors are homogeneous at the time of transplantation, they give rise to heterogeneous grafts composed not only of therapeutic DA neurons but also of other mature cell types. The mechanisms for graft diversification are unclear. We used single-nucleus RNA-seq and ATAC-seq to profile DA progenitors before transplantation combined with molecular barcode-based tracing to determine origin and shared lineages of the mature cell types in the grafts. Our data demonstrate that astrocytes, vascular leptomeningeal cells, and DA neurons are the main component of the DAergic grafts, originating from a common progenitor that is tripotent at the time of transplantation.
Collapse
Affiliation(s)
- Petter Storm
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yu Zhang
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Wallenberg Center for Molecular Medicine (WCMM) and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Fredrik Nilsson
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Alessandro Fiorenzano
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Niklas Krausse
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Malin Åkerblom
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Marcus Davidsson
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joan Yuan
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Wallenberg Center for Molecular Medicine (WCMM) and Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
40
|
Svendsen SP, Svendsen CN. Cell therapy for neurological disorders. Nat Med 2024; 30:2756-2770. [PMID: 39407034 DOI: 10.1038/s41591-024-03281-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/30/2024] [Indexed: 10/18/2024]
Abstract
Cell therapies for neurological disorders are entering the clinic and present unique challenges and opportunities compared with conventional medicines. They have the potential to replace damaged nervous tissue and integrate into the brain or spinal cord to produce functional effects for the lifetime of the patient, which could revolutionize the way clinicians treat debilitating neurological disorders. The major challenge has been cell sourcing, which historically relied mainly on fetal brain tissue. This has largely been overcome with the advent of pluripotent stem cell technology and the ability to make almost any cell of the nervous system at scale. Furthermore, advances in gene editing now allow the generation of genetically modified cells that could perform better and evade the immune system. With all the remarkable new approaches to treat neurological disorders, we take a critical look at the state of current clinical trials and how challenges may be overcome with the evolving technology and innovation occurring in the stem cell field.
Collapse
Affiliation(s)
- Soshana P Svendsen
- Cedars-Sinai Board of Governors Regenerative Medicine Institute, Los Angeles, CA, USA
| | - Clive N Svendsen
- Cedars-Sinai Board of Governors Regenerative Medicine Institute, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Zhang Y, Zhu Z, Li Z, Feng J, Long J, Deng Y, Ahmed W, Khan AA, Huang S, Fu Q, Chen L. Sbno1 mediates cell-cell communication between neural stem cells and microglia through small extracellular vesicles. Cell Biosci 2024; 14:125. [PMID: 39343943 PMCID: PMC11441009 DOI: 10.1186/s13578-024-01296-4] [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: 12/04/2023] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Neural stem cells (NSCs) play a crucial role in the progress of ischemic stroke. Research on zebrafish embryonic demonstrates an association between Strawberry Notch 1 (Sbno1) and central nervous system development. However, the regulation and underlying mechanism of Sbno1 in NSCs have not been studied yet. Here, we investigated the role and the mechanism of Sbno1 in NSCs development and the potential therapeutic value of Sbno1 in ischemic stroke. METHODS Adeno-associated virus (AAV) was used for overexpression or knockdown of Sbno1 in vitro or in vivo. A mouse model of MCAO was established to evaluate the neuroprotective effects of AAV-Sbno1, including balance beam test, rotarod test, and strength evaluation. H&E and immunofluorescence assessed neuronal impairment. Western blot and RT-qPCR were used to detect the expression of Sbno1 and its downstream target genes. RNA-seq and western blot were performed to explore further molecular mechanisms by which Sbno1 promoted endogenous repair of NSCs and macrophages M2 polarization. CCK8 was conducted to assess the effects of Sbno1 on NSCs proliferation. The impact of Sbno1 on NSCs apoptosis was evaluated by flow cytometry. NSCs derived from small extracellular vesicles (sEV) were obtained using ultracentrifugation and identified through nanoparticle tracking analysis (NTA) and western blot analysis. RESULTS Our results showed that Sbno1 is highly expressed in the central nervous system, which plays a crucial role in regulating the proliferation of NSCs through the PI3k-Akt-GSK3β-Wnt/β-catenin signaling pathway. In addition, with overexpression of Sbno1 in the hippocampus, post-stroke behavioral scores were superior to the wild-type mice, and immunofluorescence staining revealed an increased number of newly generated neurons. sEV released by NSCs overexpressing Sbno1 inhibited neuroinflammation, which mechanistically impaired the activation of the microglial NF-κB and MAPK signaling pathways. CONCLUSIONS Our studies indicate that sbno1 promotes the proliferation of NSCs and enhances endogenous repairing through the PI3k-Akt-GSK3β-Wnt/β-catenin signaling pathway. Additionally, NSCs overexpressing sbno1 improve ischemic stroke recovery and inhibit neuroinflammation after ischemia by sEV through the MAPK and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Zhihan Zhu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Zhinuo Li
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Jia Feng
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Jun Long
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Yushu Deng
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Waqas Ahmed
- Department of Neurology, Zhongda Hospital Southeast University, Nanjing, China
| | - Ahsan Ali Khan
- Department of Neurosurgery, The Aga Khan University, Karachi, Pakistan
| | - Shiying Huang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Qingling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
42
|
Wang CH, Lin GC, Fu RH, Huang YC, Chen SY, Lin SZ, Harn HJ, Shyu WC, Huang YF, Jeng LB, Liu SP. Neural stem cells derived from α-synuclein-knockdown iPS cells alleviate Parkinson's disease. Cell Death Discov 2024; 10:407. [PMID: 39285205 PMCID: PMC11405526 DOI: 10.1038/s41420-024-02176-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
Stem cells have the potential to replace damaged or defective cells and assist in the development of treatments for neurodegenerative diseases, including Parkinson's disease (PD) and Alzheimer's disease. iPS cells derived from patient-specific somatic cells are not only ethically acceptable, but they also avoid complications relating to immune rejection. Currently, researchers are developing stem cell-based therapies for PD using induced pluripotent stem (iPS) cells. iPS cells can differentiate into cells from any of the three germ layers, including neural stem cells (NSCs). Transplantation of neural stem cells (NSCs) is an emerging therapy for treating neurological disorders by restoring neuronal function. Nevertheless, there are still challenges associated with the quality and source of neural stem cells. This issue can be addressed by genetically edited iPS cells. In this study, shRNA was used to knock down the expression of mutant α-synuclein (SNCA) in iPS cells that were generated from SNCA A53T transgenic mice, and these iPS cells were differentiated to NSCs. After injecting these NSCs into SNCA A53T mice, the therapeutic effects of these cells were evaluated. We found that the transplantation of neural stem cells produced from SNCA A53T iPS cells with knocking down SNCA not only improved SNCA A53T mice coordination abilities, balance abilities, and locomotor activities but also significantly prolonged their lifespans. The results of this study suggest an innovative therapeutic approach that combines stem cell therapy and gene therapy for the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Chie-Hong Wang
- Cell Therapy Center, China Medical University Hospital, Taichung, 404, Taiwan
- Neuroscience and Brain Disease Center, College of Medicine, China Medical University, Taichung, 411, Taiwan
- Department of Neurology, China Medical University Hospital, Taichung, 404, Taiwan
| | - Guan-Cyun Lin
- Graduate Institute of Biomedical Science, China Medical University, Taichung, 411, Taiwan
| | - Ru-Huei Fu
- Ph.D. Program for Aging, College of Medicine, China Medical University, Taichung, 411, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung, 404, Taiwan
| | - Yu-Chuen Huang
- Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung, 404, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 411, Taiwan
| | - Shih-Yin Chen
- Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung, 404, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 411, Taiwan
| | - Shinn-Zong Lin
- Buddhist Tzu Chi Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, 970, Taiwan
| | - Horng-Jyh Harn
- Buddhist Tzu Chi Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
- Department of Pathology, Hualien Tzu Chi Hospital and Tzu Chi University, Hualien, 970, Taiwan
| | - Woei-Cherng Shyu
- Neuroscience and Brain Disease Center, College of Medicine, China Medical University, Taichung, 411, Taiwan
- Department of Neurology, China Medical University Hospital, Taichung, 404, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung, 404, Taiwan
| | - Yi-Fang Huang
- Department of General Dentistry, Linkou Chang Gung Memorial Hospital, Taoyuan City, 333, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Graduate Institute of Dental and Craniofacial Science, College of Medicine, Chang Gung University, Taoyuan City, 333, Taiwan
| | - Long-Bin Jeng
- Cell Therapy Center, China Medical University Hospital, Taichung, 404, Taiwan
- Organ Transplantation Center, China Medical University Hospital, Taichung, 404, Taiwan
| | - Shih-Ping Liu
- Neuroscience and Brain Disease Center, College of Medicine, China Medical University, Taichung, 411, Taiwan.
- Ph.D. Program for Aging, College of Medicine, China Medical University, Taichung, 411, Taiwan.
- Translational Medicine Research Center, China Medical University Hospital, Taichung, 404, Taiwan.
| |
Collapse
|
43
|
Galgani A, Scotto M, Giorgi FS. The Neuroanatomy of Induced Pluripotent Stem Cells: In Vitro Models of Subcortical Nuclei in Neurodegenerative Disorders. Curr Issues Mol Biol 2024; 46:10180-10199. [PMID: 39329959 PMCID: PMC11430477 DOI: 10.3390/cimb46090607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Neuromodulatory subcortical systems (NSSs) are monoaminergic and cholinergic neuronal groups that are markedly and precociously involved in the pathogenesis of many neurodegenerative disorders (NDDs), including Parkinson's and Alzheimer's diseases. In humans, although many tools have been developed to infer information on these nuclei, encompassing neuroimaging and neurophysiological methods, a detailed and specific direct evaluation of their cellular features in vivo has been difficult to obtain until recent years. The development of induced pluripotent stem cell (iPSC) models has allowed research to deeply delve into the cellular and molecular biology of NSS neurons. In fact, iPSCs can be produced easily and non-invasively from patients' fibroblasts or circulating blood monocytes, by de-differentiating those cells using specific protocols, and then be re-differentiated towards neural phenotypes, which may reproduce the specific features of the correspondent brain neurons (including NSS ones) from the same patient. In this review, we summarized findings obtained in the field of NDDs using iPSCs, with the aim to understand how reliably these might represent in vitro models of NSS. We found that most of the current literature in the field of iPSCs and NSSs in NDDs has focused on midbrain dopaminergic neurons in Parkinson's disease, providing interesting results on cellular pathophysiology and even leading to the first human autologous transplantation. Differentiation protocols for noradrenergic, cholinergic, and serotoninergic neurons have also been recently defined and published. Thus, it might be expected that in the near future, this approach could extend to other NSSs and other NDDs.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Marco Scotto
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Filippo S. Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
- IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| |
Collapse
|
44
|
Narasimhan K, Hakami A, Comini G, Patton T, Newland B, Dowd E. Cryogel microcarriers loaded with glial cell line-derived neurotrophic factor enhance the engraftment of primary dopaminergic neurons in a rat model of Parkinson's disease. J Neural Eng 2024; 21:056011. [PMID: 39231475 DOI: 10.1088/1741-2552/ad7761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
Objective.Cryogel microcarriers made of poly(ethylene glycol) diacrylate and 3-sulfopropyl acrylate have the potential to act as delivery vehicles for long-term retention of neurotrophic factors (NTFs) in the brain. In addition, they can potentially enhance stem cell-derived dopaminergic (DAergic) cell replacement strategies for Parkinson's disease (PD), by addressing the limitations of variable survival and poor differentiation of the transplanted precursors due to neurotrophic deprivation post-transplantation in the brain. In this context, to develop a proof-of-concept, the aim of this study was to determine the efficacy of glial cell line-derived NTF (GDNF)-loaded cryogel microcarriers by assessing their impact on the survival of, and reinnervation by, primary DAergic grafts after intra-striatal delivery in Parkinsonian rat brains.Approach.Rat embryonic day 14 ventral midbrain cells were transplanted into the 6-hydroxydopamine-lesioned striatum either alone, or with GDNF, or with unloaded cryogel microcarriers, or with GDNF-loaded cryogel microcarriers.Post-mortem, GDNF and tyrosine hydroxylase immunostaining were used to identify retention of the delivered GDNF within the implanted cryogel microcarriers, and to identify the transplanted DAergic neuronal cell bodies and fibres in the brains, respectively.Main results.We found an intact presence of GDNF-stained cryogel microcarriers in graft sites, indicating their ability for long-term retention of the delivered GDNF up to 4 weeks in the brain. This resulted in an enhanced survival (1.9-fold) of, and striatal reinnervation (density & volume) by, the grafted DAergic neurons, in addition to an enhanced sprouting of fibres within graft sites.Significance.This data provides an important proof-of-principle for the beneficial effects of neurotrophin-loaded cryogel microcarriers on engraftment of cells in the context of cell replacement therapy in PD. For clinical translation, further studies will be needed to assess the impact of cryogel microcarriers on the survival and differentiation of stem cell-derived DAergic precursors in Parkinsonian rat brains.
Collapse
Affiliation(s)
- Kaushik Narasimhan
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Abrar Hakami
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, United Kingdom
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Giulia Comini
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Tommy Patton
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, United Kingdom
- Leibniz-Institut für Polymerforschung Dresden e.V., 01069 Dresden, Germany
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, University of Galway, Galway, Ireland
| |
Collapse
|
45
|
Nakashima Y, Tsukahara M. Atelocollagen supports three-dimensional culture of human induced pluripotent stem cells. Mol Ther Methods Clin Dev 2024; 32:101302. [PMID: 39185274 PMCID: PMC11342089 DOI: 10.1016/j.omtm.2024.101302] [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: 01/12/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
As autologous induced pluripotent stem cell (iPSC) therapy requires a custom-made small-lot cell production line, and the cell production method differs significantly from the existing processes for producing allogeneic iPSC stocks for clinical use. Specifically, mass culture to produce stock is no longer necessary; instead, a series of operations from iPSC production to induction of differentiation of therapeutic cells must be performed continuously. A three-dimensional (3D) culture method using small, closed-cell manufacturing devices is suitable for autologous iPSC therapy. The use of such devices avoids the need to handle many patient-derived specimens in a single clean room; handling of cell cultures in an open system in a cell processing facility increases the risk of infection. In this study, atelocollagen beads were evaluated as a 3D biomaterial to assist 3D culture in the establishment, expansion culture, and induction of differentiation of iPSCs. It was found that iPSCs can be handled in a closed-cell device with the same ease as use of a two-dimensional (2D) culture when laminin-511 is added to the medium. In conclusion, atelocollagen beads enable 3D culture of iPSCs, and the quality of the obtained cells is at the same level as those derived from 2D culture.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- CiRA Foundation, Research and Development Center, Nakanoshima Qross, Osaka 530-005, Japan
| | - Masayoshi Tsukahara
- CiRA Foundation, Research and Development Center, Nakanoshima Qross, Osaka 530-005, Japan
| |
Collapse
|
46
|
Clark BJ, Lelos MJ, Loring JF. Advancing Parkinson's disease treatment: cell replacement therapy with neurons derived from pluripotent stem cells. Stem Cells 2024; 42:781-790. [PMID: 38902932 DOI: 10.1093/stmcls/sxae042] [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: 03/11/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024]
Abstract
The motor symptoms of Parkinson's disease (PD) are caused by the progressive loss of dopamine neurons from the substantia nigra. There are currently no treatments that can slow or reverse the neurodegeneration. To restore the lost neurons, international groups have initiated clinical trials using human embryonic or induced pluripotent stem cells (PSCs) to derive dopamine neuron precursors that are used as transplants to replace the lost neurons. Proof-of-principle experiments in the 1980s and 1990s showed that grafts of fetal ventral mesencephalon, which contains the precursors of the substantial nigra, could, under rare circumstances, reverse symptoms of the disease. Improvements in PSC technology and genomics have inspired researchers to design clinical trials using PSC-derived dopamine neuron precursors as cell replacement therapy for PD. We focus here on 4 such first-in-human clinical trials that have begun in the US, Europe, and Japan. We provide an overview of the sources of PSCs and the methods used to generate cells for transplantation. We discuss pros and cons of strategies for allogeneic, immune-matched, and autologous approaches and novel methods for overcoming rejection by the immune system. We consider challenges for safety and efficacy of the cells for durable engraftment, focusing on the genomics-based quality control methods to assure that the cells will not become cancerous. Finally, since clinical trials like these have never been undertaken before, we comment on the value of cooperation among rivals to contribute to advancements that will finally provide relief for the millions suffering from the symptoms of PD.
Collapse
Affiliation(s)
- Branden J Clark
- Department of Biomedical Engineering, UC Irvine, Irvine, CA 92697, United States
| | - Mariah J Lelos
- School of Biosciences, Museum Avenue, Cardiff University, Cardiff, CF10 3AX, United Kingdom
| | - Jeanne F Loring
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92030, United States
| |
Collapse
|
47
|
Zhao Y, Liu K, Wang Y, Ma Y, Guo W, Shi C. Human-mouse chimeric brain models constructed from iPSC-derived brain cells: Applications and challenges. Exp Neurol 2024; 379:114848. [PMID: 38857749 DOI: 10.1016/j.expneurol.2024.114848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The establishment of reliable human brain models is pivotal for elucidating specific disease mechanisms and facilitating the discovery of novel therapeutic strategies for human brain disorders. Human induced pluripotent stem cell (iPSC) exhibit remarkable self-renewal capabilities and can differentiate into specialized cell types. This makes them a valuable cell source for xenogeneic or allogeneic transplantation. Human-mouse chimeric brain models constructed from iPSC-derived brain cells have emerged as valuable tools for modeling human brain diseases and exploring potential therapeutic strategies for brain disorders. Moreover, the integration and functionality of grafted stem cells has been effectively assessed using these models. Therefore, this review provides a comprehensive overview of recent progress in differentiating human iPSC into various highly specialized types of brain cells. This review evaluates the characteristics and functions of the human-mouse chimeric brain model. We highlight its potential roles in brain function and its ability to reconstruct neural circuitry in vivo. Additionally, we elucidate factors that influence the integration and differentiation of human iPSC-derived brain cells in vivo. This review further sought to provide suitable research models for cell transplantation therapy. These research models provide new insights into neuropsychiatric disorders, infectious diseases, and brain injuries, thereby advancing related clinical and academic research.
Collapse
Affiliation(s)
- Ya Zhao
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Ke Liu
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Yinghua Wang
- Medical College of Yan'an University, Yan'an 716000, PR China
| | - Yifan Ma
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; Gansu University of traditional Chinese medicine, Lanzhou 730030, PR China
| | - Wenwen Guo
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China
| | - Changhong Shi
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| |
Collapse
|
48
|
Azevedo EM, Fracaro L, Hochuli AHD, Ilkiw J, Bail EL, Lisboa MDO, Rodrigues LS, Barchiki F, Correa A, Capriglione LGA, Brofman PRS, Lima MMS. Comparative analysis of uninduced and neuronally-induced human dental pulp stromal cells in a 6-OHDA model of Parkinson's disease. Cytotherapy 2024; 26:1052-1061. [PMID: 38739074 DOI: 10.1016/j.jcyt.2024.04.068] [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: 12/13/2023] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND In recent years, dental pulp stromal cells (DPSCs) have emerged as a promising therapeutic approach for Parkinson's disease (PD), owing to their inherent neurogenic potential and the lack of neuroprotective treatments for this condition. However, uncertainties persist regarding the efficacy of these cells in an undifferentiated state versus a neuronally-induced state. This study aims to delineate the distinct therapeutic potential of uninduced and neuronally-induced DPSCs in a rodent model of PD induced by 6-Hydroxydopamine (6-OHDA). METHODS DPSCs were isolated from human teeth, characterized as mesenchymal stromal cells, and induced to neuronal differentiation. Neuronal markers were assessed before and after induction. DPSCs were transplanted into the substantia nigra pars compacta (SNpc) of rats 7 days following the 6-OHDA lesion. In vivo tracking of the cells, evaluation of locomotor behavior, dopaminergic neuron survival, and the expression of essential proteins within the dopaminergic system were conducted 7 days postgrafting. RESULTS Isolated DPSCs exhibited typical characteristics of mesenchymal stromal cells and maintained a normal karyotype. DPSCs consistently expressed neuronal markers, exhibiting elevated expression of βIII-tubulin following neuronal induction. Results from the animal model showed that both DPSC types promoted substantial recovery in dopaminergic neurons, correlating with enhanced locomotion. Additionally, neuronally-induced DPSCs prevented GFAP elevation, while altering DARPP-32 phosphorylation states. Conversely, uninduced DPSCs reduced JUN levels. Both DPSC types mitigated the elevation of glycosylated DAT. CONCLUSIONS Our results suggested that uninduced DPSCs and neuronally-induced DPSCs exhibit potential in reducing dopaminergic neuron loss and improving locomotor behavior, but their underlying mechanisms differ.
Collapse
Affiliation(s)
- Evellyn M Azevedo
- Physiology Department, Parkinson's Disease and Sleep Neurophysiology Lab, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | - Letícia Fracaro
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Agner H D Hochuli
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Jéssica Ilkiw
- Physiology Department, Parkinson's Disease and Sleep Neurophysiology Lab, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | - Ellen L Bail
- Physiology Department, Parkinson's Disease and Sleep Neurophysiology Lab, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | - Mateus de O Lisboa
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Lais S Rodrigues
- Physiology Department, Parkinson's Disease and Sleep Neurophysiology Lab, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | - Fabiane Barchiki
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Alejandro Correa
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute, Fiocruz-Paraná, Curitiba, Brazil
| | - Luiz G A Capriglione
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Paulo R S Brofman
- Core for Cell Technology, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Brazil
| | - Marcelo M S Lima
- Physiology Department, Parkinson's Disease and Sleep Neurophysiology Lab, Universidade Federal do Paraná (UFPR), Curitiba, Brazil.
| |
Collapse
|
49
|
Prudon N, Cordero-Espinoza L, Abarkan M, Gurchenkov B, Morel C, Lepleux M, De Luca V, Lartigue M, Cabanas H, Pujol N, Milvoy L, Morand P, Moncaubeig F, Wurtz H, Poinçot L, De Marco M, Jonckeau A, Pletenka J, Luquet E, Sovera A, Hardoüin J, Neves IJ, Machado-Hitau A, Schmit K, Piouceau L, Guilbert S, Manache-Alberici L, Lanero Fidalgo M, Dabée G, Dufourd T, Schroeder J, Alessandri K, Bezard E, Faggiani E, Feyeux M. Bioreactor-produced iPSCs-derived dopaminergic neuron-containing neural microtissues innervate and normalize rotational bias in a dose-dependent manner in a Parkinson rat model. Neurotherapeutics 2024; 21:e00436. [PMID: 39353832 PMCID: PMC11581877 DOI: 10.1016/j.neurot.2024.e00436] [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: 02/10/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 10/04/2024] Open
Abstract
A breadth of preclinical studies now support the rationale of pluripotent stem cell-derived cell replacement therapies to alleviate motor symptoms in Parkinsonian patients. Replacement of the primary dysfunctional cell population in the disease, i.e. the A9 dopaminergic neurons, is the major focus of these therapies. To achieve this, most therapeutical approaches involve grafting single-cell suspensions of DA progenitors. However, most cells die during the transplantation process, as cells face anoïkis. One potential solution to address this challenge is to graft solid preparations, i.e. adopting a 3D format. Cryopreserving such a format remains a major hurdle and is not exempt from causing delays in the time to effect, as observed with cryopreserved single-cell DA progenitors. Here, we used a high-throughput cell-encapsulation technology coupled with bioreactors to provide a 3D culture environment enabling the directed differentiation of hiPSCs into neural microtissues. The proper patterning of these neural microtissues into a midbrain identity was confirmed using orthogonal methods, including qPCR, RNAseq, flow cytometry and immunofluorescent microscopy. The efficacy of the neural microtissues was demonstrated in a dose-dependent manner using a Parkinsonian rat model. The survival of the cells was confirmed by post-mortem histological analysis, characterised by the presence of human dopaminergic neurons projecting into the host striatum. The work reported here is the first bioproduction of a cell therapy for Parkinson's disease in a scalable bioreactor, leading to a full behavioural recovery 16 weeks after transplantation using cryopreserved 3D format.
Collapse
Affiliation(s)
- Nicolas Prudon
- Université de Bordeaux, CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; TreeFrog Therapeutics, Bât A, F-33600 Pessac, France.
| | | | | | | | - Chloé Morel
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| | | | | | | | | | - Nadège Pujol
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| | - Loanne Milvoy
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| | | | | | - Hélène Wurtz
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| | - Léa Poinçot
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| | | | | | | | - Elisa Luquet
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| | - Andrea Sovera
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| | | | | | | | | | | | | | | | | | - Guillaume Dabée
- Université de Bordeaux, CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France; PIV-EXPE, Centre Broca, Université de Bordeaux, F-33000 Bordeaux, France
| | | | | | | | - Erwan Bezard
- Université de Bordeaux, CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | | | - Maxime Feyeux
- TreeFrog Therapeutics, Bât A, F-33600 Pessac, France
| |
Collapse
|
50
|
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
|