1
|
Paisey SJ, Jones LR, Harrison DJ, Drummond NJ, Edwards OZ, Canham MA, Roberton VH, Marshall C, Parker G, Hills R, Rosser AE, Lane EL, Dunnett SB, Kunath T, Assaf Y, Lelos MJ. Imaging of human stem cell-derived dopamine grafts correlates with behavioural recovery and reveals microstructural brain changes. Neurobiol Dis 2025; 209:106910. [PMID: 40233853 DOI: 10.1016/j.nbd.2025.106910] [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: 02/18/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
Cell therapy is a promising therapeutic intervention for Parkinson's disease (PD) and is currently undergoing safety and efficacy testing in clinical trials worldwide. The goals of this project were (1) to determine whether [18F]Fluorodopa or [18F]Fallypride imaging correlates robustly with functional recovery; and (2) to explore whether diffusion-weighted MR imaging (DWI) could detect graft-induced cytoarchitectural changes in the host brain. hfVM and hESC-derived dopamine precursor cells were transplanted into the 6-OHDA lesioned rat striatum. Tests of motor function and PET and MR imaging were conducted up to 6 months post-transplantation. Our data demonstrate that [18F]Fluorodopa imaging identified presynaptic DA synthesis from hfVM and hESC-derived dopaminergic grafts and [18F]Fallypride imaging confirmed occupancy and normalisation of D2/D3 receptor expression in the grafted hemisphere. In hfVM grafted rats, [18F]Fluorodopa binding correlated robustly with motor recovery on a range of drug-induced and drug-free behavioural tasks. In hESC-DA grafted rats, improvements in [18F]Fluorodopa PET imaging signals preceded recovery of naturalistic motor behaviours. DWI revealed widespread graft-mediated microstructural changes in the rodent brain, which did not identify graft placement, but instead may reflect remodelling of neuroglia. These data further our understanding of the impact of dopaminergic grafts on brain cytoarchitecture and the potential of these radioligands to predict graft efficacy may aid in the translation of therapeutics from preclinical to clinical settings.
Collapse
Affiliation(s)
- Stephen J Paisey
- Wales Research and Diagnostic PET Imaging Centre, School of Medicine, Cardiff University, University Hospital Wales Main Building, Cardiff CF14 4XN, UK
| | - Lucy R Jones
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - David J Harrison
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Nicola J Drummond
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Olivia Z Edwards
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Maurice A Canham
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Victoria H Roberton
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Christopher Marshall
- Wales Research and Diagnostic PET Imaging Centre, School of Medicine, Cardiff University, University Hospital Wales Main Building, Cardiff CF14 4XN, UK
| | - Greg Parker
- Independent Imaging Consultant, Museum Avenue, Cardiff CF10 3AX, UK
| | - Rachel Hills
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Anne E Rosser
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Emma L Lane
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Ave, Cardiff CF10 3NB, UK
| | - Stephen B Dunnett
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK; Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Yaniv Assaf
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK; School of Biochemistry Neurobiology Biophysics, Faculty of Life Sciences, Tel Aviv University, Israel
| | - Mariah J Lelos
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK; Medicines Discovery Institute, Cardiff University, Cardiff, CF10 3AT, UK.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Fasano A, Kundrick A, Henchcliffe C. Convention vs. innovation III: The promise of stem cell therapy in Parkinson's disease remains bright (PSG debate 2024). Parkinsonism Relat Disord 2025:107849. [PMID: 40335354 DOI: 10.1016/j.parkreldis.2025.107849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025]
Affiliation(s)
- Alfonso Fasano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, Milan, Italy.
| | - Avery Kundrick
- Center for Neurological Restoration, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Claire Henchcliffe
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
5
|
Zhu X, Zhao Y, Bai X, Dong Q, Tian C, Sun R, Yan C, Ruan J, Liu Z, Gao J. Small molecules direct the generation of ameloblast-like cells from human embryonic stem cells. Stem Cell Res Ther 2025; 16:173. [PMID: 40221796 PMCID: PMC11993985 DOI: 10.1186/s13287-025-04294-6] [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: 11/06/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Ameloblasts present a promising avenue for the investigation of enamel and tooth regeneration. Previous protocols for directing the differentiation of human embryonic stem cells (hESCs) into dental epithelial (DE) cells involving the need for additional cells, conditional medium, and the use of costly cytokines. Importantly, ameloblasts have not been generated from hESCs in previous studies. Hence, we aimed to identify defined differentiation conditions that would solely utilize small molecules to achieve the production of ameloblasts. METHODS We developed a three-step strategy entailing the progression of hESCs through non-neural ectoderm (NNE) and DE to generate functional ameloblasts in vitro. Initially, the NNE fate was induced from hESCs using a 6-day differentiation protocol with 1 µmol/L Retinoic acid (RA). Subsequently, the NNE lineage was differentiated into DE by employing a combination of 1 µmol/L LDN193189 (a BMP signaling inhibitor) and 1 µmol/L XAV939 (a WNT signaling inhibitor). In the final phase, 3 µmol/L CHIR99021 (a WNT signaling activator) and 2 µmol/L DAPT (a NOTCH signaling inhibitor) were utilized to achieve the fate of ameloblasts from DE cells. Three-dimensional cultures were investigated to enhance the ameloblast differentiation ability of the induced DE cells. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunofluorescence were conducted to assess the expression of lineage-specific markers. Alizarin Red S (ARS) staining was performed to evaluate the formation of mineralization nodules. RESULTS The application of RA facilitated the efficient generation of NNE within a six-day period. Subsequently, upon stimulation with LDN193189 and XAV939, a notable emergence of DE cells was observed on the eighth days. By the tenth day, ameloblast-like cells derived from hESCs were generated. Upon cultivation in spheroids, these cells exhibited elevated levels of ameloblast markers AMBN and AMELX expression, suggesting that spheroid culture augments the differentiation of ameloblasts. CONCLUSION We established an efficient small molecule-based method to differentiate hESCs into ameloblast-like cells through the concerted modulation of RA, BMP, WNT, and NOTCH signaling pathways, potentially advancing research in enamel and tooth regeneration.
Collapse
Affiliation(s)
- Ximei Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - YiMeng Zhao
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Xiaofan Bai
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Qiannan Dong
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Chunli Tian
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Ruilin Sun
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Congjuan Yan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jianping Ruan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Laboratory Center of Stomatology, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jianghong Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
6
|
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
|
7
|
Wang Y, Cao Y, Xie W, Guo Y, Cai J, Huang T, Li P. Advances in clinical translation of stem cell-based therapy in neurological diseases. J Cereb Blood Flow Metab 2025; 45:600-616. [PMID: 39883811 PMCID: PMC11783424 DOI: 10.1177/0271678x251317374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/03/2025] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Stem cell-based therapies have raised considerable interest to develop regenerative treatment for neurological disorders with high disability. In this review, we focus on recent preclinical and clinical evidence of stem cell therapy in the treatment of degenerative neurological diseases and discuss different cell types, delivery routes and biodistribution of stem cell therapy. In addition, recent advances of mechanistic insights of stem cell therapy, including functional replacement by exogenous cells, immunomodulation and paracrine effects of stem cell therapies are also demonstrated. Finally, we also highlight the adjunction approaches that has been implemented to augment their reparative function, survival and migration to target specific tissue, including stem cell preconditioning, genetical engineering, co-transplantation and combined therapy.
Collapse
Affiliation(s)
- Yu Wang
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yirong Cao
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Wanqing Xie
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Yunlu Guo
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Jiayi Cai
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Huang
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Peiying Li
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Kirkeby A, Main H, Carpenter M. Pluripotent stem-cell-derived therapies in clinical trial: A 2025 update. Cell Stem Cell 2025; 32:10-37. [PMID: 39753110 DOI: 10.1016/j.stem.2024.12.005] [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: 10/14/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 01/28/2025]
Abstract
Since the first derivation of human pluripotent stem cells (hPSCs) 27 years ago, technologies to control their differentiation and manufacturing have advanced immensely, enabling increasing numbers of clinical trials with hPSC-derived products. Here, we revew the landscape of interventional hPSC trials worldwide, highlighting available data on clinical safety and efficacy. As of December 2024, we identify 116 clinical trials with regulatory approval, testing 83 hPSC products. The majority of trials are targeting eye, central nervous system, and cancer. To date, more than 1,200 patients have been dosed with hPSC products, accumulating to >1011 clinically administered cells, so far showing no generalizable safety concerns.
Collapse
Affiliation(s)
- Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW) and Department of Neuroscience, 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, 221 84 Lund, Sweden.
| | - Heather Main
- HOYA Consulting (ReGenMed Solutions), Stockholm, Sweden
| | | |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Wu H, Zhang ZH, Zhou P, Sui X, Liu X, Sun Y, Zhao X, Pu XP. A Single-Cell Atlas of the Substantia Nigra Reveals Therapeutic Effects of Icaritin in a Rat Model of Parkinson's Disease. Antioxidants (Basel) 2024; 13:1183. [PMID: 39456437 PMCID: PMC11505506 DOI: 10.3390/antiox13101183] [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: 08/17/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/28/2024] Open
Abstract
Degeneration and death of dopaminergic neurons in the substantia nigra of the midbrain are the main pathological changes in Parkinson's disease (PD); however, the mechanism underlying the selective vulnerability of specific neuronal populations in PD remains unclear. Here, we used single-cell RNA sequencing to identify seven cell clusters, including oligodendrocytes, neurons, astrocytes, oligodendrocyte progenitor cells, microglia, synapse-rich cells (SRCs), and endothelial cells, in the substantia nigra of a rotenone-induced rat model of PD based on marker genes and functional definitions. We found that SRCs were a previously unidentified cell subtype, and the tight interactions between SRCs and other cell populations can be improved by icaritin, which is a flavonoid extracted from Epimedium sagittatum Maxim. and exerts anti-neuroinflammatory, antioxidant, and immune-improving effects in PD. We also demonstrated that icaritin bound with transcription factors of SRCs, and icaritin application modulated synaptic characterization of SRCs, neuroinflammation, oxidative stress, and survival of dopaminergic neurons, and improved abnormal energy metabolism, amino acid metabolism, and phospholipase D metabolism of astrocytes in the substantia nigra of rats with PD. Moreover, icaritin supplementation also promotes the recovery of the physiological homeostasis of the other cell clusters to delay the pathogenesis of PD. These data uncovered previously unknown cellular diversity in a rat model of Parkinson's disease and provide insights into the promising therapeutic potential of icaritin in PD.
Collapse
Affiliation(s)
- Hao Wu
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; (H.W.); (X.L.); (Y.S.)
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Zhen-Hua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China;
| | - Ping Zhou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China;
| | - Xin Sui
- The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Xi Liu
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; (H.W.); (X.L.); (Y.S.)
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- China State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi Sun
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; (H.W.); (X.L.); (Y.S.)
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xin Zhao
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; (H.W.); (X.L.); (Y.S.)
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiao-Ping Pu
- National Key Research Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China; (H.W.); (X.L.); (Y.S.)
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Park TY, Jeon J, Cha Y, Kim KS. Past, present, and future of cell replacement therapy for parkinson's disease: a novel emphasis on host immune responses. Cell Res 2024; 34:479-492. [PMID: 38777859 PMCID: PMC11217403 DOI: 10.1038/s41422-024-00971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Parkinson's disease (PD) stands as the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence continues to rise with the aging global population. Central to the pathophysiology of PD is the specific degeneration of midbrain dopamine neurons (mDANs) in the substantia nigra. Consequently, cell replacement therapy (CRT) has emerged as a promising treatment approach, initially supported by various open-label clinical studies employing fetal ventral mesencephalic (fVM) cells. Despite the initial favorable results, fVM cell therapy has intrinsic and logistical limitations that hinder its transition to a standard treatment for PD. Recent efforts in the field of cell therapy have shifted its focus towards the utilization of human pluripotent stem cells, including human embryonic stem cells and induced pluripotent stem cells, to surmount existing challenges. However, regardless of the transplantable cell sources (e.g., xenogeneic, allogeneic, or autologous), the poor and variable survival of implanted dopamine cells remains a major obstacle. Emerging evidence highlights the pivotal role of host immune responses following transplantation in influencing the survival of implanted mDANs, underscoring an important area for further research. In this comprehensive review, building upon insights derived from previous fVM transplantation studies, we delve into the functional ramifications of host immune responses on the survival and efficacy of grafted dopamine cells. Furthermore, we explore potential strategic approaches to modulate the host immune response, ultimately aiming for optimal outcomes in future clinical applications of CRT for PD.
Collapse
Affiliation(s)
- Tae-Yoon Park
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Jeha Jeon
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Young Cha
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA.
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA.
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard Medical School, Belmont, MA, USA.
| |
Collapse
|
18
|
Kwon M, Kim BS, Yoon S, Oh SO, Lee D. Hematopoietic Stem Cells and Their Niche in Bone Marrow. Int J Mol Sci 2024; 25:6837. [PMID: 38999948 PMCID: PMC11241602 DOI: 10.3390/ijms25136837] [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: 05/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Extensive research has explored the functional correlation between stem cells and progenitor cells, particularly in blood. Hematopoietic stem cells (HSCs) can self-renew and regenerate tissues within the bone marrow, while stromal cells regulate tissue function. Recent studies have validated the role of mammalian stem cells within specific environments, providing initial empirical proof of this functional phenomenon. The interaction between bone and blood has always been vital to the function of the human body. It was initially proposed that during evolution, mammalian stem cells formed a complex relationship with the surrounding microenvironment, known as the niche. Researchers are currently debating the significance of molecular-level data to identify individual stromal cell types due to incomplete stromal cell mapping. Obtaining these data can help determine the specific activities of HSCs in bone marrow. This review summarizes key topics from previous studies on HSCs and their environment, discussing current and developing concepts related to HSCs and their niche in the bone marrow.
Collapse
Affiliation(s)
- Munju Kwon
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
19
|
Christiansen JR, Kirkeby A. Clinical translation of pluripotent stem cell-based therapies: successes and challenges. Development 2024; 151:dev202067. [PMID: 38564308 PMCID: PMC11057818 DOI: 10.1242/dev.202067] [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] [Indexed: 04/04/2024]
Abstract
The translational stem cell research field has progressed immensely in the past decade. Development and refinement of differentiation protocols now allows the generation of a range of cell types, such as pancreatic β-cells and dopaminergic neurons, from human pluripotent stem cells (hPSCs) in an efficient and good manufacturing practice-compliant fashion. This has led to the initiation of several clinical trials using hPSC-derived cells to replace lost or dysfunctional cells, demonstrating evidence of both safety and efficacy. Here, we highlight successes from some of the hPSC-based trials reporting early signs of efficacy and discuss common challenges in clinical translation of cell therapies.
Collapse
Affiliation(s)
- Josefine Rågård Christiansen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), University of Copenhagen, 2200 Copenhagen N, Denmark
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark
- Wallenberg Center for Molecular Medicine, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
20
|
Ifediora N, Canoll P, Hargus G. Human stem cell transplantation models of Alzheimer's disease. Front Aging Neurosci 2024; 16:1354164. [PMID: 38450383 PMCID: PMC10915253 DOI: 10.3389/fnagi.2024.1354164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
Alzheimer's disease (AD) is the most frequent form of dementia. It is characterized by pronounced neuronal degeneration with formation of neurofibrillary tangles and deposition of amyloid β throughout the central nervous system. Animal models have provided important insights into the pathogenesis of AD and they have shown that different brain cell types including neurons, astrocytes and microglia have important functions in the pathogenesis of AD. However, there are difficulties in translating promising therapeutic observations in mice into clinical application in patients. Alternative models using human cells such as human induced pluripotent stem cells (iPSCs) may provide significant advantages, since they have successfully been used to model disease mechanisms in neurons and in glial cells in neurodegenerative diseases in vitro and in vivo. In this review, we summarize recent studies that describe the transplantation of human iPSC-derived neurons, astrocytes and microglial cells into the forebrain of mice to generate chimeric transplantation models of AD. We also discuss opportunities, challenges and limitations in using differentiated human iPSCs for in vivo disease modeling and their application for biomedical research.
Collapse
Affiliation(s)
- Nkechime Ifediora
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States
| |
Collapse
|
21
|
Foltynie T. Scaling up GMP-grade dopaminergic cells for Parkinson's disease. Cell Stem Cell 2024; 31:5-6. [PMID: 38181750 DOI: 10.1016/j.stem.2023.12.005] [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: 12/05/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024]
Abstract
Stem cell therapy for Parkinson's disease requires demonstration of safety and efficacy of dopaminergic cells derived from a cell line, consideration of dose, and whether this is deliverable at scale. Park et al. demonstrate these requirements for a new hESC line and that their manufacturing methods allow for its scalability.
Collapse
Affiliation(s)
- Thomas Foltynie
- National Hospital for Neurology & Neurosurgery, UCL Institute of Neurology, Queen Square, London, UK.
| |
Collapse
|