1
|
Lindvall O. History of cellular grafting for central nervous system repair-A clinical perspective. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:15-40. [PMID: 39341652 DOI: 10.1016/b978-0-323-90120-8.00011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
As late as in the 1970s, the evidence supporting that brain function might be restored by replacing dead cells by transplantation of new healthy cells was scarce in experimental animals and lacking in humans. Repairing the human brain was regarded as completely unrealistic by clinicians. Fifty years later, the situation is very different, and cellular grafting has reached patient application in several conditions affecting the CNS. The clinical studies performed so far have shown that cellular grafts can survive, grow, and function also in the diseased adult human brain. However, no proven treatment based on cell transplantation is currently available for any brain disorder. Here, the history of cellular grafting is described from a clinical perspective, including some of the preclinical work that has formed the basis for its translation to patient application. The focus is on cell transplantation for Parkinson disease, which in many ways is paving the way for this field of research. The chapter gives an account of the scientific milestones, the ups and downs, as well as the positive and negative reactions from the scientific and clinical community, and how this research field despite many obstacles has continued to move forward over more than four decades.
Collapse
Affiliation(s)
- Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden; Division of Neurology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
2
|
Maheshwari S, Akram H, Bulstrode H, Kalia SK, Morizane A, Takahashi J, Natalwala A. Dopaminergic Cell Replacement for Parkinson's Disease: Addressing the Intracranial Delivery Hurdle. JOURNAL OF PARKINSON'S DISEASE 2024; 14:415-435. [PMID: 38457149 PMCID: PMC11091588 DOI: 10.3233/jpd-230328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/16/2024] [Indexed: 03/09/2024]
Abstract
Parkinson's disease (PD) is an increasingly prevalent neurological disorder, affecting more than 8.5 million individuals worldwide. α-Synucleinopathy in PD is considered to cause dopaminergic neuronal loss in the substantia nigra, resulting in characteristic motor dysfunction that is the target for current medical and surgical therapies. Standard treatment for PD has remained unchanged for several decades and does not alter disease progression. Furthermore, symptomatic therapies for PD are limited by issues surrounding long-term efficacy and side effects. Cell replacement therapy (CRT) presents an alternative approach that has the potential to restore striatal dopaminergic input and ameliorate debilitating motor symptoms in PD. Despite promising pre-clinical data, CRT has demonstrated mixed success clinically. Recent advances in graft biology have renewed interest in the field, resulting in several worldwide ongoing clinical trials. However, factors surrounding the effective neurosurgical delivery of cell grafts have remained under-studied, despite their significant potential to influence therapeutic outcomes. Here, we focus on the key neurosurgical factors to consider for the clinical translation of CRT. We review the instruments that have been used for cell graft delivery, highlighting current features and limitations, while discussing how future devices could address these challenges. Finally, we review other novel developments that may enhance graft accessibility, delivery, and efficacy. Challenges surrounding neurosurgical delivery may critically contribute to the success of CRT, so it is crucial that we address these issues to ensure that CRT does not falter at the final hurdle.
Collapse
Affiliation(s)
- Saumya Maheshwari
- The Medical School, University of Edinburgh, Edinburgh BioQuarter, UK
| | - Harith Akram
- Unit of Functional Neurosurgery, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Harry Bulstrode
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, Division of Academic Neurosurgery, University of Cambridge, Cambridge, UK
| | - Suneil K. Kalia
- Division of Neurosurgery, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Asuka Morizane
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Regenerative Medicine, Center for Clinical Research and Innovation, Kobe City Medical Center General Hospital, Hyogo, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ammar Natalwala
- Unit of Functional Neurosurgery, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
- Department for Neuromuscular Diseases, Institute of Neurology, University College London, London, UK
| |
Collapse
|
3
|
Wang F, Sun Z, Peng D, Gianchandani S, Le W, Boltze J, Li S. Cell-therapy for Parkinson's disease: a systematic review and meta-analysis. J Transl Med 2023; 21:601. [PMID: 37679754 PMCID: PMC10483810 DOI: 10.1186/s12967-023-04484-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Cell-based strategies focusing on replacement or protection of dopaminergic neurons have been considered as a potential approach to treat Parkinson's disease (PD) for decades. However, despite promising preclinical results, clinical trials on cell-therapy for PD reported mixed outcomes and a thorough synthesis of these findings is lacking. We performed a systematic review and meta-analysis to evaluate cell-therapy for PD patients. METHODS We systematically identified all clinical trials investigating cell- or tissue-based therapies for PD published before July 2023. Out of those, studies reporting transplantation of homogenous cells (containing one cell type) were included in meta-analysis. The mean difference or standardized mean difference in quantitative neurological scale scores before and after cell-therapy was analyzed to evaluate treatment effects. RESULTS The systematic literature search revealed 106 articles. Eleven studies reporting data from 11 independent trials (210 patients) were eligible for meta-analysis. Disease severity and motor function evaluation indicated beneficial effects of homogenous cell-therapy in the 'off' state at 3-, 6-, 12-, or 24-month follow-ups, and for motor function even after 36 months. Most of the patients were levodopa responders (61.6-100% in different follow-ups). Cell-therapy was also effective in improving the daily living activities in the 'off' state of PD patients. Cells from diverse sources were used and multiple transplantation modes were applied. Autografts did not improve functional outcomes, while allografts exhibited beneficial effects. Encouragingly, both transplantation into basal ganglia and to areas outside the basal ganglia were effective to reduce disease severity. Some trials reported adverse events potentially related to the surgical procedure. One confirmed and four possible cases of graft-induced dyskinesia were reported in two trials included in this meta-analysis. CONCLUSIONS This meta-analysis provides preliminary evidence for the beneficial effects of homogenous cell-therapy for PD, potentially to the levodopa responders. Allogeneic cells were superior to autologous cells, and the effective transplantation sites are not limited to the basal ganglia. PROSPERO registration number: CRD42022369760.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Zhengwu Sun
- Department of Clinical Pharmacy, Central Hospital of Dalian University of Technology, Dalian, China
| | - Daoyong Peng
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Shikha Gianchandani
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Weidong Le
- Institute of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial Hospital, Chengdu, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Beijing, 100038, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Barker RA, Björklund A. Restorative cell and gene therapies for Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:211-226. [PMID: 36803812 DOI: 10.1016/b978-0-323-85555-6.00012-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
One of the core pathological features of Parkinson's disease (PD) is the loss of the dopaminergic nigrostriatal pathway which lies at the heart of many of the motor features of this condition as well as some of the cognitive problems. The importance of this pathological event is evident through the clinical benefits that are seen when patients with PD are treated with dopaminergic agents, at least in early-stage disease. However, these agents create problems of their own through stimulation of more intact dopaminergic networks within the central nervous system causing major neuropsychiatric problems including dopamine dysregulation. In addition, over time the nonphysiological stimulation of striatal dopamine receptors by l-dopa containing drugs leads to the genesis of l-dopa-induced dyskinesias that can become very disabling in many cases. As such, there has been much interest in trying to better reconstitute the dopaminergic nigrostriatal pathway using either factors to regrow it, cells to replace it, or gene therapies to restore dopamine transmission in the striatum. In this chapter, we lay out the rationale, history and current status of these different therapies as well as highlighting where the field is heading and what new interventions might come to clinic in the coming years.
Collapse
Affiliation(s)
- Roger A Barker
- Department of Clinical Neuroscience, Cambridge Centre for Brain Repair, Cambridge, United Kingdom.
| | - Anders Björklund
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
5
|
Xue J, Wu Y, Bao Y, Zhao M, Li F, Sun J, Sun Y, Wang J, Chen L, Mao Y, Schweitzer JS, Song B. Clinical considerations in Parkinson's disease cell therapy. Ageing Res Rev 2023; 83:101792. [PMID: 36402405 DOI: 10.1016/j.arr.2022.101792] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/13/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Cell replacement therapy is an area of increasing interest for treating Parkinson's disease (PD). However, to become a clinically practical option for PD patients, it must first overcome significant barriers, including establishment of safe and standardized surgical procedures, determination of appropriate perioperative medication regimens, demonstration of long-term graft survival and incorporation, and standardized, clinically meaningful follow-up measures. In this review, we will describe the current status of cell therapy for PD with special attention to these critical requirements, to define guideposts on the road to bring the benefit of this therapy to the Parkinson's clinic.
Collapse
Affiliation(s)
- Jun Xue
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Yifan Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Yuting Bao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Minglai Zhao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Fangzhou Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Jing Sun
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yimin Sun
- Institute of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jian Wang
- Institute of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai Clinical Medical Center of Neurosurgery, Shanghai 200040, China.
| | - Jeffrey S Schweitzer
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Bin Song
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China; Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
6
|
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]
|
7
|
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
|
8
|
Schweitzer JS, Song B, Herrington TM, Park TY, Lee N, Ko S, Jeon J, Cha Y, Kim K, Li Q, Henchcliffe C, Kaplitt M, Neff C, Rapalino O, Seo H, Lee IH, Kim J, Kim T, Petsko GA, Ritz J, Cohen BM, Kong SW, Leblanc P, Carter BS, Kim KS. Personalized iPSC-Derived Dopamine Progenitor Cells for Parkinson's Disease. N Engl J Med 2020; 382:1926-1932. [PMID: 32402162 PMCID: PMC7288982 DOI: 10.1056/nejmoa1915872] [Citation(s) in RCA: 344] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We report the implantation of patient-derived midbrain dopaminergic progenitor cells, differentiated in vitro from autologous induced pluripotent stem cells (iPSCs), in a patient with idiopathic Parkinson's disease. The patient-specific progenitor cells were produced under Good Manufacturing Practice conditions and characterized as having the phenotypic properties of substantia nigra pars compacta neurons; testing in a humanized mouse model (involving peripheral-blood mononuclear cells) indicated an absence of immunogenicity to these cells. The cells were implanted into the putamen (left hemisphere followed by right hemisphere, 6 months apart) of a patient with Parkinson's disease, without the need for immunosuppression. Positron-emission tomography with the use of fluorine-18-L-dihydroxyphenylalanine suggested graft survival. Clinical measures of symptoms of Parkinson's disease after surgery stabilized or improved at 18 to 24 months after implantation. (Funded by the National Institutes of Health and others.).
Collapse
Affiliation(s)
- Jeffrey S Schweitzer
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Bin Song
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Todd M Herrington
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Tae-Yoon Park
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Nayeon Lee
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Sanghyeok Ko
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Jeha Jeon
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Young Cha
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Kyungsang Kim
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Quanzheng Li
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Claire Henchcliffe
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Michael Kaplitt
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Carolyn Neff
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Otto Rapalino
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Hyemyung Seo
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - In-Hee Lee
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Jisun Kim
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Taewoo Kim
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Gregory A Petsko
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Jerome Ritz
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Bruce M Cohen
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Sek-Won Kong
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Pierre Leblanc
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Bob S Carter
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| | - Kwang-Soo Kim
- From the Departments of Neurosurgery (J.S.S., B.S.C.), Neurology (T.M.H.), and Radiology (K.K., Q.L.), the Gordon Center for Medical Imaging (K.K., Q.L.), and the Division of Neuroradiology (O.R.), Massachusetts General Hospital, the Department of Pediatrics, Computational Health Informatics Program, Boston Children's Hospital (I.-H.L., S.-W.K.), and the Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (J.R.), Boston, and the Department of Psychiatry (B.M.C.) and the Molecular Neurobiology Laboratory (B.S., T.-Y.P., N.L., S.K., J.J., Y.C., H.S., J.K., T.K., P.L., K.-S.K.), McLean Hospital, Belmont - all in Massachusetts; the Departments of Neurology (C.H.) and Neurosurgery (M.K.) and the Brain and Mind Research Institute (G.A.P.), Weill Cornell Medical College, New York; the Department of Neurology, Kaiser Permanente, Irvine, CA (C.N.); and the Department of Molecular and Life Sciences, Hanyang University, Seoul, South Korea (H.S.)
| |
Collapse
|
9
|
Jamebozorgi K, Taghizadeh E, Rostami D, Pormasoumi H, Barreto GE, Hayat SMG, Sahebkar A. Cellular and Molecular Aspects of Parkinson Treatment: Future Therapeutic Perspectives. Mol Neurobiol 2019; 56:4799-4811. [PMID: 30397850 DOI: 10.1007/s12035-018-1419-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder accompanied by depletion of dopamine and loss of dopaminergic neurons in the brain that is believed to be responsible for the motor and non-motor symptoms in this disease. The main drug prescribed for Parkinsonian patients is L-dopa, which can be converted to dopamine by passing through the blood-brain barrier. Although L-dopa is able to improve motor function and improve the quality of life in the patients, there is inter-individual variability and some patients do not achieve the therapeutic effect. Variations in treatment response and side effects of current drugs have convinced scientists to think of treating Parkinson's disease at the cellular and molecular level. Molecular and cellular therapy for Parkinson's disease include (i) cell transplantation therapy with human embryonic stem (ES) cells, human induced pluripotent stem (iPS) cells and human fetal mesencephalic tissue, (ii) immunological and inflammatory therapy which is done using antibodies, and (iii) gene therapy with AADC-TH-GCH gene therapy, viral vector-mediated gene delivery, RNA interference-based therapy, CRISPR-Cas9 gene editing system, and alternative methods such as optogenetics and chemogenetics. Although these methods currently have a series of challenges, they seem to be promising techniques for Parkinson's treatment in future. In this study, these prospective therapeutic approaches are reviewed.
Collapse
Affiliation(s)
| | - Eskandar Taghizadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
- Departments of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Daryoush Rostami
- Department of School Allied, Zabol University of Medical Sciences, Zabol, Iran
| | - Hosein Pormasoumi
- Faculty of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, P.O. Box: 91779-48564, Mashhad, Iran.
| |
Collapse
|
10
|
Stoker TB, Torsney KM, Barker RA. Emerging Treatment Approaches for Parkinson's Disease. Front Neurosci 2018; 12:693. [PMID: 30349448 PMCID: PMC6186796 DOI: 10.3389/fnins.2018.00693] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease, manifesting as a characteristic movement disorder with a number of additional non-motor features. The pathological hallmark of PD is the presence of intra-neuronal aggregates of α-synuclein (Lewy bodies). The movement disorder of PD occurs largely due to loss of dopaminergic neurons of the substantia nigra, resulting in striatal dopamine depletion. There are currently no proven disease modifying treatments for PD, with management options consisting mainly of dopaminergic drugs, and in a limited number of patients, deep brain stimulation. Long-term use of established dopaminergic therapies for PD results in significant adverse effects, and there is therefore a requirement to develop better means of restoring striatal dopamine, as well as treatments that are able to slow progression of the disease. A number of exciting treatments have yielded promising results in pre-clinical and early clinical trials, and it now seems likely that the landscape for the management of PD will change dramatically in the short to medium term future. Here, we discuss the promising regenerative cell-based and gene therapies, designed to treat the dopaminergic aspects of PD whilst limiting adverse effects, as well as novel approaches to reducing α-synuclein pathology.
Collapse
Affiliation(s)
- Thomas B Stoker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Kelli M Torsney
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Department of Medicine for the Elderly, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
11
|
Sonntag KC, Song B, Lee N, Jung JH, Cha Y, Leblanc P, Neff C, Kong SW, Carter BS, Schweitzer J, Kim KS. Pluripotent stem cell-based therapy for Parkinson's disease: Current status and future prospects. Prog Neurobiol 2018; 168:1-20. [PMID: 29653250 PMCID: PMC6077089 DOI: 10.1016/j.pneurobio.2018.04.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 03/13/2018] [Accepted: 04/05/2018] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative disorders, which affects about 0.3% of the general population. As the population in the developed world ages, this creates an escalating burden on society both in economic terms and in quality of life for these patients and for the families that support them. Although currently available pharmacological or surgical treatments may significantly improve the quality of life of many patients with PD, these are symptomatic treatments that do not slow or stop the progressive course of the disease. Because motor impairments in PD largely result from loss of midbrain dopamine neurons in the substantia nigra pars compacta, PD has long been considered to be one of the most promising target diseases for cell-based therapy. Indeed, numerous clinical and preclinical 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 as a standardized therapeutic regimen has been fraught with fundamental ethical, practical, and clinical issues, prompting scientists to explore alternative cell sources. Based on groundbreaking establishments of human embryonic stem cells and induced pluripotent stem cells, these human pluripotent stem cells have been the subject of extensive research, leading to tremendous advancement in our understanding of these novel classes of stem cells and promising great potential for regenerative medicine. In this review, we discuss the prospects and challenges of human pluripotent stem cell-based cell therapy for PD.
Collapse
Affiliation(s)
- Kai-C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Laboratory for Translational Research on Neurodegeneration, 115 Mill Street, Belmont, MA, 02478, United States; Program for Neuropsychiatric Research, 115 Mill Street, Belmont, MA, 02478, United States
| | - Bin Song
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Nayeon Lee
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Jin Hyuk Jung
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Young Cha
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Pierre Leblanc
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States
| | - Carolyn Neff
- Kaiser Permanente Medical Group, Irvine, CA, 92618, United States
| | - Sek Won Kong
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, United States; Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, 02115, United States
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, 02114, United States
| | - Jeffrey Schweitzer
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, 02114, United States.
| | - Kwang-Soo Kim
- Department of Psychiatry, McLean Hospital, Harvard Medical School, United States; Molecular Neurobiology Laboratory, Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, MA, 02478, United States.
| |
Collapse
|
12
|
Corey S, Ghanekar S, Sokol J, Zhang JH, Borlongan CV. An update on stem cell therapy for neurological disorders: cell death pathways as therapeutic targets. Chin Neurosurg J 2017. [DOI: 10.1186/s41016-016-0071-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
13
|
Torres N, Molet J, Moro C, Mitrofanis J, Benabid AL. Neuroprotective Surgical Strategies in Parkinson's Disease: Role of Preclinical Data. Int J Mol Sci 2017; 18:ijms18102190. [PMID: 29053638 PMCID: PMC5666871 DOI: 10.3390/ijms18102190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/18/2022] Open
Abstract
Although there have been many pharmacological agents considered to be neuroprotective therapy in Parkinson's disease (PD) patients, neurosurgical approaches aimed to neuroprotect or restore the degenerative nigrostriatal system have rarely been the focus of in depth reviews. Here, we explore the neuroprotective strategies involving invasive surgical approaches (NSI) using neurotoxic models 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA), which have led to clinical trials. We focus on several NSI approaches, namely deep brain stimulation of the subthalamic nucleus, glial neurotrophic derived factor (GDNF) administration and cell grafting methods. Although most of these interventions have produced positive results in preclinical animal models, either from behavioral or histological studies, they have generally failed to pass randomized clinical trials to validate each approach. We argue that NSI are promising approaches for neurorestoration in PD, but preclinical studies should be planned carefully in order not only to detect benefits but also to detect potential adverse effects. Further, clinical trials should be designed to be able to detect and disentangle neuroprotection from symptomatic effects. In summary, our review study evaluates the pertinence of preclinical models to study NSI for PD and how this affects their efficacy when translated into clinical trials.
Collapse
Affiliation(s)
- Napoleon Torres
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - Jenny Molet
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - Cecile Moro
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - John Mitrofanis
- Department of Anatomy, University of Sydney; Sydney Medical School, Sydney NSW 2006, Australia.
| | - Alim Louis Benabid
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| |
Collapse
|
14
|
Wianny F, Vezoli J. Transplantation in the nonhuman primate MPTP model of Parkinson's disease: update and perspectives. Primate Biol 2017; 4:185-213. [PMID: 32110706 PMCID: PMC7041537 DOI: 10.5194/pb-4-185-2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/31/2017] [Indexed: 12/22/2022] Open
Abstract
In order to calibrate stem cell exploitation for cellular therapy in neurodegenerative diseases, fundamental and preclinical research in NHP (nonhuman primate) models is crucial. Indeed, it is consensually recognized that it is not possible to directly extrapolate results obtained in rodent models to human patients. A large diversity of neurological pathologies should benefit from cellular therapy based on neural differentiation of stem cells. In the context of this special issue of Primate Biology on NHP stem cells, we describe past and recent advances on cell replacement in the NHP model of Parkinson's disease (PD). From the different grafting procedures to the various cell types transplanted, we review here diverse approaches for cell-replacement therapy and their related therapeutic potential on behavior and function in the NHP model of PD.
Collapse
Affiliation(s)
- Florence Wianny
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Julien Vezoli
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, 60528 Frankfurt, Germany
| |
Collapse
|
15
|
Boronat-García A, Guerra-Crespo M, Drucker-Colín R. Historical perspective of cell transplantation in Parkinson’s disease. World J Transplant 2017; 7:179-192. [PMID: 28698835 PMCID: PMC5487308 DOI: 10.5500/wjt.v7.i3.179] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/27/2017] [Accepted: 05/15/2017] [Indexed: 02/05/2023] Open
Abstract
Cell grafting has been considered a therapeutic approach for Parkinson’s disease (PD) since the 1980s. The classical motor symptoms of PD are caused by the loss of dopaminergic neurons in the substantia nigra pars compacta, leading to a decrement in dopamine release in the striatum. Consequently, the therapy of cell-transplantation for PD consists in grafting dopamine-producing cells directly into the brain to reestablish dopamine levels. Different cell sources have been shown to induce functional benefits on both animal models of PD and human patients. However, the observed motor improvements are highly variable between individual subjects, and the sources of this variability are not fully understood. The purpose of this review is to provide a general overview of the pioneering studies done in animal models of PD that established the basis for the first clinical trials in humans, and compare these with the latest findings to identify the most relevant aspects that remain unanswered to date. The main focus of the discussions presented here will be on the mechanisms associated with the survival and functionality of the transplants. These include the role of the dopamine released by the grafts and the capacity of the grafted cells to extend fibers and to integrate into the motor circuit. The complete understanding of these aspects will require extensive research on basic aspects of molecular and cellular physiology, together with neuronal network function, in order to uncover the real potential of cell grafting for treating PD.
Collapse
|
16
|
Kordower JH, Liu YT, Winn S, Emerich DF. Encapsulated PC 12 Cell Transplants into Hemiparkinsonian Monkeys: A Behavioral, Neuroanatomical, and Neurochemical Analysis. Cell Transplant 2017; 4:155-71. [PMID: 7773555 DOI: 10.1177/096368979500400203] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Four cynomolgus monkeys were trained on a hand reaching task and then rendered hemiparkinsonian with an intracarotid injection of n-methyl 4 phenyl 1,2,3,6, tetrahydropyridine (MPTP). Performance on this task with the limb contralateral to the MPTP injection was significantly impaired following the lesion. Three monkeys received implants of polymer-encapsulated containing PC12 cells into the caudate nucleus and putamen. One monkey received identical implants of empty capsules and served as a control. After a transient improvement, limb use in the control monkey dissipated and returned to post-MPTP disability. Two of the three PC12 cell grafted monkeys recovered performance on the hand reach task to near normal levels for up to 6.5 mo posttransplantation. Capsules retrieved from the monkeys who recovered limb function postimplantation contained numerous viable PC12 cells that continued to release levodopa, basal dopamine, and potassium evoked dopamine. In contrast, capsules retrieved from the PC12 cell-grafted monkey which did not recover limb use on the hand reach task contained few cells which secreted negligible or undetectable levels of levodopa and dopamine. Interestingly, functional disability was not reinstated following removal of the capsules. Neuroanatomical and neurochemical evaluation of the grafted striatum did not reveal a host-derived sprouting response of catecholaminergic or indolaminergic fibers. These data indicate that xenografts of PC12 cells can survive for up to 6.5 mo in nonimmunosuppressed monkeys when immunoisolated via polymer encapsulation. Moreover, these cells continue to secrete high levels of levodopa and dopamine and induce recovery of motor function in parkinsonian nonhuman primates.
Collapse
Affiliation(s)
- J H Kordower
- Department of Neurological Sciences, Rush Presbyterian Medical Center, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
17
|
Abstract
Cell transplantation is now being explored as a new therapeutic strategy to restore function in the diseased human central nervous system. Neural grafts show long-term survival and function in patients with Parkinson's disease but the symptomatic relief needs to be increased. Cell transplantation seems justified in patients with Huntington's disease and, at a later stage, possibly also in demyelinating disorders. The further development in this research field will require systematic studies in animal experiments but also well-designed clinical trials in small groups of patients.
Collapse
Affiliation(s)
- O Lindvall
- Department of Neurology, University Hospital, Lund, Sweden
| |
Collapse
|
18
|
Soderstrom K, O'Malley J, Steece-Collier K, Kordower JH. Neural Repair Strategies for Parkinson's Disease: Insights from Primate Models. Cell Transplant 2017; 15:251-65. [PMID: 16719060 DOI: 10.3727/000000006783982025] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Nonhuman primate models of Parkinson's disease (PD) have been invaluable to our understanding of the human disease and in the advancement of novel therapies for its treatment. In this review, we attempt to give a brief overview of the animal models of PD currently used, with a more comprehensive focus on the advantages and disadvantages presented by their use in the nonhuman primate. In particular, discussion addresses the 6-hydroxydopamine (6-OHDA), 1-methyl-1,2,3,6-tetrahydopyridine (MPTP), rotenone, paraquat, and maneb parkinsonian models. Additionally, the role of primate PD models in the development of novel therapies, such as trophic factor delivery, grafting, and deep brain stimulation, are described. Finally, the contribution of primate PD models to our understanding of the etiology and pathology of human PD is discussed.
Collapse
Affiliation(s)
- Katherine Soderstrom
- Department of Neurological Science, Research Center for Brain Repair, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
19
|
Abstract
Neural transplantation has been extensively applied in Parkinson's disease, including numerous clinical studies, studies in animal models, and related basic research on cell biology. There is evidence that the clinical trials of both adrenal medulla transplantation and fetal substantia nigra transplantation have produced a detectable clinical effect, although it is not yet clear whether the clinical benefit is sufficient to justify a more widespread application of these procedures. Studies of long-term outcome and quantitative tests are important in assaying the degree of benefit produced by transplantation procedures in Parkinson's disease and for developing improved and refined procedures. Other disease-related applications of neural transplantation are beginning to be developed. These include Huntington's disease, chronic pain, epilepsy, spinal cord injury, and perhaps even demyelinating diseases and cortical ischemic injury. Although most of these applications lie in the future, it is not too soon to begin to consider the scientific justification that should be required for initiation of human clinical trials.
Collapse
Affiliation(s)
- William J. Freed
- Preclinical Neurosciences Section, Neuropsychiatry Branch, NIMH Neuroscience Center at St. Elizabeths, 2700 Martin Luther King Ave., Washington, DC 20032, USA
| |
Collapse
|
20
|
Kirkeby A, Parmar M, Barker RA. Strategies for bringing stem cell-derived dopamine neurons to the clinic. PROGRESS IN BRAIN RESEARCH 2017; 230:165-190. [DOI: 10.1016/bs.pbr.2016.11.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
21
|
Björklund A, Lindvall O. Replacing Dopamine Neurons in Parkinson's Disease: How did it happen? JOURNAL OF PARKINSON'S DISEASE 2017; 7:S21-S31. [PMID: 28282811 PMCID: PMC5345652 DOI: 10.3233/jpd-179002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The efforts to develop a dopamine cell replacement therapy for Parkinson's disease have spanned over more than three decades. Based on almost 10 years of transplantation studies in animal models, the first patients receiving grafts of fetal-derived dopamine neuroblasts were operated in Lund in 1987. Over the following two decades, a total of 18 patients were transplanted and followed closely by our team with mixed but also very encouraging results. In this article we tell the story of how the preclinical and clinical transplantation program in Lund evolved. We recall the excitement when we obtained the first evidence for survival and function of transplanted neurons in the diseased human brain. We also remember the setbacks that we have experienced during these 30 years and discuss the very interesting developments that are now taking place in this exciting field.
Collapse
Affiliation(s)
- Anders Björklund
- Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden
| | - Olle Lindvall
- Department of Clinical Sciences, and Lund Stem Cell Center, Division of Neurology, University Hospital, Lund, Sweden
| |
Collapse
|
22
|
Abstract
Over the past three decades, significant progress has been made in the development of potential regenerative cell-based therapies for neurodegenerative disease, with most success being seen in Parkinson's disease. Cell-based therapies face many challenges including ethical considerations, potential for immune-mediated rejection with allogeneic and xenogeneic tissue, pathological spread of protein-related disease into the grafted tissue as well as the risk of graft overgrowth and tumorigenesis in stem cell-derived transplants. Preclinical trials have looked at many tissue types of which the most successful to date have been those using fetal ventral mesencephalon grafts, which led to clinical trials, which have shown that in some cases they can work very well. With important proof-of-concept derived from these studies, there is now much interest in how dopaminergic neurons derived from stem cell sources could be used to develop cell-based therapies suitable for clinical use, with clinical trials poised to enter the clinic in the next couple of years.
Collapse
Affiliation(s)
- Thomas B Stoker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge, CB2 0PY, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge
| |
Collapse
|
23
|
Pramanik S, Sulistio YA, Heese K. Neurotrophin Signaling and Stem Cells-Implications for Neurodegenerative Diseases and Stem Cell Therapy. Mol Neurobiol 2016; 54:7401-7459. [PMID: 27815842 DOI: 10.1007/s12035-016-0214-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
Neurotrophins (NTs) are members of a neuronal growth factor protein family whose action is mediated by the tropomyosin receptor kinase (TRK) receptor family receptors and the p75 NT receptor (p75NTR), a member of the tumor necrosis factor (TNF) receptor family. Although NTs were first discovered in neurons, recent studies have suggested that NTs and their receptors are expressed in various types of stem cells mediating pivotal signaling events in stem cell biology. The concept of stem cell therapy has already attracted much attention as a potential strategy for the treatment of neurodegenerative diseases (NDs). Strikingly, NTs, proNTs, and their receptors are gaining interest as key regulators of stem cells differentiation, survival, self-renewal, plasticity, and migration. In this review, we elaborate the recent progress in understanding of NTs and their action on various stem cells. First, we provide current knowledge of NTs, proNTs, and their receptor isoforms and signaling pathways. Subsequently, we describe recent advances in the understanding of NT activities in various stem cells and their role in NDs, particularly Alzheimer's disease (AD) and Parkinson's disease (PD). Finally, we compile the implications of NTs and stem cells from a clinical perspective and discuss the challenges with regard to transplantation therapy for treatment of AD and PD.
Collapse
Affiliation(s)
- Subrata Pramanik
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Yanuar Alan Sulistio
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 133-791, Republic of Korea.
| |
Collapse
|
24
|
Corey S, Lippert T, Borlongan CV. Translational lab-to-clinic hurdles in stem cell therapy. Chin Neurosurg J 2016. [DOI: 10.1186/s41016-016-0058-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
25
|
Lindvall O. Treatment of Parkinson's disease using cell transplantation. Philos Trans R Soc Lond B Biol Sci 2016; 370:20140370. [PMID: 26416681 DOI: 10.1098/rstb.2014.0370] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The clinical trials with intrastriatal transplantation of human fetal mesencephalic tissue, rich in dopaminergic neurons, in Parkinson's disease (PD) patients show that cell replacement can work and in some cases induce major, long-lasting improvement. However, owing to poor tissue availability, this approach can only be applied in very few patients, and standardization is difficult, leading to wide variation in functional outcome. Stem cells and reprogrammed cells could potentially be used to produce dopaminergic neurons for transplantation. Importantly, dopaminergic neurons of the correct substantia nigra phenotype can now be generated from human embryonic stem cells in large numbers and standardized preparations, and will soon be ready for application in patients. Also, human induced pluripotent stem cell-derived dopaminergic neurons are being considered for clinical translation. Available data justify moving forward in a responsible way with these dopaminergic neurons, which should be tested, using optimal patient selection, cell preparation and transplantation procedures, in controlled clinical studies.
Collapse
Affiliation(s)
- Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, 221 84 Lund, Sweden
| |
Collapse
|
26
|
Marxreiter F, Storch A, Winkler J. [Cellular replacement strategies and adult neurogenesis in idiopathic Parkinson's disease]. DER NERVENARZT 2016; 87:805-13. [PMID: 27389601 DOI: 10.1007/s00115-016-0157-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is the most common age-related movement disorder and characterized by slowly progressive neurodegeneration resulting in motor symptoms, such as bradykinesia, rigidity, tremor and postural instability. Moreover, non-motor symptoms, such as hyposmia, anxiety and depression reduce the quality of life in PD. Motor symptoms are associated with a distinct striatal dopaminergic deficit resulting from axonal dysfunction and neuronal loss in the substantia nigra (SN). Recent progress in stem cell technology allows the optimization of cellular transplantation strategies in order to alleviate the motor deficit, which potentially leads to a reactivation of this therapeutic strategy. Besides neurodegenerative processes impaired adult neurogenesis and consequentially reduced endogenous cellular plasticity may play an important role in PD. This article discusses the notion that non-motor symptoms in PD may partly be explained by reduced adult neurogenesis in the olfactory bulb and hippocampus.
Collapse
Affiliation(s)
- F Marxreiter
- Abteilung für Molekulare Neurologie, Universitätsklinikum Erlangen, Schwabachanlage 6, 91054, Erlangen, Deutschland
| | - A Storch
- Klinik und Poliklinik für Neurologie, Universität Rostock, Gehlsheimer Straße 20, 18147, Rostock, Deutschland.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock, Gehlsheimer Straße 20, 18147, Rostock, Deutschland
| | - J Winkler
- Abteilung für Molekulare Neurologie, Universitätsklinikum Erlangen, Schwabachanlage 6, 91054, Erlangen, Deutschland.
| |
Collapse
|
27
|
Abstract
In Parkinson's disease (PD), the main pathology underlying the motor symptoms is a loss of nigrostriatal dopaminergic neurons. Clinical trials of intrastriatal transplantation of human foetal mesencephalic tissue have shown that the grafted dopaminergic neurons re-innervate the striatum, restore striatal dopamine release and, in some cases, induce major, long-lasting improvement of motor function. However, nonmotor symptoms originating from degeneration outside the striatum or in nondopaminergic systems are not alleviated by intrastriatal implantation of dopaminergic neurons. Stem cells and reprogrammed cells could potentially be used to produce dopaminergic neurons for transplantation in patients with PD. Recent studies demonstrate that standardized preparations of dopaminergic neurons of the correct substantia nigra phenotype can be generated from human embryonic stem cells in large numbers, and they will soon be available for patient application. In addition, dopaminergic neurons derived from human induced pluripotent stem cells are being considered for clinical translation. Important challenges include the demonstration of potency (growth capacity and functional efficacy) and safety of the generated dopaminergic neurons in preclinical animal models. The dopaminergic neurons should subsequently be tested, using optimal patient selection and cell preparation and transplantation procedures, in controlled clinical studies.
Collapse
Affiliation(s)
- O Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, University Hospital, Lund, Sweden
| |
Collapse
|
28
|
Foetal Cell Transplantation for Parkinson's Disease: Focus on Graft-Induced Dyskinesia. PARKINSONS DISEASE 2015; 2015:563820. [PMID: 26881178 PMCID: PMC4736211 DOI: 10.1155/2015/563820] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 12/02/2015] [Accepted: 12/16/2015] [Indexed: 02/05/2023]
Abstract
Transplantation of dopamine- (DA-) rich foetal ventral mesencephalic cells emerged as a promising therapy for Parkinson's disease (PD), as it allowed significant improvement of motor symptoms in several PD patients in open-label studies. However, double-blind clinical trials have been largely disappointing. The general agreement in the field is that the lack of standardization of tissue collection and preparation, together with the absence of postsurgical immunosuppression, played a key role in the failure of these studies. Moreover, a further complication that emerged in previous studies is the appearance of the so-called graft-induced dyskinesia (GID), in a subset of grafted patients, which resembles dyskinesia induced by L-DOPA but in the absence of medication. Preclinical evidence pointed to the serotonin neurons as possible players in the appearance of GID. In agreement, clinical investigations have shown that grafted tissue may contain a large number of serotonin neurons, in the order of half of the DA cells; moreover, the serotonin 5-HT1A receptor agonist buspirone has been found to produce significant dampening of GID in grafted patients. In this paper, we will review the recent preclinical and clinical studies focusing on cell transplantation for PD and on the mechanisms underlying GID.
Collapse
|
29
|
Barker RA, Drouin-Ouellet J, Parmar M. Cell-based therapies for Parkinson disease—past insights and future potential. Nat Rev Neurol 2015; 11:492-503. [PMID: 26240036 DOI: 10.1038/nrneurol.2015.123] [Citation(s) in RCA: 214] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Parkinson disease (PD) is characterized by loss of the A9 nigral neurons that provide dopaminergic innervation to the striatum. This discovery led to the successful instigation of dopaminergic drug treatments in the 1960s, although these drugs were soon recognized to lose some of their efficacy and generate their own adverse effects over time. Despite the fact that PD is now known to have extensive non-nigral pathology with a wide range of clinical features, dopaminergic drug therapies are still the mainstay of therapy, and work well for many years. Given the success of pharmacological dopamine replacement, pursuit of cell-based dopamine replacement strategies seemed to be the next logical step, and studies were initiated over 30 years ago to explore the possibility of dopaminergic cell transplantation. In this Review, we outline the history of this therapeutic approach to PD and highlight the lessons that we have learned en route. We discuss how the best clinical outcomes have been obtained with fetal ventral mesencephalic allografts, while acknowledging inconsistencies in the results owing to problems in trial design, patient selection, tissue preparation, and immunotherapy used post-grafting. We conclude by discussing the challenges of bringing the new generation of stem cell-derived dopamine cells to the clinic.
Collapse
Affiliation(s)
- Roger A Barker
- John van Geest Centre for Brain Repair &Department of Neurology, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Cambridge CB2 0PY, UK
| | - Janelle Drouin-Ouellet
- Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, Lund University, BMC A11, S-221 84 Lund, Sweden
| | - Malin Parmar
- Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, Lund University, BMC A11, S-221 84 Lund, Sweden
| |
Collapse
|
30
|
Borlongan CV, Jolkkonen J, Detante O. The future of stem cell therapy for stroke rehabilitation. FUTURE NEUROLOGY 2015; 10:313-319. [PMID: 26997918 DOI: 10.2217/fnl.15.27] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612 USA
| | - Jukka Jolkkonen
- University of Eastern Finland, Institute of Clinical Medicine - Neurology, Yliopistonranta 1 C, 70210 Kuopio, Finland
| | - Olivier Detante
- University Hospital of Grenoble, Stroke Unit, Department of Neurology, CS 10217, 38043 Grenoble, France; Inserm, U 836, BP 170, 38042 Grenoble, France; University Grenoble Alpes, Grenoble Institute of Neurosciences, BP 170, 38042 Grenoble, France
| |
Collapse
|
31
|
Barker RA. What have open label studies of cell based therapies for Parkinson's disease told us, if anything? ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.baga.2014.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
32
|
Remy P. Biotherapies for Parkinson disease. Rev Neurol (Paris) 2014; 170:763-9. [DOI: 10.1016/j.neurol.2014.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 10/08/2014] [Indexed: 11/17/2022]
|
33
|
Canet-Aviles R, Lomax GP, Feigal EG, Priest C. Proceedings: cell therapies for Parkinson's disease from discovery to clinic. Stem Cells Transl Med 2014; 3:979-91. [PMID: 25150264 DOI: 10.5966/sctm.2014-0146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In March 2013, the California Institute for Regenerative Medicine, in collaboration with the NIH Center for Regenerative Medicine, held a 2-day workshop on cell therapies for Parkinson's disease (PD), with the goals of reviewing the state of stem cell research for the treatment of PD and discussing and refining the approach and the appropriate patient populations in which to plan and conduct new clinical trials using stem cell-based therapies for PD. Workshop participants identified priorities for research, development, and funding; discussed existing resources and initiatives; and outlined a path to the clinic for a stem cell-based therapy for PD. A consensus emerged among participants that the development of cell replacement therapies for PD using stem cell-derived products could potentially offer substantial benefits to patients. As with all stem cell-based therapeutic approaches, however, there are many issues yet to be resolved regarding the safety, efficacy, and methodology of transplanting cell therapies into patients. Workshop participants agreed that designing an effective stem cell-based therapy for PD will require further research and development in several key areas. This paper summarizes the meeting.
Collapse
Affiliation(s)
- Rosa Canet-Aviles
- California Institute for Regenerative Medicine, San Francisco, California, USA
| | - Geoffrey P Lomax
- California Institute for Regenerative Medicine, San Francisco, California, USA
| | - Ellen G Feigal
- California Institute for Regenerative Medicine, San Francisco, California, USA
| | - Catherine Priest
- California Institute for Regenerative Medicine, San Francisco, California, USA
| |
Collapse
|
34
|
Cisbani G, Cicchetti F. Review: The fate of cell grafts for the treatment of Huntington's disease: thepost-mortemevidence. Neuropathol Appl Neurobiol 2014; 40:71-90. [DOI: 10.1111/nan.12104] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 12/03/2013] [Indexed: 12/13/2022]
Affiliation(s)
- G. Cisbani
- Centre de Recherche du CHU de Québec (CHUL); Québec QC Canada
| | - F. Cicchetti
- Centre de Recherche du CHU de Québec (CHUL); Québec QC Canada
- Département de Psychiatrie et Neurosciences; Université Laval; Québec QC Canada
| |
Collapse
|
35
|
Sydow O, Hansson P, Young D, Meyerson B, Backlund EO, Ebendal T, Farnebo LO, Freedman R, Hamberger B, Hoffer B, Seiger A, Strömberq I, Olson L. Long-term beneficial effects of adrenal medullary autografts supported by nerve growth factor in Parkinson's disease. Eur J Neurol 2013; 2:445-54. [PMID: 24283725 DOI: 10.1111/j.1468-1331.1995.tb00154.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Parkinson's disease has been the object of several therapeutic strategies based upon replacement of the degenerating dopaminergic neurons. Adrenal medullary transplants were tried initially, because of the biochemical relationship between chromaffin cells of the medulla and dopaminergic neurons of the substantia nigra. Compared to transplant of fetal neurons, autologous grafts of adrenal medullary tissue has the advantage of using a readily available source of tissue without the problems of immunosuppression. However, these cells have not proven to be as effective as fetal neurons, probably because they do not fully differentiate into neurons. In animal models, brief treatment with nerve growth factor can facilitate such differentiation. This study is a clinical evaluation of the efficacy of adrenal medullary cell transplantation, combined with nerve growth factor infusion. Two patients were selected who were moderately to severely affected (Hoehn-Yahr stage 2 in on-phase and stage 4 in off-phase). After adrenalectomy, small pieces of medulla were prepared and implanted stereotactically into the dorsal putamen on one side of the brain. A catheter filled with mouse beta-nerve growth factor (NGF) was placed close to the grafts. Infusion of NGF was continued for one month. Despite a progressively deteriorating course prior to surgery, both patients showed improvement on the rating scales postoperatively. There was also significant improvement in timed motor tests. Motor readiness evoked potentials showed increased voltage over the operated hemisphere. The study points to methods and feasibility of supplying nerve growth factor intraparenchymally to the human brain. Possible implications with respect to other growth factors, particularly Glial cell-line Derived Neurotrophic factor (GDNF) are discussed.
Collapse
Affiliation(s)
- O Sydow
- Department of Neurology, Karolinska Institutet, Danderyd Hospital, Danderyd, SwedenNeurogenic Pain Unit, Department of Rehabilitation Medicine, Karolinska Hospital, Stockholm, SwedenDepartments of Neurosurgery, Karolinska Hospital, Stockholm, SwedenGeneral Surgery, Karolinska Hospital, Stockholm, SwedenDepartments of NeuroscienceGeriatric Medicine, Karolinska Institutet, Stockholm, SwedenDepartment of Neurosurgery, Linköping University Hospital, Linköping, SwedenDepartment of Developmental Biology, Biomedical Center, Uppsala University, Uppsala, SwedenDepartments of Psychiatry and PharmacologyPreventive Medicine and Biostatistics, Denver Veterans Administration Medical Center and University of Colorado, Denver, Colorado, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bjorklund A, Kordower JH. Cell therapy for Parkinson's disease: what next? Mov Disord 2013; 28:110-5. [PMID: 23390097 DOI: 10.1002/mds.25343] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 12/11/2012] [Accepted: 12/13/2012] [Indexed: 11/08/2022] Open
Abstract
The idea to use transplants of dopamine-producing cells to substitute for the lost midbrain dopamine neurons in Parkinson's disease (PD) goes back to the 1970s. In this review we give an overview of the history of cell transplantation in animal models of PD, and summarize the experience gained from the open-label and placebo-controlled clinical trials performed so far using intrastriatal transplants of human fetal dopamine neuroblasts. Further development of this therapeutic approach face numerous challenges, for example in the development of protocols that allow generation of fully functional and safe midbrain dopamine neurons from stem cells. Based on recent promising advancements, efforts are now being made to develop standardized and efficient protocols, and adapt these protocols to good laboratory practice (GLP)/good manufacturing practice (GMP) conditions, to move this technology closer to clinical translation.
Collapse
|
37
|
Lindvall O. Developing dopaminergic cell therapy for Parkinson's disease-give up or move forward? Mov Disord 2013; 28:268-73. [DOI: 10.1002/mds.25378] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/25/2012] [Accepted: 01/03/2013] [Indexed: 01/24/2023] Open
Affiliation(s)
- Olle Lindvall
- Lund Stem Cell Center; University Hospital; Lund; Sweden
| |
Collapse
|
38
|
Ambriz-Tututi M, Monjaraz-Fuentes F, Drucker-Colín R. Chromaffin cell transplants: From the lab to the clinic. Life Sci 2012; 91:1243-51. [DOI: 10.1016/j.lfs.2012.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/13/2012] [Accepted: 10/05/2012] [Indexed: 11/29/2022]
|
39
|
Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. J Transl Med 2012. [PMID: 23190582 PMCID: PMC3543237 DOI: 10.1186/1479-5876-10-239] [Citation(s) in RCA: 323] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The physiological role of the neurotrophin nerve growth factor (NGF) has been characterized, since its discovery in the 1950s, first in the sensory and autonomic nervous system, then in central nervous, endocrine and immune systems. NGF plays its trophic role both during development and in adulthood, ensuring the maintenance of phenotypic and functional characteristic of several populations of neurons as well as immune cells. From a translational standpoint, the action of NGF on cholinergic neurons of the basal forebrain and on sensory neurons in dorsal root ganglia first gained researcher's attention, in view of possible clinical use in Alzheimer's disease patients and in peripheral neuropathies respectively. The translational and clinical research on NGF have, since then, enlarged the spectrum of diseases that could benefit from NGF treatment, at the same time highlighting possible limitations in the use of the neurotrophin as a drug. In this review we give a comprehensive account for almost all of the clinical trials attempted until now by using NGF. A perspective on future development for translational research on NGF is also discussed, in view of recent proposals for innovative delivery strategies and/or for additional pathologies to be treated, such as ocular and skin diseases, gliomas, traumatic brain injuries, vascular and immune diseases.
Collapse
Affiliation(s)
- Luigi Aloe
- Cellular Biology and Neurobiology Institute, CNR, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | | | | | | |
Collapse
|
40
|
Clinical and Experimental Experiences of Graft-Induced Dyskinesia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 98:173-86. [DOI: 10.1016/b978-0-12-381328-2.00007-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
41
|
Abstract
The history of cell transplantation in the nervous system is reviewed in four main sections. The "early era" spans the period from 1890 to 1940, during which the first attempts at cell transplantation in the brain were undertaken. Many contemporary themes were first addressed such as surgical factors to achieve survival of grafted cells and how that should be assessed, immunological factors, use of tumors as a readily viable cell source; and use of the anterior eye chamber as a model transplantation site. However, such studies generally exhibited only low levels of viability or successful implantation. The "middle era" from 1940 to 1970 spans the period when the techniques for viable and reliable cell transplantation using embryonic donor tissues implanted into sites with effective vascularization were first established in brain and neuroendocrine systems in a limited number of specialist centers. However, although sometimes impressive, these results were at variance with the prevailing view that the adult mammalian brain is immutable and resistant to plasticity, growth or regeneration, and were largely ignored. The "modern era," since 1970, began with the pioneering studies that combined cell transplantation with the use of improved histochemical and ultrastructural anatomical techniques to demonstrate selectivity, specificity and regenerative capacity of implanted cells, and the slow acceptance that the adult brain does exhibit considerable potential for plasticity and repair. The last three decades have witnessed the identification of reliable and efficient transplantation technologies combined with progressively refined methods of molecular, cellular, biochemical, physiological and functional analysis. This now enables the ready use of cell transplantation as a powerful novel method within the neuroscience tool-kit, which is being used: to analyze normal organization and function of the nervous system; to reveal the biological mechanisms and principles of neuronal development, regeneration and plasticity; and to study the principles of surgically directed cell therapies for promoting plasticity, replacement and repair in response to injury and disease. The final section reviews recent progress in translating cell transplantation to the clinic for application in Parkinson's and other central nervous system diseases.
Collapse
|
42
|
Pérez-Alvarez A, Hernández-Vivanco A, Albillos A. Past, present and future of human chromaffin cells: role in physiology and therapeutics. Cell Mol Neurobiol 2010; 30:1407-15. [PMID: 21107679 PMCID: PMC11498861 DOI: 10.1007/s10571-010-9582-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Accepted: 09/02/2010] [Indexed: 12/26/2022]
Abstract
Chromaffin cells are neuroendocrine cells mainly found in the medulla of the adrenal gland. Most existing knowledge of these cells has been the outcome of extensive research performed in animals, mainly in the cow, cat, mouse and rat. However, some insight into the physiology of this neuroendocrine cell in humans has been gained. This review summarizes the main findings reported in human chromaffin cells under physiological or disease conditions and discusses the clinical implications of these results.
Collapse
Affiliation(s)
- Alberto Pérez-Alvarez
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, c/Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Alicia Hernández-Vivanco
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, c/Arzobispo Morcillo 4, 28029 Madrid, Spain
| | - Almudena Albillos
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, c/Arzobispo Morcillo 4, 28029 Madrid, Spain
| |
Collapse
|
43
|
Huang H, Chen L, Sanberg P. Cell Therapy From Bench to Bedside Translation in CNS Neurorestoratology Era. CELL MEDICINE 2010; 1:15-46. [PMID: 21359168 DOI: 10.3727/215517910x516673] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent advances in cell biology, neural injury and repair, and the progress towards development of neurorestorative interventions are the basis for increased optimism. Based on the complexity of the processes of demyelination and remyelination, degeneration and regeneration, damage and repair, functional loss and recovery, it would be expected that effective therapeutic approaches will require a combination of strategies encompassing neuroplasticity, immunomodulation, neuroprotection, neurorepair, neuroreplacement, and neuromodulation. Cell-based restorative treatment has become a new trend, and increasing data worldwide have strongly proven that it has a pivotal therapeutic value in CNS disease. Moreover, functional neurorestoration has been achieved to a certain extent in the CNS clinically. Up to now, the cells successfully used in preclinical experiments and/or clinical trial/treatment include fetal/embryonic brain and spinal cord tissue, stem cells (embryonic stem cells, neural stem/progenitor cells, hematopoietic stem cells, adipose-derived adult stem/precursor cells, skin-derived precursor, induced pluripotent stem cells), glial cells (Schwann cells, oligodendrocyte, olfactory ensheathing cells, astrocytes, microglia, tanycytes), neuronal cells (various phenotypic neurons and Purkinje cells), mesenchymal stromal cells originating from bone marrow, umbilical cord, and umbilical cord blood, epithelial cells derived from the layer of retina and amnion, menstrual blood-derived stem cells, Sertoli cells, and active macrophages, etc. Proof-of-concept indicates that we have now entered a new era in neurorestoratology.
Collapse
Affiliation(s)
- Hongyun Huang
- Center for Neurorestoratology, Beijing Rehabilitation Center, Beijing, P.R. China
| | | | | |
Collapse
|
44
|
Abstract
Parkinson's disease is the second most common age-related neurodegenerative disorder, typified by the progressive loss of substantia nigra pars compacta dopamine neurons and the consequent decrease in the neurotransmitter dopamine. Patients exhibit a range of clinical symptoms, with the most common affecting motor function and including resting tremor, rigidity, akinesia, bradykinesia and postural instability. Current pharmacological interventions are palliative and largely aimed at increasing dopamine levels through increased production and/or inhibition of metabolism of this key neurotransmitter. The gold standard for treatment of both familial and sporadic Parkinson's disease is the peripheral administration of the dopamine precursor, levodopa. However, many patients gradually develop levodopa-induced dyskinesias and motor fluctuations. In addition, dopamine enhancement therapies are most useful when a portion of the nigrostriatal pathway is intact. Consequently, as the number of substantia nigra dopamine neurons and striatal projections decrease, these treatments become less efficacious. Current translational research is focused on the development of novel disease-modifying therapies, including those utilizing gene therapeutic approaches. Herein we present an overview of current gene therapy clinical trials for Parkinson's disease. Employing either recombinant adeno-associated virus type 2 (rAAV2) or lentivirus vectors, these clinical trials are focused on three overarching approaches: augmentation of dopamine levels via increased neurotransmitter production; modulation of the neuronal phenotype; and neuroprotection. The first two therapies discussed in this article focus on increasing dopamine production via direct delivery of genes involved in neurotransmitter synthesis (amino acid decarboxylase, tyrosine hydroxylase and GTP [guanosine triphosphate] cyclohydrolase 1). In an attempt to bypass the degenerating nigrostriatal pathway, a third clinical trial utilizes rAAV2 to deliver glutamic acid decarboxylase to the subthalamic nucleus, converting a subset of excitatory neurons to GABA-producing cells. In contrast, the final clinical trial is aimed at protecting the degenerating nigrostriatum by striatal delivery of rAAV2 harbouring the neuroprotective gene, neurturin. Based on preclinical studies, this gene therapeutic approach is posited to slow disease progression by enhancing neuronal survival. In addition, we discuss the outcome of each clinical trial and discuss the potential rationale for the marginal yet incremental clinical advancements that have thus far been realized for Parkinson's disease gene therapy.
Collapse
|
45
|
Wijeyekoon R, Barker RA. Cell replacement therapy for Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2009; 1792:688-702. [DOI: 10.1016/j.bbadis.2008.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 10/12/2008] [Accepted: 10/13/2008] [Indexed: 12/21/2022]
|
46
|
|
47
|
Maguire-Zeiss KA. alpha-Synuclein: a therapeutic target for Parkinson's disease? Pharmacol Res 2008; 58:271-80. [PMID: 18840530 PMCID: PMC2630208 DOI: 10.1016/j.phrs.2008.09.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 09/10/2008] [Accepted: 09/11/2008] [Indexed: 12/21/2022]
Abstract
Parkinson's disease is a progressive age-related neurodegenerative disease with invariant loss of substantia nigra dopamine neurons and striatal projections. This disorder is well known for the associated motoric symptoms including resting tremor and the inability to initiate movement. However, it is now apparent that Parkinson's disease is a multisystem disorder with patients exhibiting symptoms derived from peripheral nervous system and extra-nigral dysfunctions in addition to the prototypical nigrostriatal damage. Although the etiology for sporadic Parkinson's disease is unknown, information gleaned from both familial forms of the disease and animal models places misfolded alpha-synuclein at the forefront. The disease is currently without a cure and most therapies target the motoric symptoms relying on increasing dopamine tone. In this review, the role of alpha-synuclein in disease pathogenesis and as a potential therapeutic target focusing on toxic conformers of this protein is considered. The addition of protofibrillar/oligomer-directed neurotherapeutics to the existing armamentarium may extend the symptom-free stage of Parkinson's disease as well as alleviate pathogenesis.
Collapse
Affiliation(s)
- Kathleen A Maguire-Zeiss
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Road, Washington, DC 20057, United States.
| |
Collapse
|
48
|
Chen Z, Palmer TD. Cellular repair of CNS disorders: an immunological perspective. Hum Mol Genet 2008; 17:R84-92. [PMID: 18632702 DOI: 10.1093/hmg/ddn104] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cellular repair is a promising strategy for treating central nervous system (CNS) disorders. Several strategies have been contemplated including replacement of neurons or glia that have been lost due to injury or disease, use of cellular grafts to modify or augment the functions of remaining neurons and/or use of cellular grafts to protect neural tissue by local delivery of growth or trophic factors. Depending on the specific disease target, there may be one or many cell types that could be considered for therapy. In each case, an additional variable must be considered--the role of the immune system in both the injury process itself and in the response to incoming cells. Cellular transplants can be roughly categorized into autografts, allografts and xenografts. Despite the immunological privilege of the CNS, allografts and xenografts can elicit activation of the innate and adaptive immune system. In this article, we evaluate the various effects that immune cells and signals may have on the survival, proliferation, differentiation and migration/integration of transplanted cells in therapeutic approaches to CNS injury and disease.
Collapse
Affiliation(s)
- Zhiguo Chen
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
49
|
Tan SA, Aebischer P. The problems of delivering neuroactive molecules to the CNS. CIBA FOUNDATION SYMPOSIUM 2007; 196:211-36; discussion 236-9. [PMID: 8866136 DOI: 10.1002/9780470514863.ch14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
At present, the aetiologies of many neurological and neurodegenerative diseases are unknown. However, emergence of a better understanding of these diseases, at both cellular and molecular levels, opens up the possibility of replacement therapies. The presence of the blood-brain barrier complicates the delivery of molecules to the central nervous system. Numerous attempts have been made to bypass this barrier either by delivering the drugs directly into the brain or by transplanting cells to produce the missing molecules in situ. This review explores several methods for delivering bioactive molecules into the CNS, including the use of permeabilizers, osmotic pumps, slow polymer release systems and transplantation of cells with or without the use of the encapsulation technology.
Collapse
Affiliation(s)
- S A Tan
- Gene Therapy Center, Lausanne University Medical School, CHU Vaudois, Switzerland
| | | |
Collapse
|
50
|
Nakao N, Shintani-Mizushima A, Kakishita K, Itakura T. Transplantation of autologous sympathetic neurons as a potential strategy to restore metabolic functions of the damaged nigrostriatal dopamine nerve terminals in Parkinson's disease. ACTA ACUST UNITED AC 2006; 52:244-56. [PMID: 16644019 DOI: 10.1016/j.brainresrev.2006.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Revised: 02/18/2006] [Accepted: 03/07/2006] [Indexed: 11/30/2022]
Abstract
Grafting of catecholamine-producing cells can be a possible therapeutic strategy for attenuating motor symptoms in Parkinson's disease (PD). The potential of autologous sympathetic neurons has been investigated as a donor for cell therapy of PD. The clinical trials of autotransplantation of sympathetic ganglion cells in PD have revealed that the grafts increase the duration of L-DOPA (L-dihydroxy phenyl alanine)-induced beneficial effects, and that the graft-mediated effect is detectable during a follow-up period of at least 1 year postgrafting. In an in vitro analysis of the ability of human sympathetic neurons to release catecholamines, although DA was not detectable under basal conditions, DA levels were significantly increased upon exposure to exogenous L-DOPA. Furthermore, animal experiments with xenografting of human sympathetic ganglionic neurons in the DA-denervated striatum of rats demonstrated that a significant increase in striatal DA levels is noted after systemic L-DOPA treatment, and that the DA levels remain high for longer periods of time in the grafted rats than in control animals with sham surgery. The L-DOPA-induced rise of striatal DA levels was significantly attenuated when given reserpine pretreatment. This suggests that DA derived from exogenously administered L-DOPA is subjected to, at least in part, vesicular storage in grafted sympathetic neurons. Histological examinations indeed showed that the grafts express aromatic-L-amino acid decarboxylase and vesicular monoamine transporter-2, both of which are important molecules for the synthesis and the storage of DA, respectively. Taken together, grafted sympathetic neurons can provide a site for both the conversion of exogenous L-DOPA to DA and the storage of the synthesized DA in the DA-denervated striatum. This might be an explanation for a mechanism by which sympathetic neuron autografts can increase the duration of L-DOPA effects in PD patients. This review article summarizes the clinical effect of transplantation of autologous sympathetic neurons in PD and discusses the underlying mechanism for the effect based on experimental evidence previously obtained.
Collapse
Affiliation(s)
- Naoyuki Nakao
- Department of Neurological Surgery, Wakayama Medical University, Wakayama 641-0012, Japan.
| | | | | | | |
Collapse
|