1
|
Zhang Z, Wang Y, Zhang Z, Qi Z. Network Meta-Analysis of Stem Cell Therapies for Parkinson's Disease: Exploring the Optimal Strategy Based on Animal Models. Stem Cells Dev 2025. [PMID: 40421708 DOI: 10.1089/scd.2025.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Stem cell therapy holds promise for Parkinson's disease (PD). To identify optimal stem cell regimens in PD mouse models and inform translational research, we conducted a network meta-analysis (NMA). Specifically, we systematically searched for studies on stem cell therapy in PD mouse models up to September 2024 in PubMed, Embase, Scopus, Web of Science, China National Knowledge Infrastructure, WANFANG, and VIP. Based on the data collected, we conducted an NMA using GeMTC-0.14.3 software. The results of traditional meta-analysis of 148 studies demonstrated superior efficacy of most interventions versus controls at biweekly intervals (2-8 weeks post-treatment), with neural stem cells engineered with neurotrophic factors (NSC-NFs) showing the lowest weighted mean difference, indicating optimal therapeutic effect. NMA demonstrated that NF-engineered NSC therapy ranked the highest at biweekly time points (2-8 weeks post-treatment). Doses of 105 cells showed optimal efficacy at 2, 4, and 6 weeks, peaking within this range, whereas doses of 103 cells showed the best efficacy at 8 weeks. Medial forebrain bundle (MFB) administration showed superior efficacy at weeks 2 and 8, while striatum (STR) infusion showed greater therapeutic effects at weeks 4 and 6, with both approaches significantly outperforming nasal and intravenous delivery at all evaluated time points (2, 4, 6, and 8 weeks). Taken together, these results suggest that NSC-NF (dosage of 105) delivered via MFB (at 2 and 8 weeks) or STR (at 4 and 6 weeks) may represent the optimal strategy. It provides important guidance for optimizing preclinical and clinical trial designs and offers valuable insights for clinical translation.
Collapse
Affiliation(s)
- Zehong Zhang
- School of Medicine, Guangxi University, Nanning, China
| | - Yingkai Wang
- School of Medicine, Guangxi University, Nanning, China
| | - Zhengmian Zhang
- Fujian Maternity and Child Health Hospital, Fuzhou, China
- Stem Cell Therapy Research Center, Fujian, China
- Fujian Provincial Human Sperm Bank, Fujian, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Zhongquan Qi
- School of Medicine, Guangxi University, Nanning, China
- Fujian Maternity and Child Health Hospital, Fuzhou, China
- Stem Cell Therapy Research Center, Fujian, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Tang P, Wei F, Qiao W, Chen X, Ji C, Yang W, Zhang X, Chen S, Wu Y, Jiang M, Ma C, Shen W, Dong Q, Cao H, Xie M, Cai Z, Xu L, Shi J, Dong N, Chen J, Wang N. Engineering aortic valves via transdifferentiating fibroblasts into valvular endothelial cells without using viruses or iPS cells. Bioact Mater 2025; 45:181-200. [PMID: 39651397 PMCID: PMC11625219 DOI: 10.1016/j.bioactmat.2024.11.018] [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: 07/31/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The technology of induced pluripotent stem cells (iPSCs) has enabled the conversion of somatic cells into primitive undifferentiated cells via reprogramming. This approach provides possibilities for cell replacement therapies and drug screening, but the potential risk of tumorigenesis hampers its further development and in vivo application. How to generate differentiated cells such as valvular endothelial cells (VECs) has remained a major challenge. Utilizing a combinatorial strategy of selective soluble chemicals, cytokines and substrate stiffness modulation, mouse embryonic fibroblasts are directly and efficiently transdifferentiated into induced aortic endothelial cell-like cells (iAECs), or human primary adult fibroblasts are transdifferentiated into induced valvular endothelial cell-like cells (hiVECs), without expressing pluripotency stem cell markers. These iAECs and hiVECs express VEC-associated genes and proteins and VEC-specific marker NFATC1 and are functional in culture and on decellularized porcine aortic valves, like mouse aortic endothelial cells or human primary aortic valvular endothelial cells. The iAECs and hiVECs seeded on decellularized porcine aortic valves stay intact and express VEC-associated proteins for 60 days after grafting into abdominal aorta of immune-compromised rats. In contrast, induced pluripotent stem cells (iPSCs) are less efficient in differentiating into VEC-like cells and pluripotency marker Nanog is expressed in a small subpopulation of iPSC-derived VEC-like cells that generate teratomas in SCID mice whereas hiVECs derived from transdifferentiation do not generate teratomas in vivo. Our findings highlight an approach to efficiently convert fibroblasts into iAECs and hiVECs and seed them onto decellularized aortic valves for safely generating autologous tissue-engineered aortic valves without using viruses or first reprogramming the cells into pluripotent stem cells.
Collapse
Affiliation(s)
- Peng Tang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Fuxiang Wei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chenyang Ji
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Wanzhi Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Xinyu Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Sihan Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yanyan Wu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Mingxing Jiang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Chenyu Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Weiqiang Shen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Qi Dong
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Minghui Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ziwen Cai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junwei Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Ning Wang
- Institute for Mechanobiology, Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
5
|
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
|
6
|
Cai H, Huang J, Wang W, Lin W, Ahmed W, Lu D, Quan J, Chen L. Characteristics of Parthenogenetic Stem Cells and Their Potential Treatment Strategy for Central Nervous System Diseases. Neuropsychiatr Dis Treat 2025; 21:213-227. [PMID: 39926116 PMCID: PMC11804250 DOI: 10.2147/ndt.s497758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/14/2025] [Indexed: 02/11/2025] Open
Abstract
Stem cells hold significant promise in treating neurological illnesses, such as stroke, spinal cord injury and neurodegenerative diseases. The origins and characteristics of human parthenogenetic stem cells might lead to a new research area in treating nervous system diseases. The current clinical studies in the field of traumatic brain injury and neurodegenerative diseases are reviewed. Some variables that influence common stem cells' survival, proliferation, and therapeutic efficacy will be mentioned in this paper because they may play an important role in studying parthenogenetic stem cells.
Collapse
Affiliation(s)
- Hengsen Cai
- Department of Neurosurgery, The second People’s Hospital of Pingnan, Pingnan, Guangxi, People’s Republic of China
| | - Jiajun Huang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, 510310, People’s Republic of China
| | - Wei Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, 510310, People’s Republic of China
| | - Wentong Lin
- Department of Orthopaedics, Chaozhou Hospital of Traditional Chinese Medicine, Chaozhou, Guangdong, People’s Republic of China
| | - Waqas Ahmed
- Department of Neurology, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Deng Lu
- Department of Neurosurgery, The second People’s Hospital of Pingnan, Pingnan, Guangxi, People’s Republic of China
| | - Jiewei Quan
- Department of Neurosurgery, The second People’s Hospital of Pingnan, Pingnan, Guangxi, People’s Republic of China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, 510310, People’s Republic of China
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Yasuda S, Bando K, Henry MP, Libertini S, Watanabe T, Bando H, Chen C, Fujimori K, Harada K, Kuroda T, Lemmens M, Marginean D, Moss D, Pereira Mouriès L, Nicholas NS, Smart MJK, Terai O, Sato Y. Detection of residual pluripotent stem cells in cell therapy products utilizing droplet digital PCR: an international multisite evaluation study. Stem Cells Transl Med 2024; 13:1001-1014. [PMID: 39120125 PMCID: PMC11465167 DOI: 10.1093/stcltm/szae058] [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: 02/28/2024] [Accepted: 06/08/2024] [Indexed: 08/10/2024] Open
Abstract
The presence of residual undifferentiated pluripotent stem cells (PSCs) in PSC-derived cell therapy products (CTPs) is a major safety issue for their clinical application, due to the potential risk of PSC-derived tumor formation. An international multidisciplinary multisite study to evaluate a droplet digital PCR (ddPCR) approach to detect residual undifferentiated PSCs in PSC-derived CTPs was conducted as part of the Health and Environmental Sciences Institute Cell Therapy-TRAcking, Circulation & Safety Technical Committee. To evaluate the use of ddPCR in quantifying residual iPSCs in a cell sample, different quantities of induced pluripotent stem cells (iPSCs) were spiked into a background of iPSC-derived cardiomyocytes (CMs) to mimic different concentrations of residual iPSCs. A one step reverse transcription ddPCR (RT-ddPCR) was performed to measure mRNA levels of several iPSC-specific markers and to evaluate the assay performance (precision, sensitivity, and specificity) between and within laboratories. The RT-ddPCR assay variability was initially assessed by measuring the same RNA samples across all participating facilities. Subsequently, each facility independently conducted the entire process, incorporating the spiking step, to discern the parameters influencing potential variability. Our results show that a RT-ddPCR assay targeting ESRG, LINC00678, and LIN28A genes offers a highly sensitive and robust detection of impurities of iPSC-derived CMs and that the main contribution to variability between laboratories is the iPSC-spiking procedure, and not the RT-ddPCR. The RT-ddPCR assay would be generally applicable for tumorigenicity evaluation of PSC-derived CTPs with appropriate marker genes suitable for each CTP.
Collapse
Affiliation(s)
- Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | | | | | | | | | | | - Connie Chen
- Health and Environmental Sciences Institute, Washington, DC, United States
| | | | - Kosuke Harada
- Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | | | | | - David Moss
- Cell and Gene Therapy Catapult, London, United Kingdom
| | | | | | | | | | - Yoji Sato
- Division of Drugs, National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
11
|
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
|
12
|
Khandia R, Gurjar P, Priyanka, Romashchenko V, Al-Hussain SA, Zaki MEA. Recent advances in stem cell therapy: efficacy, ethics, safety concerns, and future directions focusing on neurodegenerative disorders - a review. Int J Surg 2024; 110:6367-6381. [PMID: 39705668 DOI: 10.1097/js9.0000000000001609] [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: 01/21/2024] [Accepted: 04/29/2024] [Indexed: 12/22/2024]
Abstract
Neurodegeneration refers to the gradual loss of neurons and extensive changes in glial cells like tau inclusions in astrocytes and oligodendrocytes, α-synuclein inclusions in oligodendrocytes and SOD1 aggregates in astrocytes along with deterioration in the motor, cognition, learning, and behavior. Common neurodegenerative disorders are Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), spinocerebellar ataxia (SCA), and supranuclear palsy. There is a lack of effective treatment for neurodegenerative diseases, and scientists are putting their efforts into developing therapies against them. Stem cell therapy has emerged as a hope for neurodegenerative disorders since it is not only the damaged neurons that might be replaced, but other neuromodulators and neuroprotectors are secreted. Stem cell terminal differentiation before implantation ensures the implantation of correct cells and molecular markers like carbonic anhydrase II, CNPase (2',3'-cyclic nucleotide 3'-phosphohydrolase), myelin basic protein (MBP), and myelin oligodendrocyte glycoprotein (MOG) elucidate the differentiation. Secretion of various growth factors like epidermal growth factor (EGF), keratinocyte growth factor (KGF), vascular endothelial growth factor-α (VEGF-α), transforming growth factor (TGF), and macrophage inflammatory protein (MIP) supports cell survival, cell proliferation, blood vessel formation, axon regeneration, and neuroglial functional connection formation at the site of degeneration. Adverse effects of stem cell therapy, like teratogenicity and differentiation in different cells other than the desired one under the influence of microenvironment, are a few key concerns. Post-transplantation improved synaptic plasticity, apoptosis inhibition, and reduction in tau-phosphorylation and amyloid beta (Aβ) production has been observed in Alzheimer's patients. A large number of experimental, preclinical, and clinical studies have been conducted, and encouraging results have been obtained. The present review exhaustively discusses various kinds of stem cells, their usage in treating neurodegenerative disorders, limitations and challenges, and ethical issues related to stem cell therapy.
Collapse
Affiliation(s)
- Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh
| | - Pankaj Gurjar
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
| | - Priyanka
- Department of Veterinary Microbiology, College of Veterinary Science, Guru AngadDev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda, Punjab, India
| | | | - Sami A Al-Hussain
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E A Zaki
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
13
|
Wang F, Cheng XY, Zhang YT, Bai QR, Zhang XQ, Sun XC, Ma QH, Zhao XF, Liu CF. Transplantation of human neural stem cell prevents symptomatic motor behavior disability in a rat model of Parkinson's disease. Open Life Sci 2024; 19:20220834. [PMID: 38465343 PMCID: PMC10921471 DOI: 10.1515/biol-2022-0834] [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: 09/25/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 03/12/2024] Open
Abstract
Parkinson's disease (PD) is a ubiquitous brain cell degeneration disease and presents a significant therapeutic challenge. By injecting 6-hydroxydopamine (6-OHDA) into the left medial forebrain bundle, rats were made to exhibit PD-like symptoms and treated by intranasal administration of a low-dose (2 × 105) or high-dose (1 × 106) human neural stem cells (hNSCs). Apomorphine-induced rotation test, stepping test, and open field test were implemented to evaluate the motor behavior and high-performance liquid chromatography was carried out to detect dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC), serotonin, and 5-hydroxyindole-3-acetic acid in the striatum of rats. Animals injected with 6-OHDA showed significant motor function deficits and damaged dopaminergic system compared to the control group, which can be restored by hNSCs treatment. Treatment with hNSCs significantly increased the tyrosine hydroxylase-immunoreactive cell count in the substantia nigra of PD animals. Moreover, the levels of neurotransmitters exhibited a significant decline in the striatum tissue of animals injected with 6-OHDA when compared to that of the control group. However, transplantation of hNSCs significantly elevated the concentration of DA and DOPAC in the injured side of the striatum. Our study offered experimental evidence to support prospects of hNSCs for clinical application as a cell-based therapy for PD.
Collapse
Affiliation(s)
- Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou215004, China
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Yu Cheng
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou215004, China
| | - Yu-Ting Zhang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou215004, China
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Qing-Ran Bai
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200333, China
| | - Xiao-Qi Zhang
- Shanghai Angecon Biotechnology Co., Ltd, Shanghai, 201318, China
| | - Xi-Cai Sun
- Shanghai Angecon Biotechnology Co., Ltd, Shanghai, 201318, China
| | - Quan-Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou215004, China
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiong-Fei Zhao
- Shanghai Angecon Biotechnology Co., Ltd, Shanghai, 201318, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou215004, China
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| |
Collapse
|
14
|
Zhang L, Yang H. Research progress of neural stem cells as a source of dopaminergic neurons for cell therapy in Parkinson's disease. Mol Biol Rep 2024; 51:347. [PMID: 38400887 DOI: 10.1007/s11033-024-09294-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, the most characteristic pathological feature is the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compactus (SNpc) of the mesencephalon, along with reduced dopamine content in the striatum. Researchers have been searching for drugs and therapies to treat PD in decades. However, no approach could stop the progression of the disease, and even some of them caused adverse clinical side effects. PD has a well-defined lesion. Therefore, it is considered to be one of the most curable central nervous system diseases by cell replacement treatment. Fetal ventral mesencephalic tissue transplantation has been used to treat patients with PD and obtained positive treatment results. However, ethical issues, such as limited donor tissue, and side effects including graft-induced dyskinesias, limit its clinical applications. Neural stem cell (NSC) transplantation is a viable therapy choice because it possesses multipotency, self-renewal ability, and differentiation into DA neurons, which may substitute for lost DA neurons and slow down the neurodegenerative process in PD. Studies that investigated the delivery of NSCs by using animal models of PD revealed survival, migration, and even amelioration of behavioral deficits. Here, the research progress of NSCs or NSC-derived DA neurons in treating PD was reviewed, and the practicability of present manufacturing processes for clinical testing was considered. This review is expected to offer ideas for practical strategies to solve the present technical and biological problems related to the clinical application of NSCs in PD.
Collapse
Affiliation(s)
- Lingling Zhang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, 555 East Youyi Road, Beilin District, Xi'an, 710054, China.
| | - Hao Yang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, 555 East Youyi Road, Beilin District, Xi'an, 710054, China
| |
Collapse
|
15
|
Park S, Park CW, Eom JH, Jo MY, Hur HJ, Choi SK, Lee JS, Nam ST, Jo KS, Oh YW, Lee J, Kim S, Kim DH, Park CY, Kim SJ, Lee HY, Cho MS, Kim DS, Kim DW. Preclinical and dose-ranging assessment of hESC-derived dopaminergic progenitors for a clinical trial on Parkinson's disease. Cell Stem Cell 2024; 31:25-38.e8. [PMID: 38086390 DOI: 10.1016/j.stem.2023.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/25/2023] [Accepted: 11/17/2023] [Indexed: 01/07/2024]
Abstract
Human embryonic stem cell (hESC)-derived midbrain dopaminergic (mDA) cell transplantation is a promising therapeutic strategy for Parkinson's disease (PD). Here, we present the derivation of high-purity mDA progenitors from clinical-grade hESCs on a large scale under rigorous good manufacturing practice (GMP) conditions. We also assessed the toxicity, biodistribution, and tumorigenicity of these cells in immunodeficient rats in good laboratory practice (GLP)-compliant facilities. Various doses of mDA progenitors were transplanted into hemi-parkinsonian rats, and a significant dose-dependent behavioral improvement was observed with a minimal effective dose range of 5,000-10,000 mDA progenitor cells. These results provided insights into determining a low cell dosage (3.15 million cells) for human clinical trials. Based on these results, approval for a phase 1/2a clinical trial for PD cell therapy was obtained from the Ministry of Food and Drug Safety in Korea, and a clinical trial for treating patients with PD has commenced.
Collapse
Affiliation(s)
- Sanghyun Park
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Chan Wook Park
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | | | - Mi-Young Jo
- S. Biomedics Co., Ltd., Seoul 04797, Republic of Korea
| | - Hye-Jin Hur
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; S. Biomedics Co., Ltd., Seoul 04797, Republic of Korea
| | | | - Jae Souk Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | | | - Ki-Sang Jo
- S. Biomedics Co., Ltd., Seoul 04797, Republic of Korea
| | - Young Woo Oh
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea, 21 PLUS Program for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jungil Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea, 21 PLUS Program for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sieun Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea, 21 PLUS Program for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Do-Hun Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; S. Biomedics Co., Ltd., Seoul 04797, Republic of Korea
| | - Chul-Yong Park
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; S. Biomedics Co., Ltd., Seoul 04797, Republic of Korea
| | - Su Jin Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Gyeonggi-do, Republic of Korea
| | - Myung Soo Cho
- S. Biomedics Co., Ltd., Seoul 04797, Republic of Korea
| | - Dae-Sung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; Department of Pediatrics, Korea University College of Medicine, Guro Hospital, Seoul 08308, Republic of Korea.
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; S. Biomedics Co., Ltd., Seoul 04797, Republic of Korea; Brain Korea, 21 PLUS Program for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
16
|
Song XY, Fan CX, Atta-ur-Rahman FRS, Choudhary MI, Wang XP. Neuro-regeneration or Repair: Cell Therapy of Neurological Disorders as A Way Forward. Curr Neuropharmacol 2024; 22:2272-2283. [PMID: 38939990 PMCID: PMC11451317 DOI: 10.2174/1570159x22666240509092903] [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: 01/25/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 06/29/2024] Open
Abstract
The human central nervous system (CNS) has a limited capacity for regeneration and repair, as many other organs do. Partly as a result, neurological diseases are the leading cause of medical burden globally. Most neurological disorders cannot be cured, and primary treatments focus on managing their symptoms and slowing down their progression. Cell therapy for neurological disorders offers several therapeutic potentials and provides hope for many patients. Here we provide a general overview of cell therapy in neurological disorders such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Wilson's disease (WD), stroke and traumatic brain injury (TBI), involving many forms of stem cells, including embryonic stem cells and induced pluripotent stem cells. We also address the current concerns and perspectives for the future. Most studies for cell therapy in neurological diseases are in the pre-clinical stage, and there is still a great need for further research to translate neural replacement and regenerative therapies into clinical settings.
Collapse
Affiliation(s)
- Xiao-Yan Song
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Cun-Xiu Fan
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Atta-ur-Rahman FRS
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Muhammad Iqbal Choudhary
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Xiao-Ping Wang
- Department of Neurology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Harary PM, Jgamadze D, Kim J, Wolf JA, Song H, Ming GL, Cullen DK, Chen HI. Cell Replacement Therapy for Brain Repair: Recent Progress and Remaining Challenges for Treating Parkinson's Disease and Cortical Injury. Brain Sci 2023; 13:1654. [PMID: 38137103 PMCID: PMC10741697 DOI: 10.3390/brainsci13121654] [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: 10/19/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Neural transplantation represents a promising approach to repairing damaged brain circuitry. Cellular grafts have been shown to promote functional recovery through "bystander effects" and other indirect mechanisms. However, extensive brain lesions may require direct neuronal replacement to achieve meaningful restoration of function. While fetal cortical grafts have been shown to integrate with the host brain and appear to develop appropriate functional attributes, the significant ethical concerns and limited availability of this tissue severely hamper clinical translation. Induced pluripotent stem cell-derived cells and tissues represent a more readily scalable alternative. Significant progress has recently been made in developing protocols for generating a wide range of neural cell types in vitro. Here, we discuss recent progress in neural transplantation approaches for two conditions with distinct design needs: Parkinson's disease and cortical injury. We discuss the current status and future application of injections of dopaminergic cells for the treatment of Parkinson's disease as well as the use of structured grafts such as brain organoids for cortical repair.
Collapse
Affiliation(s)
- Paul M. Harary
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.M.H.)
| | - Dennis Jgamadze
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.M.H.)
| | - Jaeha Kim
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.M.H.)
| | - John A. Wolf
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.M.H.)
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - D. Kacy Cullen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.M.H.)
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - H. Isaac Chen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.M.H.)
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Cherkashova E, Namestnikova D, Leonov G, Gubskiy I, Sukhinich K, Melnikov P, Chekhonin V, Yarygin K, Goldshtein D, Salikhova D. Comparative study of the efficacy of intra-arterial and intravenous transplantation of human induced pluripotent stem cells-derived neural progenitor cells in experimental stroke. PeerJ 2023; 11:e16358. [PMID: 38025691 PMCID: PMC10640846 DOI: 10.7717/peerj.16358] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Background Cell therapy using neural progenitor cells (NPCs) is a promising approach for ischemic stroke treatment according to the results of multiple preclinical studies in animal stroke models. In the vast majority of conducted animal studies, the therapeutic efficacy of NPCs was estimated after intracerebral transplantation, while the information of the effectiveness of systemic administration is limited. Nowadays, several clinical trials aimed to estimate the safety and efficacy of NPCs transplantation in stroke patients were also conducted. In these studies, NPCs were transplanted intracerebrally in the subacute/chronic phase of stroke. The results of clinical trials confirmed the safety of the approach, however, the degree of functional improvement (the primary efficacy endpoint) was not sufficient in the majority of the studies. Therefore, more studies are needed in order to investigate the optimal transplantation parameters, especially the timing of cell transplantation after the stroke onset. This study aimed to evaluate the therapeutic effects of intra-arterial (IA) and intravenous (IV) administration of NPCs derived from induced pluripotent stem cells (iNPCs) in the acute phase of experimental stroke in rats. Induced pluripotent stem cells were chosen as the source of NPCs as this technology is perspective, has no ethical concerns and provides the access to personalized medicine. Methods Human iNPCs were transplanted IA or IV into male Wistar rats 24 h after the middle cerebral artery occlusion stroke modeling. Therapeutic efficacy was monitored for 14 days and evaluated in comparison with the cell transplantation-free control group. Additionally, cell distribution in the brain was assessed. Results The obtained results show that both routes of systemic transplantation (IV and IA) significantly reduced the mortality and improved the neurological deficit of experimental animals compared to the control group. At the same time, according to the MRI data, only IA administration led to faster and prominent reduction of the stroke volume. After IA administration, iNPCs transiently trapped in the brain and were not detected on day 7 after the transplantation. In case of IV injection, transplanted cells were not visualized in the brain. The obtained data demonstrated that the systemic transplantation of human iNPCs in the acute phase of ischemic stroke can be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Elvira Cherkashova
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency of Russian Federation, Moscow, Russian Federation
| | - Daria Namestnikova
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency of Russian Federation, Moscow, Russian Federation
| | - Georgiy Leonov
- Orekhovich Research Institute of Biomedical Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Ilya Gubskiy
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency of Russian Federation, Moscow, Russian Federation
| | - Kirill Sukhinich
- Orekhovich Research Institute of Biomedical Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Pavel Melnikov
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Vladimir Chekhonin
- Pirogov Russian National Research Medical University of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Healthcare of Russian Federation, Moscow, Russian Federation
| | - Konstantin Yarygin
- Orekhovich Research Institute of Biomedical Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
- Russian Medical Academy of Continuous Professional Education of the Ministry of Healthcare of the Russian Federation, Moscow, Russian Federation
| | | | - Diana Salikhova
- Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University, Moscow, Russian Federation
| |
Collapse
|
19
|
Capelli C, Cuofano C, Pavoni C, Frigerio S, Lisini D, Nava S, Quaroni M, Colombo V, Galli F, Bezukladova S, Panina-Bordignon P, Gaipa G, Comoli P, Cossu G, Martino G, Biondi A, Introna M, Golay J. Potency assays and biomarkers for cell-based advanced therapy medicinal products. Front Immunol 2023; 14:1186224. [PMID: 37359560 PMCID: PMC10288881 DOI: 10.3389/fimmu.2023.1186224] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Advanced Therapy Medicinal Products (ATMPs) based on somatic cells expanded in vitro, with or without genetic modification, is a rapidly growing area of drug development, even more so following the marketing approval of several such products. ATMPs are produced according to Good Manufacturing Practice (GMP) in authorized laboratories. Potency assays are a fundamental aspect of the quality control of the end cell products and ideally could become useful biomarkers of efficacy in vivo. Here we summarize the state of the art with regard to potency assays used for the assessment of the quality of the major ATMPs used clinic settings. We also review the data available on biomarkers that may substitute more complex functional potency tests and predict the efficacy in vivo of these cell-based drugs.
Collapse
Affiliation(s)
- Chiara Capelli
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Carolina Cuofano
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Chiara Pavoni
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Simona Frigerio
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Lisini
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Nava
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Michele Quaroni
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Valentina Colombo
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Francesco Galli
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, Manchester, United Kingdom
| | - Svetlana Bezukladova
- Università Vita-Salute San Raffaele, Milan, Italy
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
| | - Paola Panina-Bordignon
- Università Vita-Salute San Raffaele, Milan, Italy
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
| | - Giuseppe Gaipa
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giulio Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, Manchester, United Kingdom
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Gianvito Martino
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Biondi
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Martino Introna
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Josée Golay
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
20
|
Skidmore S, Barker RA. Challenges in the clinical advancement of cell therapies for Parkinson's disease. Nat Biomed Eng 2023; 7:370-386. [PMID: 36635420 PMCID: PMC7615223 DOI: 10.1038/s41551-022-00987-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 11/04/2022] [Indexed: 01/14/2023]
Abstract
Cell therapies as potential treatments for Parkinson's disease first gained traction in the 1980s, owing to the clinical success of trials that used transplants of foetal midbrain dopaminergic tissue. However, the poor standardization of the tissue for grafting, and constraints on its availability and ethical use, have hindered this treatment strategy. Recent advances in stem-cell technologies and in the understanding of the development of dopaminergic neurons have enabled preclinical advancements of promising stem-cell therapies. To move these therapies to the clinic, appropriate levels of safety screening, as well as optimization of the cell products and the scalability of their manufacturing, will be required. In this Review, we discuss how challenges pertaining to cell sources, functional and safety testing, manufacturing and storage, and clinical-trial design are being addressed to advance the translational and clinical development of cell therapies for Parkinson's disease.
Collapse
Affiliation(s)
- Sophie Skidmore
- Wellcome and MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre Cambridge Biomedical Campus, Cambridge, UK
| | - Roger A Barker
- Wellcome and MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre Cambridge Biomedical Campus, Cambridge, UK.
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, For vie Site, Cambridge, UK.
| |
Collapse
|
21
|
Watanabe T, Yasuda S, Chen CL, Delsing L, Fellows MD, Foldes G, Kusakawa S, Mouriès LP, Sato Y. International evaluation study of a highly efficient culture assay for detection of residual human pluripotent stem cells in cell therapies. Regen Med 2023; 18:219-227. [PMID: 36852420 DOI: 10.2217/rme-2022-0207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Aim & methods: The Health and Environmental Sciences Institute Cell Therapy-TRAcking, Circulation & Safety Technical Committee launched an international, multisite study to evaluate the sensitivity and reproducibility of the highly efficient culture (HEC) assay, an in vitro assay to detect residual undifferentiated human pluripotent stem cells (hPSCs) in cell therapy products. Results: All facilities detected colonies of human induced pluripotent stem cells (hiPSCs) when five hiPSCs were spiked into 1 million hiPSC-derived cardiomyocytes. Spiking with a trace amount of hiPSCs revealed that repeatability accounts for the majority of reproducibility while the true positive rate was high. Conclusion: The results indicate that the HEC assay is highly sensitive and robust and can be generally applicable for tumorigenicity evaluation of hPSC-derived cell therapy products.
Collapse
Affiliation(s)
- Takeshi Watanabe
- Drug Safety Research & Evaluation, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Connie L Chen
- Health & Environmental Sciences Institute, 740 Fifteenth Street NW, Suite 600, Washington, DC 20005, USA
| | - Louise Delsing
- CVRM Safety, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Mick D Fellows
- CVRM Safety, Clinical Pharmacology & Safety Science, R&D, AstraZeneca, Darwin Building 310, Milton Science Park, Cambridge, CB4 OWG, UK
| | - Gabor Foldes
- National Heart & Lung Institute, Imperial College London, London, W120NN, UK.,Current address, BioPharmaceuticals R&D, AstraZeneca, Milstein Building, Granta Park, Cambridge, CB21 6GH, UK
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Lucilia Pereira Mouriès
- Health & Environmental Sciences Institute, 740 Fifteenth Street NW, Suite 600, Washington, DC 20005, USA
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| |
Collapse
|
22
|
Li X, Zhu Y, Wang Y, Xia X, Zheng JC. Neural stem/progenitor cell-derived extracellular vesicles: A novel therapy for neurological diseases and beyond. MedComm (Beijing) 2023; 4:e214. [PMID: 36776763 PMCID: PMC9905070 DOI: 10.1002/mco2.214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
As bilayer lipid membrane vesicles secreted by neural stem/progenitor cells (NSCs), NSC-derived extracellular vesicles (NSC-EVs) have attracted growing attention for their promising potential to serve as novel therapeutic agents in treatment of neurological diseases due to their unique physicochemical characteristics and biological functions. NSC-EVs exhibit advantages such as stable physical and chemical properties, low immunogenicity, and high penetration capacity to cross blood-brain barrier to avoid predicaments of the clinical applications of NSCs that include autoimmune responses, ethical/religious concerns, and the problematic logistics of acquiring fetal tissues. More importantly, NSC-EVs inherit excellent neuroprotective and neuroregenerative potential and immunomodulatory capabilities from parent cells, and display outstanding therapeutic effects on mitigating behavioral alterations and pathological phenotypes of patients or animals with neurological diseases. In this review, we first comprehensively summarize the progress in functional research and application of NSC-EVs in different neurological diseases, including neurodegenerative diseases, acute neurological diseases, dementia/cognitive dysfunction, and peripheral diseases. Next, we provide our thoughts on current limitations/concerns as well as tremendous potential of NSC-EVs in clinical applications. Last, we discuss future directions of further investigations on NSC-EVs and their probable applications in both basic and clinical research.
Collapse
Affiliation(s)
- Xiangyu Li
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
| | - Yingbo Zhu
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
| | - Yi Wang
- Center for Translational Neurodegeneration and Regenerative TherapyYangzhi Rehabilitation Hospital, Tongji UniversityShanghaiChina
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Frontiers Science Center of Nanocatalytic MedicineTongji University School of MedicineShanghaiChina
- Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, Tongji University School of MedicineShanghaiChina
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Tongji UniversityMinistry of EducationShanghaiChina
| | - Jialin C. Zheng
- Center for Translational Neurodegeneration and Regenerative TherapyTongji Hospital, Tongji University School of MedicineShanghaiChina
- Shanghai Frontiers Science Center of Nanocatalytic MedicineTongji University School of MedicineShanghaiChina
- Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's Hospital, Tongji University School of MedicineShanghaiChina
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Tongji UniversityMinistry of EducationShanghaiChina
| |
Collapse
|
23
|
Yeap YJ, Teddy TJW, Lee MJ, Goh M, Lim KL. From 2D to 3D: Development of Monolayer Dopaminergic Neuronal and Midbrain Organoid Cultures for Parkinson's Disease Modeling and Regenerative Therapy. Int J Mol Sci 2023; 24:ijms24032523. [PMID: 36768843 PMCID: PMC9917335 DOI: 10.3390/ijms24032523] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Parkinson's Disease (PD) is a prevalent neurodegenerative disorder that is characterized pathologically by the loss of A9-specific dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain. Despite intensive research, the etiology of PD is currently unresolved, and the disease remains incurable. This, in part, is due to the lack of an experimental disease model that could faithfully recapitulate the features of human PD. However, the recent advent of induced pluripotent stem cell (iPSC) technology has allowed PD models to be created from patient-derived cells. Indeed, DA neurons from PD patients are now routinely established in many laboratories as monolayers as well as 3D organoid cultures that serve as useful toolboxes for understanding the mechanism underlying PD and also for drug discovery. At the same time, the iPSC technology also provides unprecedented opportunity for autologous cell-based therapy for the PD patient to be performed using the patient's own cells as starting materials. In this review, we provide an update on the molecular processes underpinning the development and differentiation of human pluripotent stem cells (PSCs) into midbrain DA neurons in both 2D and 3D cultures, as well as the latest advancements in using these cells for drug discovery and regenerative medicine. For the novice entering the field, the cornucopia of differentiation protocols reported for the generation of midbrain DA neurons may seem daunting. Here, we have distilled the essence of the different approaches and summarized the main factors driving DA neuronal differentiation, with the view to provide a useful guide to newcomers who are interested in developing iPSC-based models of PD.
Collapse
Affiliation(s)
- Yee Jie Yeap
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Tng J. W. Teddy
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Interdisciplinary Graduate Programme (IGP-Neuroscience), Nanyang Technological University, Singapore 639798, Singapore
| | - Mok Jung Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Micaela Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Kah Leong Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- National Neuroscience Institute, Singapore 308433, Singapore
- Department of Brain Sciences, Imperial College London, London SW7 2AZ, UK
- Department of Anatomy, Shanxi Medical University, Taiyuan 030001, China
- Correspondence:
| |
Collapse
|
24
|
Galiakberova AA, Brovkina OI, Kondratyev NV, Artyuhov AS, Momotyuk ED, Kulmukhametova ON, Lagunin AA, Shilov BV, Zadorozhny AD, Zakharov IS, Okorokova LS, Golimbet VE, Dashinimaev EB. Different iPSC-derived neural stem cells shows various spectrums of spontaneous differentiation during long term cultivation. Front Mol Neurosci 2023; 16:1037902. [PMID: 37201156 PMCID: PMC10186475 DOI: 10.3389/fnmol.2023.1037902] [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: 09/06/2022] [Accepted: 03/23/2023] [Indexed: 05/20/2023] Open
Abstract
Introduction Culturing of human neural stem cells (NSCs) derived from induced pluripotent stem cells (iPSC) is a promising area of research, as these cells have the potential to treat a wide range of neurological, neurodegenerative and psychiatric diseases. However, the development of optimal protocols for the production and long-term culturing of NSCs remains a challenge. One of the most important aspects of this problem is to determine the stability of NSCs during long-term in vitro passaging. To address this problem, our study was aimed at investigating the spontaneous differentiation profile in different iPSC-derived human NSCs cultures during long-term cultivation using. Methods Four different IPSC lines were used to generate NSC and spontaneously differentiated neural cultures using DUAL SMAD inhibition. These cells were analyzed at different passages using immunocytochemistry, qPCR, bulk transcriptomes and scRNA-seq. Results We found that various NSC lines generate significantly different spectrums of differentiated neural cells, which can also change significantly during long-term cultivation in vitro. Discussion Our results indicate that both internal (genetic and epigenetic) and external (conditions and duration of cultivation) factors influence the stability of NSCs. These results have important implications for the development of optimal NSCs culturing protocols and highlight the need to further investigate the factors influencing the stability of these cells in vitro.
Collapse
Affiliation(s)
- Adelya Albertovna Galiakberova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Igorevna Brovkina
- Federal Research and Clinical Center, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | | | - Alexander Sergeevich Artyuhov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Ekaterina Dmitrievna Momotyuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - Alexey Aleksandrovich Lagunin
- Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | - Igor Sergeevitch Zakharov
- Department of Bioinformatics, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | - Erdem Bairovich Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of Bioinformatics, Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
- *Correspondence: Erdem Bairovich Dashinimaev,
| |
Collapse
|
25
|
Cha Y, Park TY, Leblanc P, Kim KS. Current Status and Future Perspectives on Stem Cell-Based Therapies for Parkinson's Disease. J Mov Disord 2023; 16:22-41. [PMID: 36628428 PMCID: PMC9978267 DOI: 10.14802/jmd.22141] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 01/12/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, affecting 1%-2% of the population over the age of 65. As the population ages, it is anticipated that the burden on society will significantly escalate. Although symptom reduction by currently available pharmacological and/or surgical treatments improves the quality of life of many PD patients, there are no treatments that can slow down, halt, or reverse disease progression. Because the loss of a specific cell type, midbrain dopamine neurons in the substantia nigra, is the main cause of motor dysfunction in PD, it is considered a promising target for cell replacement therapy. Indeed, numerous preclinical and clinical studies using fetal cell transplantation have provided proof of concept that cell replacement therapy may be a viable therapeutic approach for PD. However, the use of human fetal cells remains fraught with controversy due to fundamental ethical, practical, and clinical limitations. Groundbreaking work on human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, coupled with extensive basic research in the stem cell field offers promising potential for hPSC-based cell replacement to become a realistic treatment regimen for PD once several major issues can be successfully addressed. In this review, we will discuss the prospects and challenges of hPSC-based cell therapy for PD.
Collapse
Affiliation(s)
- Young Cha
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Tae-Yoon Park
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Pierre Leblanc
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Kwang-Soo Kim
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| |
Collapse
|
26
|
McGinley LM, Chen KS, Mason SN, Rigan DM, Kwentus JF, Hayes JM, Glass ED, Reynolds EL, Murphy GG, Feldman EL. Monoclonal antibody-mediated immunosuppression enables long-term survival of transplanted human neural stem cells in mouse brain. Clin Transl Med 2022; 12:e1046. [PMID: 36101963 PMCID: PMC9471059 DOI: 10.1002/ctm2.1046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/14/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND As the field of stem cell therapy advances, it is important to develop reliable methods to overcome host immune responses in animal models. This ensures survival of transplanted human stem cell grafts and enables predictive efficacy testing. Immunosuppressive drugs derived from clinical protocols are frequently used but are often inconsistent and associated with toxic side effects. Here, using a molecular imaging approach, we show that immunosuppression targeting costimulatory molecules CD4 and CD40L enables robust survival of human xenografts in mouse brain, as compared to conventional tacrolimus and mycophenolate mofetil. METHODS Human neural stem cells were modified to express green fluorescent protein and firefly luciferase. Cells were implanted in the fimbria fornix of the hippocampus and viability assessed by non-invasive bioluminescent imaging. Cell survival was assessed using traditional pharmacologic immunosuppression as compared to monoclonal antibodies directed against CD4 and CD40L. This paradigm was also implemented in a transgenic Alzheimer's disease mouse model. RESULTS Graft rejection occurs within 7 days in non-immunosuppressed mice and within 14 days in mice on a traditional regimen. The addition of dual monoclonal antibody immunosuppression extends graft survival past 7 weeks (p < .001) on initial studies. We confirm dual monoclonal antibody treatment is superior to either antibody alone (p < .001). Finally, we demonstrate robust xenograft survival at multiple cell doses up to 6 months in both C57BL/6J mice and a transgenic Alzheimer's disease model (p < .001). The dual monoclonal antibody protocol demonstrated no significant adverse effects, as determined by complete blood counts and toxicity screen. CONCLUSIONS This study demonstrates an effective immunosuppression protocol for preclinical testing of stem cell therapies. A transition towards antibody-based strategies may be advantageous by enabling stem cell survival in preclinical studies that could inform future clinical trials.
Collapse
Affiliation(s)
- Lisa M. McGinley
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Kevin S. Chen
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Shayna N. Mason
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Diana M. Rigan
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - John M. Hayes
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Emily D. Glass
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Michigan Neuroscience InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Evan L. Reynolds
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| | - Geoffrey G. Murphy
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
- Michigan Neuroscience InstituteUniversity of MichiganAnn ArborMichiganUSA
| | - Eva L. Feldman
- Department of NeurologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
27
|
McComish SF, MacMahon Copas AN, Caldwell MA. Human Brain-Based Models Provide a Powerful Tool for the Advancement of Parkinson’s Disease Research and Therapeutic Development. Front Neurosci 2022; 16:851058. [PMID: 35651633 PMCID: PMC9149087 DOI: 10.3389/fnins.2022.851058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease and affects approximately 2–3% of the population over the age of 65. PD is characterised by the loss of dopaminergic neurons from the substantia nigra, leading to debilitating motor symptoms including bradykinesia, tremor, rigidity, and postural instability. PD also results in a host of non-motor symptoms such as cognitive decline, sleep disturbances and depression. Although existing therapies can successfully manage some motor symptoms for several years, there is still no means to halt progression of this severely debilitating disorder. Animal models used to replicate aspects of PD have contributed greatly to our current understanding but do not fully replicate pathological mechanisms as they occur in patients. Because of this, there is now great interest in the use of human brain-based models to help further our understanding of disease processes. Human brain-based models include those derived from embryonic stem cells, patient-derived induced neurons, induced pluripotent stem cells and brain organoids, as well as post-mortem tissue. These models facilitate in vitro analysis of disease mechanisms and it is hoped they will help bridge the existing gap between bench and bedside. This review will discuss the various human brain-based models utilised in PD research today and highlight some of the key breakthroughs they have facilitated. Furthermore, the potential caveats associated with the use of human brain-based models will be detailed.
Collapse
Affiliation(s)
- Sarah F. McComish
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Adina N. MacMahon Copas
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Maeve A. Caldwell
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Maeve A. Caldwell,
| |
Collapse
|
28
|
Monsour M, Ebedes D, Borlongan CV. A review of the pathology and treatment of TBI and PTSD. Exp Neurol 2022; 351:114009. [PMID: 35150737 DOI: 10.1016/j.expneurol.2022.114009] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/25/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023]
Abstract
This literature review focuses on the underlying pathophysiology of TBI and PTSD symptoms, while also examining the plethora of stem cell treatment options to ameliorate these neuronal and functional changes. As more veterans return suffering from TBI and/or PTSD, it is vital that researchers discover novel therapies to mitigate the detrimental symptoms of both diagnoses. A variety of stem cell treatments have been studied and offer hopeful options for TBI and PTSD recovery.
Collapse
Affiliation(s)
- Molly Monsour
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Dominique Ebedes
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Cesario V Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA.
| |
Collapse
|
29
|
Wang G, Wu HL, Liu YP, Yan DQ, Yuan ZL, Chen L, Yang Q, Gao YS, Diao B. Pre-clinical study of human umbilical cord mesenchymal stem cell transplantation for the treatment of traumatic brain injury: safety evaluation from immunogenic and oncogenic perspectives. Neural Regen Res 2022; 17:354-361. [PMID: 34269210 PMCID: PMC8463980 DOI: 10.4103/1673-5374.317985] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Stem cell therapy is a promising strategy for the treatment of traumatic brain injury (TBI). However, animal experiments are needed to evaluate safety; in particular, to examine the immunogenicity and tumorigenicity of human umbilical cord mesenchymal stem cells (huMSCs) before clinical application. In this study, huMSCs were harvested from human amniotic membrane and umbilical cord vascular tissue. A rat model of TBI was established using the controlled cortical impact method. Starting from the third day after injury, the rats were injected with 10 μL of 5 × 106/mL huMSCs by cerebral stereotaxis or with 500 μL of 1 × 106/mL huMSCs via the tail vein for 3 successive days. huMSC transplantation decreased the serum levels of proinflammatory cytokines in rats with TBI and increased the serum levels of anti-inflammatory cytokines, thereby exhibiting good immunoregulatory function. The transplanted huMSCs were distributed in the liver, lung and brain injury sites. No abnormal proliferation or tumorigenesis was found in these organs up to 12 months after transplantation. The transplanted huMSCs negligibly proliferated in vivo, and apoptosis was gradually observed at later stages. These findings suggest that huMSC transplantation for the treatment of traumatic brain injury displays good safety. In addition, huMSCs exhibit good immunoregulatory function, which can help prevent and reduce secondary brain injury caused by the rapid release of inflammatory factors after TBI. This study was approved by the Ethics Committee of Wuhan General Hospital of PLA (approval No. 20160054) on November 1, 2016.
Collapse
Affiliation(s)
- Gang Wang
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Hua-Ling Wu
- Department of Clinical Laboratory, The Third People's Hospital of Yongzhou, Yongzhou, Hunan Province, China
| | - Yue-Ping Liu
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - De-Qi Yan
- Department of Neurosurgery, 990th Hospital of Joint Logistic Support Troops of PLA, Zhumadian, Henan Province, China
| | - Zi-Lin Yuan
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Li Chen
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Qian Yang
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| | - Yu-Song Gao
- Department of Neurosurgery, 990th Hospital of Joint Logistic Support Troops of PLA, Zhumadian, Henan Province, China
| | - Bo Diao
- Basic Medical Laboratory, General Hospital of the Central Theater Command; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei Province, China
| |
Collapse
|
30
|
Wang N, Ji X, Wu Y, Zhou S, Peng H, Wang J, Yu S, Zhang J. The Different Molecular Code in Generation of Dopaminergic Neurons from Astrocytes and Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms222212141. [PMID: 34830023 PMCID: PMC8622032 DOI: 10.3390/ijms222212141] [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: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 11/26/2022] Open
Abstract
Transplantation of exogenous dopaminergic (DA) neurons is an alternative strategy to replenish DA neurons that have lost along the course of Parkinson’s disease (PD). From the perspective of ethical acceptation, the source limitations, and the intrinsic features of PD pathology, astrocytes (AS) and mesenchymal stem cells (MSCs) are the two promising candidates of DA induction. In the present study, we induced AS or MSCs primary culture by the combination of the classical transcription-factor cocktails Mash1, Lmx1a, and Nurr1 (MLN), the chemical cocktails (S/C/D), and the morphogens SHH, FGF8, and FGF2 (S/F8/F2); the efficiency of induction into DA neurons was further analyzed by using immunostaining against the DA neuronal markers. AS could be efficiently converted into the DA neurons in vitro by the transcriptional regulation of MLN, and the combination with S/C/D or S/F8/F2 further increased the conversion efficiency. In contrast, MSCs from umbilical cord (UC-MSCs) or adipose tissue (AD-MSCs) showed moderate TH immunoreactivity after the induction with S/F8/F2 instead of with MLN or S/C/D. Our data demonstrated that AS and MSCs held lineage-specific molecular codes on the induction into DA neurons and highlighted the unique superiority of AS in the potential of cell replacement therapy for PD.
Collapse
Affiliation(s)
- Nana Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Xingrui Ji
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Yue Wu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Zhengzhou Zhongke Institute of Biomedical Engineering and Technology, Zhengzhou 450001, China
| | - Shaocong Zhou
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Huiyu Peng
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
| | - Jingwen Wang
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Zhengzhou Zhongke Institute of Biomedical Engineering and Technology, Zhengzhou 450001, China
| | - Shuang Yu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Correspondence: (S.Y.); (J.Z.); Tel.: +86-13951103916 (S.Y.); +86-13913161723 (J.Z.)
| | - Jingzhong Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (N.W.); (X.J.); (S.Z.); (H.P.)
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China; (Y.W.); (J.W.)
- Zhengzhou Zhongke Institute of Biomedical Engineering and Technology, Zhengzhou 450001, China
- Correspondence: (S.Y.); (J.Z.); Tel.: +86-13951103916 (S.Y.); +86-13913161723 (J.Z.)
| |
Collapse
|
31
|
Jovanovich N, Habib A, Kodavali C, Edwards L, Amankulor N, Zinn PO. The Evolving Role of Induced Pluripotent Stem Cells and Cerebral Organoids in Treating and Modeling Neurosurgical Diseases. World Neurosurg 2021; 155:171-179. [PMID: 34454068 PMCID: PMC11572538 DOI: 10.1016/j.wneu.2021.08.081] [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: 07/02/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 11/20/2022]
Abstract
Over the past decade, the use of induced pluripotent stem cells (IPSCs), as both direct therapeutics and building blocks for 3D in vitro models, has exhibited exciting potential in both helping to elucidate pathogenic mechanisms and treating diseases relevant to neurosurgery. Transplantation of IPSCs is being studied in neurological injuries and diseases, such as spinal cord injury and Parkinson's disease, whose clinical manifestations stem from underlying neuronal and/or axonal degeneration. Both animal models and clinical trials have shown that IPSCs have the ability to regenerate damaged neural tissue. Such evidence makes IPSCs a potentially promising therapeutic modality for patients who suffer from these neurological injuries/diseases. In addition, the cerebral organoid, a 3D assembly of IPSC aggregates that develops heterogeneous brain regions, has become the first in vitro model to closely recapitulate the complexity of the brain extracellular matrix, a 3-dimensional network of molecules that structurally and biochemically support neighboring cells. Cerebral organoids have become an exciting prospect for modeling and testing drug susceptibility of brain tumors, such as glioblastoma and metastatic brain cancer. As patient-derived organoid models are becoming more faithful to the brain, they are becoming an increasingly accurate substitute for patient clinical trials; such patient-less trials would protect the patient from potentially ineffective drugs, and speed up trial results and optimize cost. In this review, we aim to describe the role of IPSCs and cerebral organoids in treating and modeling diseases that are relevant to neurosurgery.
Collapse
Affiliation(s)
- Nicolina Jovanovich
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ahmed Habib
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Chowdari Kodavali
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Lincoln Edwards
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Nduka Amankulor
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Pascal O Zinn
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA; Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
32
|
Noh J, Jeong J, Park S, Jin Jung K, Lee B, Kim W, Han J, Cho M, Sung DK, Ahn SY, Chang YS, Son H, Jeong EJ. Preclinical assessment of thrombin-preconditioned human Wharton's jelly-derived mesenchymal stem cells for neonatal hypoxic-ischaemic brain injury. J Cell Mol Med 2021; 25:10430-10440. [PMID: 34651412 PMCID: PMC8581315 DOI: 10.1111/jcmm.16971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/23/2021] [Accepted: 09/09/2021] [Indexed: 01/17/2023] Open
Abstract
Hypoxic-ischaemic encephalopathy (HIE) is a type of brain injury affecting approximately 1 million newborn babies per year worldwide, the only treatment for which is therapeutic hypothermia. Thrombin-preconditioned mesenchymal stem cells (MSCs) exert neuroprotective effects by enriching cargo contents and boosting exosome biogenesis, thus showing promise as a new therapeutic strategy for HIE. This study was conducted to evaluate the tissue distribution and potential toxicity of thrombin-preconditioned human Wharton's jelly-derived mesenchymal stem cells (th-hWJMSCs) in animal models before the initiation of clinical trials. We investigated the biodistribution, tumorigenicity and general toxicity of th-hWJMSCs. MSCs were administered the maximum feasible dose (1 × 105 cells/10 µL/head) once, or at lower doses into the cerebral ventricle. To support the clinical use of th-hWJMSCs for treating brain injury, preclinical safety studies were conducted in newborn Sprague-Dawley rats and BALB/c nude mice. In addition, growth parameters were evaluated to assess the impact of th-hWJMSCs on the growth of newborn babies. Our results suggest that th-hWJMSCs are non-toxic and non-tumorigenic in rodent models, survive for up to 7 days in the brain and hold potential for HIE therapy.
Collapse
Affiliation(s)
- Jung‐Ho Noh
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
- College of Veterinary MedicineChungnam National UniversityDaejeonRepublic of Korea
| | - Ji‐Seong Jeong
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Sang‐Jin Park
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Kyung Jin Jung
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Byoung‐Seok Lee
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Woo‐Jin Kim
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Ji‐Seok Han
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Min‐Kyung Cho
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Dong Kyung Sung
- Stem Cell and Regenerative Medicine InstituteSamsung Medical CenterSamsung Biomedical Research InstituteSeoulRepublic of Korea
| | - So Yoon Ahn
- Stem Cell and Regenerative Medicine InstituteSamsung Medical CenterSamsung Biomedical Research InstituteSeoulRepublic of Korea
| | - Yun Sil Chang
- Stem Cell and Regenerative Medicine InstituteSamsung Medical CenterSamsung Biomedical Research InstituteSeoulRepublic of Korea
- Department of PediatricsSamsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Hwa‐Young Son
- College of Veterinary MedicineChungnam National UniversityDaejeonRepublic of Korea
| | - Eun Ju Jeong
- Department of Toxicological Evaluation and ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| |
Collapse
|
33
|
Sun Y, Feng L, Liang L, Stacey GN, Wang C, Wang Y, Hu B. Neuronal cell-based medicines from pluripotent stem cells: Development, production, and preclinical assessment. Stem Cells Transl Med 2021; 10 Suppl 2:S31-S40. [PMID: 34724724 PMCID: PMC8560198 DOI: 10.1002/sctm.20-0522] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 04/21/2021] [Accepted: 06/06/2021] [Indexed: 12/14/2022] Open
Abstract
Brain degeneration and damage is difficult to cure due to the limited endogenous repair capability of the central nervous system. Furthermore, drug development for treatment of diseases of the central nervous system remains a major challenge. However, it now appears that using human pluripotent stem cell-derived neural cells to replace degenerating cells provides a promising cell-based medicine for rejuvenation of brain function. Accordingly, a large number of studies have carried out preclinical assessments, which have involved different neural cell types in several neurological diseases. Recent advances in animal models identify the transplantation of neural derivatives from pluripotent stem cells as a promising path toward the clinical application of cell therapies [Stem Cells Transl Med 2019;8:681-693; Drug Discov Today 2019;24:992-999; Nat Med 2019;25:1045-1053]. Some groups are moving toward clinical testing in humans. However, the difficulty in selection of valuable critical quality criteria for cell products and the lack of functional assays that could indicate suitability for clinical effect continue to hinder neural cell-based medicine development [Biologicals 2019;59:68-71]. In this review, we summarize the current status of preclinical studies progress in this area and outline the biological characteristics of neural cells that have been used in new developing clinical studies. We also discuss the requirements for translation of stem cell-derived neural cells in examples of stem cell-based clinical therapy.
Collapse
Affiliation(s)
- Yun Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People's Republic of China
| | - Lin Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Lingmin Liang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Glyn N Stacey
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- International Stem Cell Banking Initiative, Barley, Hertfordshire, UK
| | - Chaoqun Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
34
|
Nandakumar S, Shahani P, Datta I, Pal R. Interventional Strategies for Parkinson Disease: Can Neural Precursor Cells Forge a Path Ahead? ACS Chem Neurosci 2021; 12:3785-3794. [PMID: 34628850 DOI: 10.1021/acschemneuro.1c00525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neural precursor cells (NPCs), derived from pluripotent stem cells (PSCs), with their unique ability to generate multiple neuronal and glial cell types are extremely useful for understanding biological mechanisms in normal and diseased states. However, generation of specific neuronal subtypes (mature) from NPCs in large numbers adequate for cell therapy is challenging due to lack of a thorough understanding of the cues that govern their differentiation. Interestingly, neural stem cells (NSCs) themselves are in consideration for therapy given their potency to form different neural cell types, release of trophic factors, and immunomodulatory effects that confer neuroprotection. With the recent COVID-19 outbreak and its accompanying neurological indications, the immunomodulatory role of NSCs may gain additional significance in the prevention of disease progression in vulnerable populations. In this regard, small-molecule mediated NPC generation from PSCs via NSC formation has become an important strategy that ensures consistency and robustness of the process. The development of the mammalian brain occurs along the rostro-caudal axis, and the establishment of anterior identity is an early event. Wnt signaling, along with fibroblast growth factor and retinoic acid, acts as a caudalization signal. Further, the increasing amount of epigenetic data available from human fetal brain development has enhanced both our understanding of and ability to experimentally manipulate these developmental regulatory programs in vitro. However, the impact on homing and engraftment after transplantation and subsequently on therapeutic efficacy of NPCs based on their derivation strategy is not yet clear. Another formidable challenge in cell replacement therapy for neurodegenerative disorders is the mode of delivery. In this Perspective, we discuss these core ideas with insights from our preliminary studies exploring the role of PSC-derived NPCs in rat models of MPTP-induced Parkinson's disease following intranasal injections.
Collapse
Affiliation(s)
- Swapna Nandakumar
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, Karnataka, India
| | - Pradnya Shahani
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru 560029, Karnataka, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru 560029, Karnataka, India
| | - Rajarshi Pal
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, Karnataka, India
| |
Collapse
|
35
|
Fernandez-Muñoz B, Garcia-Delgado AB, Arribas-Arribas B, Sanchez-Pernaute R. Human Neural Stem Cells for Cell-Based Medicinal Products. Cells 2021; 10:2377. [PMID: 34572024 PMCID: PMC8469920 DOI: 10.3390/cells10092377] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells represent an attractive tool for the development of regenerative therapies and are being tested in clinical trials for several neurological disorders. Human neural stem cells can be isolated from the central nervous system or can be derived in vitro from pluripotent stem cells. Embryonic sources are ethically controversial and other sources are less well characterized and/or inefficient. Recently, isolation of NSC from the cerebrospinal fluid of patients with spina bifida and with intracerebroventricular hemorrhage has been reported. Direct reprogramming may become another alternative if genetic and phenotypic stability of the reprogrammed cells is ensured. Here, we discuss the advantages and disadvantages of available sources of neural stem cells for the production of cell-based therapies for clinical applications. We review available safety and efficacy clinical data and discuss scalability and quality control considerations for manufacturing clinical grade cell products for successful clinical application.
Collapse
Affiliation(s)
- Beatriz Fernandez-Muñoz
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Ana Belen Garcia-Delgado
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| | - Blanca Arribas-Arribas
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Sevilla, 41012 Sevilla, Spain
| | - Rosario Sanchez-Pernaute
- Cellular Reprogramming and Production Unit, Andalusian Network for the Design and Translation of Advanced Therapies, 41092 Sevilla, Spain; (A.B.G.-D.); (B.A.-A.)
| |
Collapse
|
36
|
Das M, Pethe P. Differential expression of retinoic acid alpha and beta receptors in neuronal progenitors generated from human embryonic stem cells in response to TTNPB (a retinoic acid mimetic). Differentiation 2021; 121:13-24. [PMID: 34419635 DOI: 10.1016/j.diff.2021.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/26/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023]
Abstract
Retinoic acid (RA), an active metabolite of vitamin A, plays a critical role in the morphogenesis and differentiation of various tissues, especially in the central nervous system. RA is the most commonly used morphogen for the differentiation of human embryonic stem cells (hESCs) into neuronal progenitor cells (NPCs), an abundant source of healthy neuronal tissues for regenerative therapy. During the differentiation process, the activity of RA is governed by the involvement of RA receptor subtypes (RAR α, β, and γ) and their isoforms in the nucleus. However, little is known about the involvement of specific RAR subtypes during neuronal differentiation in humans. It is essential to elucidate the dynamic function of different RAR subtypes and their influence on the phenotypic outcome. Here in this study, we used TTNPB, an analog and stabilized form of retinoic acid that potently and selectively activates retinoic acid receptors. Here we determined the optimum concentration of TTNPBfor the efficient generation of early NPCs from hESCs. Using the optimized concentration of -TTNPB, we found that RARα is the functionally dominant subtype and controls the RA-mediated neurogenesis of hESCs. Importantly, we also found that the RARγ subtype also played a role in neuronal differentiation. In contrast, the RARβ subtype negatively correlates with neuronal differentiation. Therefore, pharmacological inhibition of RARβ in the TTNPB-mediated differentiation process could be used as a strategy to generate a large number of NPCs in vitro. In summary, our results show that RARα and RARγ play a vital role in the TTNPB-mediated neuronal differentiation of hESCs, -whereas RARβ acts as a negative regulator.
Collapse
Affiliation(s)
- Madhurima Das
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India.
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India.
| |
Collapse
|
37
|
Gordián-Vélez WJ, Chouhan D, España RA, Chen HI, Burdick JA, Duda JE, Cullen DK. Restoring lost nigrostriatal fibers in Parkinson's disease based on clinically-inspired design criteria. Brain Res Bull 2021; 175:168-185. [PMID: 34332016 DOI: 10.1016/j.brainresbull.2021.07.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022]
Abstract
Parkinson's disease is a neurodegenerative disease affecting around 10 million people worldwide. The death of dopaminergic neurons in the substantia nigra and the axonal fibers that constitute the nigrostriatal pathway leads to a loss of dopamine in the striatum that causes the motor symptoms of this disease. Traditional treatments have focused on reducing symptoms, while therapies with human fetal or stem cell-derived neurons have centered on implanting these cells in the striatum to restore its innervation. An alternative approach is pathway reconstruction, which aims to rebuild the entire structure of neurons and axonal fibers of the nigrostriatal pathway in a way that matches its anatomy and physiology. This type of repair could be more capable of reestablishing the signaling mechanisms that ensure proper dopamine release in the striatum and regulation of other motor circuit regions in the brain. In this manuscript, we conduct a review of the literature related to pathway reconstruction as a treatment for Parkinson's disease, delve into the limitations of these studies, and propose the requisite design criteria to achieve this goal at a human scale. We then present our tissue engineering-based platform to fabricate hydrogel-encased dopaminergic axon tracts in vitro for later implantation into the brain to replace and reconstruct the pathway. These tissue-engineered nigrostriatal pathways (TE-NSPs) can be characterized and optimized for cell number and phenotype, axon growth lengths and rates, and the capacity for synaptic connectivity and dopamine release. We then show original data of advances in creating these constructs matching clinical design criteria using human iPSC-derived dopaminergic neurons and a hyaluronic acid hydrogel. We conclude with a discussion of future steps that are needed to further optimize human-scale TE-NSPs and translate them into clinical products.
Collapse
Affiliation(s)
- Wisberty J Gordián-Vélez
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Dimple Chouhan
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Rodrigo A España
- Department of Neurobiology & Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - H Isaac Chen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - Jason A Burdick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - John E Duda
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - D Kacy Cullen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States.
| |
Collapse
|
38
|
Tomishima M, Kirkeby A. Bringing Advanced Therapies for Parkinson's Disease to the Clinic: The Scientist's Perspective. JOURNAL OF PARKINSONS DISEASE 2021; 11:S135-S140. [PMID: 34250954 PMCID: PMC8543246 DOI: 10.3233/jpd-212685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
After many years of preclinical development, cell and gene therapies have advanced from research tools in the lab to clinical-grade products for patients, and today they constitute more than a quarter of all new Phase I clinical trials for Parkinson’s disease. Whereas efficacy has been convincingly proven for many of these products in preclinical models, the field is now entering a new phase where the functionality and safety of these products will need to stand the test in clinical trials. If successful, these new products can have the potential to provide patients with a one-time administered treatment which may alleviate them from daily symptomatic dopaminergic medication.
Collapse
Affiliation(s)
| | - Agnete Kirkeby
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Wallenberg Center for Molecular Medicine (WCMM) and Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
39
|
Sivandzade F, Cucullo L. Regenerative Stem Cell Therapy for Neurodegenerative Diseases: An Overview. Int J Mol Sci 2021; 22:2153. [PMID: 33671500 PMCID: PMC7926761 DOI: 10.3390/ijms22042153] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases resulting from the progressive loss of structure and/or function of neurons contribute to different paralysis degrees and loss of cognition and sensation. The lack of successful curative therapies for neurodegenerative disorders leads to a considerable burden on society and a high economic impact. Over the past 20 years, regenerative cell therapy, also known as stem cell therapy, has provided an excellent opportunity to investigate potentially powerful innovative strategies for treating neurodegenerative diseases. This is due to stem cells' capability to repair injured neuronal tissue by replacing the damaged or lost cells with differentiated cells, providing a conducive environment that is in favor of regeneration, or protecting the existing healthy neurons and glial cells from further damage. Thus, in this review, the various types of stem cells, the current knowledge of stem-cell-based therapies in neurodegenerative diseases, and the recent advances in this field are summarized. Indeed, a better understanding and further studies of stem cell technologies cause progress into realistic and efficacious treatments of neurodegenerative disorders.
Collapse
Affiliation(s)
- Farzane Sivandzade
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA;
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Luca Cucullo
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
40
|
Zhang H, Su B, Jiao L, Xu ZH, Zhang CJ, Nie J, Gao ML, Zhang YV, Jin ZB. Transplantation of GMP-grade human iPSC-derived retinal pigment epithelial cells in rodent model: the first pre-clinical study for safety and efficacy in China. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:245. [PMID: 33708872 PMCID: PMC7940887 DOI: 10.21037/atm-20-4707] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly due in large part to age-dependent atrophy of retinal pigment epithelium (RPE) cells. RPE cells form a monolayer located between the choroid and the outer segments of photoreceptors, playing multifarious roles in maintenance of visual function. Allogeneically induced pluripotent stem cell-derived RPE (iPSC-RPE or iRPE) has become a potential approach for providing an abundant source of donors for clinical cell products. Transplantation of iRPE has been proven effective in rescuing impaired retinas in Royal College of Surgeons (RCS) rats after approximately 5 to 6 weeks. Here, we explore the long-term (19 weeks) safety and efficacy of human iRPE cell transplantation in pre-clinical animal models. Methods The expression of human RPE-specific markers in iRPE cells was determined using immunofluorescence staining. For the proliferative test, Ki-67 expression was also verified by immunofluorescence and flow cytometric analysis. Then, iRPE cells were transplanted into the subretinal space of immune-deficient NOD/SCID/IL-2Rgcnull (NSG) mice to assess their safety. To evaluate whether the transplanted cells could survive and rescue visual function, we performed color fundus photography, focal electroretinogram and immunostaining after delivering iRPE cells into the subretinal space of RCS rats. Results Human iRPE cells expressed native RPE-specific markers, such as microphthalmia-associated transcription factor (MiTF), retinal pigment epithelium-specific 65-kDa protein (RPE65) and tight-junction associated structural protein (ZO-1), and their proliferative capacity (Ki-67 expression) was poor after 25 days of induction. A tumorigenicity test revealed no tumor formation or abnormal proliferation in the immunodeficient mice after subretinal injection of 5×105 iRPE cells. The transplanted iRPE cells survived for at least 19 weeks and maintained visual function for 15 weeks. Conclusions In the present study, we provided further evidence for the use of human iRPE transplantation to treat retinal degenerative disease in pre-clinical animal models. Therefore, we consider human iRPE cells a promising source of cell replacement therapy for AMD.
Collapse
Affiliation(s)
- Hang Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China.,Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Bingnan Su
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China
| | - Luyan Jiao
- Nuwacell Biotechnologies Co., Ltd, Hefei, China
| | - Ze-Hua Xu
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chang-Jun Zhang
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jinfu Nie
- Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Mei-Ling Gao
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | | | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, China.,National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
41
|
Katolikova NV, Malashicheva AB, Gainetdinov RR. Cell Replacement Therapy in Parkinson’s Disease—History of Development and Prospects for Use in Clinical Practice. Mol Biol 2021. [DOI: 10.1134/s0026893320060060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Gopurappilly R. Pluripotent Stem Cell Derived Neurons as In Vitro Models for Studying Autosomal Recessive Parkinson's Disease (ARPD): PLA2G6 and Other Gene Loci. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:115-133. [PMID: 33990932 PMCID: PMC7612166 DOI: 10.1007/5584_2021_643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative motor disorder which is largely sporadic; however, some familial forms have been identified. Genetic PD can be inherited by autosomal, dominant or recessive mutations. While the dominant mutations mirror the prototype of PD with adult-onset and L-dopa-responsive cases, autosomal recessive PD (ARPD) exhibit atypical phenotypes with additional clinical manifestations. Young-onset PD is also very common with mutations in recessive gene loci. The main genes associated with ARPD are Parkin, PINK1, DJ-1, ATP13A2, FBXO7 and PLA2G6. Calcium dyshomeostasis is a mainstay in all types of PD, be it genetic or sporadic. Intriguingly, calcium imbalances manifesting as altered Store-Operated Calcium Entry (SOCE) is suggested in PLA2G6-linked PARK 14 PD. The common pathways underlying ARPD pathology, including mitochondrial abnormalities and autophagic dysfunction, can be investigated ex vivo using induced pluripotent stem cell (iPSC) technology and are discussed here. PD pathophysiology is not faithfully replicated by animal models, and, therefore, nigral dopaminergic neurons generated from iPSC serve as improved human cellular models. With no cure to date and treatments aiming at symptomatic relief, these in vitro models derived through midbrain floor-plate induction provide a platform to understand the molecular and biochemical pathways underlying PD etiology in a patient-specific manner.
Collapse
Affiliation(s)
- Renjitha Gopurappilly
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, India.
| |
Collapse
|
43
|
Watanabe T, Yasuda S, Kusakawa S, Kuroda T, Futamura M, Ogawa M, Mochizuki H, Kikkawa E, Furukawa H, Nagaoka M, Sato Y. Multisite studies for validation and improvement of a highly efficient culture assay for detection of undifferentiated human pluripotent stem cells intermingled in cell therapy products. Cytotherapy 2020; 23:176-183. [PMID: 32978066 DOI: 10.1016/j.jcyt.2020.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND AIMS The Multisite Evaluation Study on Analytical Methods for Non-Clinical Safety Assessment of Human-Derived Regenerative Medical Products (MEASURE) is a Japanese experimental public-private partnership initiative, which aims to standardize methodology for tumorigenicity evaluation of human pluripotent stem cell (hPSC)-derived cell therapy products (CTPs). Undifferentiated hPSCs possess tumorigenic potential, and thus residual undifferentiated hPSCs are one of the major hazards for the risk of tumor formation from hPSC-derived CTPs. Among currently available assays, a highly efficient culture (HEC) assay is reported to be one of the most sensitive for the detection of residual undifferentiated hPSCs. METHODS MEASURE first validated the detection sensitivity of HEC assay and then investigated the feasibility of magnetic-activated cell sorting (MACS) to improve sensitivity. RESULTS The multisite experiments confirmed that the lower limit of detection under various conditions to which the human induced pluripotent stem cell lines and culture medium/substrate were subjected was 0.001%. In addition, MACS concentrated cells expressing undifferentiated cell markers and consequently achieved a detection sensitivity of 0.00002%. CONCLUSIONS These results indicate that HEC assay is highly sensitive and robust and that the application of MACS on this assay is a promising tool for further mitigation of the potential tumorigenicity risk of hPSC-derived CTPs.
Collapse
Affiliation(s)
- Takeshi Watanabe
- Drug Safety Research and Evaluation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan; The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan.
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Shinji Kusakawa
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| | - Mayumi Futamura
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Drug Discovery Support Division, Tsukuba Research Institute, BoZo Research Center Inc, Tsukuba, Japan
| | - Mitsuhide Ogawa
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; CMIC Bioresearch Center, CMIC Pharma Science Co, Ltd, Hokuto, Japan
| | - Hidemi Mochizuki
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Research Planning Section, Ina Research Inc, Ina-shi, Japan
| | - Eri Kikkawa
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Research Division, HEALIOS K.K., Kobe, Japan
| | - Hatsue Furukawa
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Integrated & Translational Science, Axcelead Drug Discovery Partners, Inc, Fujisawa, Japan
| | - Masato Nagaoka
- The Committee for Non-Clinical Safety Evaluation of Pluripotent Stem Cell-Derived Product, Forum for Innovative Regenerative Medicine, Tokyo, Japan; Life Science Research Laboratory, Tosoh Corporation, Ayase-shi, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
44
|
Desgres M, Menasché P. Clinical Translation of Pluripotent Stem Cell Therapies: Challenges and Considerations. Cell Stem Cell 2020; 25:594-606. [PMID: 31703770 DOI: 10.1016/j.stem.2019.10.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although the clinical outcomes of cell therapy trials have not met initial expectations, emerging evidence suggests that injury-mediated tissue damage might benefit from the delivery of cells or their secreted products. Pluripotent stem cells (PSCs) are promising cell sources primarily because of their capacity to generate stage- and lineage-specific differentiated derivatives. However, they carry inherent challenges for safe and efficacious clinical translation. This Review describes completed or ongoing trials of PSCs, discusses their potential mechanisms of action, and considers how to address the challenges required for them to become a major therapy, using heart repair as a case study.
Collapse
Affiliation(s)
- Manon Desgres
- Université de Paris, PARCC, INSERM, 75015 Paris, France
| | - Philippe Menasché
- Université de Paris, PARCC, INSERM, 75015 Paris, France; Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou 20, rue Leblanc, 75015 Paris, France.
| |
Collapse
|
45
|
McGinley LM, Willsey MS, Kashlan ON, Chen KS, Hayes JM, Bergin IL, Mason SN, Stebbins AW, Kwentus JF, Pacut C, Kollmer J, Sakowski SA, Bell CB, Chestek CA, Murphy GG, Patil PG, Feldman EL. Magnetic resonance imaging of human neural stem cells in rodent and primate brain. Stem Cells Transl Med 2020; 10:83-97. [PMID: 32841522 PMCID: PMC7780819 DOI: 10.1002/sctm.20-0126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/03/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Stem cell transplantation therapies are currently under investigation for central nervous system disorders. Although preclinical models show benefit, clinical translation is somewhat limited by the absence of reliable noninvasive methods to confirm targeting and monitor transplanted cells in vivo. Here, we assess a novel magnetic resonance imaging (MRI) contrast agent derived from magnetotactic bacteria, magneto‐endosymbionts (MEs), as a translatable methodology for in vivo tracking of stem cells after intracranial transplantation. We show that ME labeling provides robust MRI contrast without impairment of cell viability or other important therapeutic features. Labeled cells were visualized immediately post‐transplantation and over time by serial MRI in nonhuman primate and mouse brain. Postmortem tissue analysis confirmed on‐target grft location, and linear correlations were observed between MRI signal, cell engraftment, and tissue ME levels, suggesting that MEs may be useful for determining graft survival or rejection. Overall, these findings indicate that MEs are an effective tool for in vivo tracking and monitoring of cell transplantation therapies with potential relevance to many cellular therapy applications.
Collapse
Affiliation(s)
- Lisa M McGinley
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew S Willsey
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Osama N Kashlan
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Kevin S Chen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA
| | - John M Hayes
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ingrid L Bergin
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shayna N Mason
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Aaron W Stebbins
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Crystal Pacut
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer Kollmer
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Caleb B Bell
- Bell Biosystems, San Francisco, California, USA.,G4S Capital & Ikigai Accelerator, Santa Clara, California, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Electrical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Neuroscience and Robotics Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Parag G Patil
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
46
|
Salado-Manzano C, Perpiña U, Straccia M, Molina-Ruiz FJ, Cozzi E, Rosser AE, Canals JM. Is the Immunological Response a Bottleneck for Cell Therapy in Neurodegenerative Diseases? Front Cell Neurosci 2020; 14:250. [PMID: 32848630 PMCID: PMC7433375 DOI: 10.3389/fncel.2020.00250] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders such as Parkinson's (PD) and Huntington's disease (HD) are characterized by a selective detrimental impact on neurons in a specific brain area. Currently, these diseases have no cures, although some promising trials of therapies that may be able to slow the loss of brain cells are underway. Cell therapy is distinguished by its potential to replace cells to compensate for those lost to the degenerative process and has shown a great potential to replace degenerated neurons in animal models and in clinical trials in PD and HD patients. Fetal-derived neural progenitor cells, embryonic stem cells or induced pluripotent stem cells are the main cell sources that have been tested in cell therapy approaches. Furthermore, new strategies are emerging, such as the use of adult stem cells, encapsulated cell lines releasing trophic factors or cell-free products, containing an enriched secretome, which have shown beneficial preclinical outcomes. One of the major challenges for these potential new treatments is to overcome the host immune response to the transplanted cells. Immune rejection can cause significant alterations in transplanted and endogenous tissue and requires immunosuppressive drugs that may produce adverse effects. T-, B-lymphocytes and microglia have been recognized as the main effectors in striatal graft rejection. This review aims to summarize the preclinical and clinical studies of cell therapies in PD and HD. In addition, the precautions and strategies to ensure the highest quality of cell grafts, the lowest risk during transplantation and the reduction of a possible immune rejection will be outlined. Altogether, the wide-ranging possibilities of advanced therapy medicinal products (ATMPs) could make therapeutic treatment of these incurable diseases possible in the near future.
Collapse
Affiliation(s)
- Cristina Salado-Manzano
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Unai Perpiña
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Francisco J. Molina-Ruiz
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Emanuele Cozzi
- Department of Cardio-Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
- Transplant Immunology Unit, Padua University Hospital, Padua, Italy
| | - Anne E. Rosser
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Josep M. Canals
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| |
Collapse
|
47
|
Pre-clinical study of induced pluripotent stem cell-derived dopaminergic progenitor cells for Parkinson's disease. Nat Commun 2020; 11:3369. [PMID: 32632153 PMCID: PMC7338530 DOI: 10.1038/s41467-020-17165-w] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/16/2020] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived dopaminergic (DA) neurons are an expected source for cell-based therapies for Parkinson’s disease (PD). The regulatory criteria for the clinical application of these therapies, however, have not been established. Here we show the results of our pre-clinical study, in which we evaluate the safety and efficacy of dopaminergic progenitors (DAPs) derived from a clinical-grade human iPSC line. We confirm the characteristics of DAPs by in vitro analyses. We also verify that the DAP population include no residual undifferentiated iPSCs or early neural stem cells and have no genetic aberration in cancer-related genes. Furthermore, in vivo studies using immunodeficient mice reveal no tumorigenicity or toxicity of the cells. When the DAPs are transplanted into the striatum of 6-OHDA-lesioned rats, the animals show behavioral improvement. Based on these results, we started a clinical trial to treat PD patients in 2018. Induced pluripotent stem cell (iPSC) derived dopaminergic neurons are a promising source for cell-based Parkinson’s disease (PD) therapy. Here the authors report a comprehensive pre-clinical evaluation of the safety and efficacy of dopaminergic progenitors derived from a clinical-grade human iPSC line.
Collapse
|
48
|
Cardiac cell therapy: Current status, challenges and perspectives. Arch Cardiovasc Dis 2020; 113:285-292. [PMID: 32171698 DOI: 10.1016/j.acvd.2020.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/08/2020] [Indexed: 12/28/2022]
Abstract
Although the initial clinical trials of cardiac cell therapy have failed to demonstrate unequivocal clinical benefits, the accumulation of preclinical data gathered in parallel can now help us to understand the main causes of failures, while providing mechanistic insights that may be leveraged to improve the outcomes of subsequent clinical studies using cells or their secreted products. This review briefly describes the current status of clinical trials, discusses the potential mechanisms of action of the grafted cells, and the impact of this knowledge on the design of future studies, and finally draws some perspectives.
Collapse
|
49
|
Desgres M, Menasché P. [Pluripotent stem cells for the treatment of heart failure: current status, persisting issues and perspectives]. Med Sci (Paris) 2019; 35:771-778. [PMID: 31625899 DOI: 10.1051/medsci/2019155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Although the first wave of cell therapy trials has not commonly yielded clinically meaningful improvements, some encouraging hints have emerged which suggest that stem cells or their secreted products could ultimately find a place within the armamentarium of therapies that can be offered to patients with heart failure. In this setting, pluripotent stem cells raise a particular interest because of their unique ability to generate lineage-specific cells which can be transplanted at the desired stage of differentiation. This review discusses the current status of research in this field, the persisting roadblocks that need to be overcome and the approaches which might hasten the clinical applications of this cell type.
Collapse
Affiliation(s)
- Manon Desgres
- Université de Paris, PARCC, Inserm, F-75015 Paris, France
| | - Philippe Menasché
- Université de Paris, PARCC, Inserm, F-75015 Paris, France - Département de chirurgie cardio-vasculaire, Hôpital Européen Georges Pompidou, 20 rue Leblanc, 75015 Paris, France
| |
Collapse
|
50
|
Garitaonandia I, Gonzalez R, Sherman G, Semechkin A, Evans A, Kern R. Novel Approach to Stem Cell Therapy in Parkinson's Disease. Stem Cells Dev 2019; 27:951-957. [PMID: 29882481 DOI: 10.1089/scd.2018.0001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In this commentary we discuss International Stem Cell Corporation's (ISCO's) approach to developing a pluripotent stem cell based treatment for Parkinson's disease (PD). In 2016, ISCO received approval to conduct the world's first clinical study of a pluripotent stem cell based therapy for PD. The Australian regulatory agency Therapeutic Goods Administration (TGA) and the Melbourne Health's Human Research Ethics Committee (HREC) independently reviewed ISCO's extensive preclinical data and granted approval for the evaluation of a novel human parthenogenetic derived neural stem cell (NSC) line, ISC-hpNSC, in a PD phase 1 clinical trial ( ClinicalTrials.gov NCT02452723). This is a single-center, open label, dose escalating 12-month study with a 5-year follow-up evaluating a number of objective and patient-reported safety and efficacy measures. A total of 6 years of safety and efficacy data will be collected from each patient. Twelve participants are recruited in this study with four participants per single dose cohort of 30, 50, and 70 million ISC-hpNSC. The grafts are placed bilaterally in the caudate nucleus, putamen, and substantia nigra by magnetic resonance imaging-guided stereotactic surgery. Participants are 30-70 years old with idiopathic PD ≤13 years duration and unified PD rating scale motor score (Part III) in the "OFF" state ≤49. This trial is fully funded by ISCO with no economic involvement from the patients. It is worth noting that ISCO underwent an exhaustive review process and successfully answered the very comprehensive, detailed, and specific questions posed by the TGA and HREC. The regulatory/ethic review process is based on applying scientific and clinical expertise to decision-making, to ensure that the benefits to consumers outweigh any risks associated with the use of medicines or novel therapies.
Collapse
Affiliation(s)
| | | | - Glenn Sherman
- 1 International Stem Cell Corporation , Carlsbad, California
| | | | - Andrew Evans
- 2 Royal Melbourne Hospital , Parkville, Australia
| | - Russell Kern
- 1 International Stem Cell Corporation , Carlsbad, California.,3 Cyto Therapeutics , Melbourne, Australia
| |
Collapse
|