1
|
Dong W, Liu S, Li S, Wang Z. Cell reprogramming therapy for Parkinson's disease. Neural Regen Res 2024; 19:2444-2455. [PMID: 38526281 PMCID: PMC11090434 DOI: 10.4103/1673-5374.390965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/23/2023] [Accepted: 10/08/2023] [Indexed: 03/26/2024] Open
Abstract
Parkinson's disease is typically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta. Many studies have been performed based on the supplementation of lost dopaminergic neurons to treat Parkinson's disease. The initial strategy for cell replacement therapy used human fetal ventral midbrain and human embryonic stem cells to treat Parkinson's disease, which could substantially alleviate the symptoms of Parkinson's disease in clinical practice. However, ethical issues and tumor formation were limitations of its clinical application. Induced pluripotent stem cells can be acquired without sacrificing human embryos, which eliminates the huge ethical barriers of human stem cell therapy. Another widely considered neuronal regeneration strategy is to directly reprogram fibroblasts and astrocytes into neurons, without the need for intermediate proliferation states, thus avoiding issues of immune rejection and tumor formation. Both induced pluripotent stem cells and direct reprogramming of lineage cells have shown promising results in the treatment of Parkinson's disease. However, there are also ethical concerns and the risk of tumor formation that need to be addressed. This review highlights the current application status of cell reprogramming in the treatment of Parkinson's disease, focusing on the use of induced pluripotent stem cells in cell replacement therapy, including preclinical animal models and progress in clinical research. The review also discusses the advancements in direct reprogramming of lineage cells in the treatment of Parkinson's disease, as well as the controversy surrounding in vivo reprogramming. These findings suggest that cell reprogramming may hold great promise as a potential strategy for treating Parkinson's disease.
Collapse
Affiliation(s)
- Wenjing Dong
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shuyi Liu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| |
Collapse
|
2
|
Muleiro Alvarez M, Cano-Herrera G, Osorio Martínez MF, Vega Gonzales-Portillo J, Monroy GR, Murguiondo Pérez R, Torres-Ríos JA, van Tienhoven XA, Garibaldi Bernot EM, Esparza Salazar F, Ibarra A. A Comprehensive Approach to Parkinson's Disease: Addressing Its Molecular, Clinical, and Therapeutic Aspects. Int J Mol Sci 2024; 25:7183. [PMID: 39000288 PMCID: PMC11241043 DOI: 10.3390/ijms25137183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Parkinson's disease (PD) is a gradually worsening neurodegenerative disorder affecting the nervous system, marked by a slow progression and varied symptoms. It is the second most common neurodegenerative disease, affecting over six million people in the world. Its multifactorial etiology includes environmental, genomic, and epigenetic factors. Clinical symptoms consist of non-motor and motor symptoms, with motor symptoms being the classic presentation. Therapeutic approaches encompass pharmacological, non-pharmacological, and surgical interventions. Traditional pharmacological treatment consists of administering drugs (MAOIs, DA, and levodopa), while emerging evidence explores the potential of antidiabetic agents for neuroprotection and gene therapy for attenuating parkinsonian symptoms. Non-pharmacological treatments, such as exercise, a calcium-rich diet, and adequate vitamin D supplementation, aim to slow disease progression and prevent complications. For those patients who have medically induced side effects and/or refractory symptoms, surgery is a therapeutic option. Deep brain stimulation is the primary surgical option, associated with motor symptom improvement. Levodopa/carbidopa intestinal gel infusion through percutaneous endoscopic gastrojejunostomy and a portable infusion pump succeeded in reducing "off" time, where non-motor and motor symptoms occur, and increasing "on" time. This article aims to address the general aspects of PD and to provide a comparative comprehensive review of the conventional and the latest therapeutic advancements and emerging treatments for PD. Nevertheless, further studies are required to optimize treatment and provide suitable alternatives.
Collapse
Affiliation(s)
- Mauricio Muleiro Alvarez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Gabriela Cano-Herrera
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - María Fernanda Osorio Martínez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | | | - Germán Rivera Monroy
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Renata Murguiondo Pérez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Jorge Alejandro Torres-Ríos
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Ximena A. van Tienhoven
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Ernesto Marcelo Garibaldi Bernot
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Felipe Esparza Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Campus México Norte, Huixquilucan 52786, Mexico
- Secretaria de la Defensa Nacional, Escuela Militar de Graduados en Sanidad, Ciudad de México 11200, Mexico
| |
Collapse
|
3
|
Pappolla MA, Wu P, Fang X, Poeggeler B, Sambamurti K, Wisniewski T, Perry G. Stem Cell Interventions in Neurology: From Bench to Bedside. J Alzheimers Dis 2024; 101:S395-S416. [PMID: 39422938 DOI: 10.3233/jad-230897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Stem cell therapies are progressively redefining the treatment landscape for a spectrum of neurological and age-related disorders. This review discusses the molecular and functional attributes of stem cells, emphasizing the roles of neural stem cells and mesenchymal stem cells in the context of neurological diseases such as stroke, multiple sclerosis, amyotrophic lateral sclerosis, traumatic brain injury, Parkinson's disease, and Alzheimer's disease. The review also explores the potential of stem cells in addressing the aging process. The paper analyzes stem cells' intrinsic properties of self-renewal, differentiation, and paracrine effects, alongside the importance of laboratory-modified stem cells like induced pluripotent stem cells and transgenic stem cells. Insights into disease-specific stem cell treatments are offered, reviewing both successes and challenges in the field. This includes the translational difficulties from rodent studies to human trials. The review concludes by acknowledging the uncharted territories that warrant further investigation, emphasizing the potential roles of stem cell-derived exosomes and indole-related molecules, and aiming at providing a basic understanding of stem cell therapies.
Collapse
Affiliation(s)
- Miguel A Pappolla
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ping Wu
- Department of Neuroscience & Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiang Fang
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Burkhard Poeggeler
- Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, Faculty of Biology and Psychology, Georg August University Göttingen, Gütersloh, Germany
| | - Kumar Sambamurti
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA
| | - Thomas Wisniewski
- Departments of Neurology, Pathology, and Psychiatry, New York University Alzheimer's Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
4
|
Toh HSY, Choo XY, Sun AX. Midbrain organoids-development and applications in Parkinson's disease. OXFORD OPEN NEUROSCIENCE 2023; 2:kvad009. [PMID: 38596240 PMCID: PMC10913847 DOI: 10.1093/oons/kvad009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/31/2023] [Indexed: 04/11/2024]
Abstract
Human brain development is spatially and temporally complex. Insufficient access to human brain tissue and inadequacy of animal models has limited the study of brain development and neurodegenerative diseases. Recent advancements of brain organoid technology have created novel opportunities to model human-specific neurodevelopment and brain diseases. In this review, we discuss the use of brain organoids to model the midbrain and Parkinson's disease. We critically evaluate the extent of recapitulation of PD pathology by organoids and discuss areas of future development that may lead to the model to become a next-generation, personalized therapeutic strategy for PD and beyond.
Collapse
Affiliation(s)
- Hilary S Y Toh
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
| | - Xin Yi Choo
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
| | - Alfred Xuyang Sun
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
- National Neuroscience Institute, 11 Jln Tan Tock Seng, Singapore
| |
Collapse
|
5
|
Cha Y, Park TY, Leblanc P, Kim KS. Current Status and Future Perspectives on Stem Cell-Based Therapies for Parkinson's Disease. J Mov Disord 2023; 16:22-41. [PMID: 36628428 PMCID: PMC9978267 DOI: 10.14802/jmd.22141] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/29/2022] [Indexed: 01/12/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, affecting 1%-2% of the population over the age of 65. As the population ages, it is anticipated that the burden on society will significantly escalate. Although symptom reduction by currently available pharmacological and/or surgical treatments improves the quality of life of many PD patients, there are no treatments that can slow down, halt, or reverse disease progression. Because the loss of a specific cell type, midbrain dopamine neurons in the substantia nigra, is the main cause of motor dysfunction in PD, it is considered a promising target for cell replacement therapy. Indeed, numerous preclinical and clinical studies using fetal cell transplantation have provided proof of concept that cell replacement therapy may be a viable therapeutic approach for PD. However, the use of human fetal cells remains fraught with controversy due to fundamental ethical, practical, and clinical limitations. Groundbreaking work on human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, coupled with extensive basic research in the stem cell field offers promising potential for hPSC-based cell replacement to become a realistic treatment regimen for PD once several major issues can be successfully addressed. In this review, we will discuss the prospects and challenges of hPSC-based cell therapy for PD.
Collapse
Affiliation(s)
- Young Cha
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Tae-Yoon Park
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Pierre Leblanc
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Kwang-Soo Kim
- Department of Psychiatry and Molecular Neurobiology Laboratory, McLean Hospital and Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| |
Collapse
|
6
|
Lane EL, Lelos MJ. Defining the unknowns for cell therapies in Parkinson's disease. Dis Model Mech 2022; 15:dmm049543. [PMID: 36165848 PMCID: PMC9555765 DOI: 10.1242/dmm.049543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
First-in-human clinical trials have commenced to test the safety and efficacy of cell therapies for people with Parkinson's disease (PD). Proof of concept that this neural repair strategy is efficacious is based on decades of preclinical studies and clinical trials using primary foetal cells, as well as a significant literature exploring more novel stem cell-derived products. Although several measures of efficacy have been explored, including the successful in vitro differentiation of stem cells to dopamine neurons and consistent alleviation of motor dysfunction in rodent models, many unknowns still remain regarding the long-term clinical implications of this treatment strategy. Here, we consider some of these outstanding questions, including our understanding of the interaction between anti-Parkinsonian medication and the neural transplant, the impact of the cell therapy on cognitive or neuropsychiatric symptoms of PD, the role of neuroinflammation in the therapeutic process and the development of graft-induced dyskinesias. We identify questions that are currently pertinent to the field that require further exploration, and pave the way for a more holistic understanding of this neural repair strategy for treatment of PD.
Collapse
Affiliation(s)
- Emma L. Lane
- Cardiff School of Pharmacy and Pharmaceutical Sciences, King Edward VII Avenue, Cardiff University, Cardiff CF10 3NB, UK
| | - Mariah J. Lelos
- School of Biosciences, Museum Avenue, Cardiff University, Cardiff CF10 3AX, UK
| |
Collapse
|
7
|
McComish SF, MacMahon Copas AN, Caldwell MA. Human Brain-Based Models Provide a Powerful Tool for the Advancement of Parkinson’s Disease Research and Therapeutic Development. Front Neurosci 2022; 16:851058. [PMID: 35651633 PMCID: PMC9149087 DOI: 10.3389/fnins.2022.851058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease and affects approximately 2–3% of the population over the age of 65. PD is characterised by the loss of dopaminergic neurons from the substantia nigra, leading to debilitating motor symptoms including bradykinesia, tremor, rigidity, and postural instability. PD also results in a host of non-motor symptoms such as cognitive decline, sleep disturbances and depression. Although existing therapies can successfully manage some motor symptoms for several years, there is still no means to halt progression of this severely debilitating disorder. Animal models used to replicate aspects of PD have contributed greatly to our current understanding but do not fully replicate pathological mechanisms as they occur in patients. Because of this, there is now great interest in the use of human brain-based models to help further our understanding of disease processes. Human brain-based models include those derived from embryonic stem cells, patient-derived induced neurons, induced pluripotent stem cells and brain organoids, as well as post-mortem tissue. These models facilitate in vitro analysis of disease mechanisms and it is hoped they will help bridge the existing gap between bench and bedside. This review will discuss the various human brain-based models utilised in PD research today and highlight some of the key breakthroughs they have facilitated. Furthermore, the potential caveats associated with the use of human brain-based models will be detailed.
Collapse
Affiliation(s)
- Sarah F. McComish
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Adina N. MacMahon Copas
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Maeve A. Caldwell
- Department of Physiology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Maeve A. Caldwell,
| |
Collapse
|
8
|
Gene-independent therapeutic interventions to maintain and restore light sensitivity in degenerating photoreceptors. Prog Retin Eye Res 2022; 90:101065. [PMID: 35562270 DOI: 10.1016/j.preteyeres.2022.101065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 12/14/2022]
Abstract
Neurodegenerative retinal diseases are a prime cause of blindness in industrialized countries. In many cases, there are no therapeutic treatments, although they are essential to improve patients' quality of life. A set of disease-causing genes, which primarily affect photoreceptors, has already been identified and is of major interest for developing gene therapies. Nevertheless, depending on the nature and the state of the disease, gene-independent strategies are needed. Various strategies to halt disease progression or maintain function of the retina are under research. These therapeutic interventions include neuroprotection, direct reprogramming of affected photoreceptors, the application of non-coding RNAs, the generation of artificial photoreceptors by optogenetics and cell replacement strategies. During recent years, major breakthroughs have been made such as the first optogenetic application to a blind patient whose visual function partially recovered by targeting retinal ganglion cells. Also, RPE cell transplantation therapies are under clinical investigation and show great promise to improve visual function in blind patients. These cells are generated from human stem cells. Similar therapies for replacing photoreceptors are extensively tested in pre-clinical models. This marks just the start of promising new cures taking advantage of developments in the areas of genetic engineering, optogenetics, and stem-cell research. In this review, we present the recent therapeutic advances of gene-independent approaches that are currently under clinical evaluation. Our main focus is on photoreceptors as these sensory cells are highly vulnerable to degenerative diseases, and are crucial for light detection.
Collapse
|
9
|
Elabi OF, Pass R, Sormonta I, Nolbrant S, Drummond N, Kirkeby A, Kunath T, Parmar M, Lane EL. Human Embryonic Stem Cell-Derived Dopaminergic Grafts Alleviate L-DOPA Induced Dyskinesia. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1881-1896. [PMID: 35466951 DOI: 10.3233/jpd-212920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND First-in-human studies to test the efficacy and safety of human embryonic stem cells (hESC)-derived dopaminergic cells in the treatment of Parkinson's disease (PD) are imminent. Pre-clinical studies using hESC-derived dopamine neuron transplants in rat models have indicated that the benefits parallel those shown with fetal tissue but have thus far failed to consider how ongoing L-DOPA administration might impact on the graft. OBJECTIVE To determine whether L-DOPA impacts on survival and functional recovery following grafting of hESC-derived dopaminergic neurons. METHODS Unilateral 6-OHDA lesioned rats were administered with either saline or L-DOPA prior to, and for 18 weeks following surgical implantation of dopaminergic neural progenitors derived from RC17 hESCs according to two distinct protocols in independent laboratories. RESULTS Grafts from both protocols elicited reduction in amphetamine-induced rotations. Reduced L-DOPA-induced dyskinesia preceded the improvement in amphetamine-induced rotations. Furthermore, L-DOPA had no effect on overall survival (HuNu) or dopaminergic neuron content of the graft (TH positive cells) but did lead to an increase in the number of GIRK2 positive neurons. CONCLUSION Critically, we found that L-DOPA was not detrimental to graft function, potentially enhancing graft maturation and promoting an A9 phenotype. Early improvement of L-DOPA-induced dyskinesia suggests that grafts may support the handling of exogenously supplied dopamine earlier than improvements in amphetamine-induced behaviours indicate. Given that one of the protocols will be employed in the production of cells for the European STEM-PD clinical trial, this is vital information for the management of patients and achieving optimal outcomes following transplantation of hESC-derived grafts for PD.
Collapse
Affiliation(s)
- Osama F Elabi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Rachel Pass
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Irene Sormonta
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| | - Sara Nolbrant
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Nicola Drummond
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Agnete Kirkeby
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Neuroscience and The Novo Nordisk Foundation Center for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tilo Kunath
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Emma L Lane
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
10
|
Kusena JWT, Shariatzadeh M, Thomas RJ, Wilson SL. Understanding cell culture dynamics: a tool for defining protocol parameters for improved processes and efficient manufacturing using human embryonic stem cells. Bioengineered 2021; 12:979-996. [PMID: 33757391 PMCID: PMC8806349 DOI: 10.1080/21655979.2021.1902696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
Standardization is crucial when culturing cells including human embryonic stem cells (hESCs) which are valuable for therapy development and disease modeling. Inherent issues regarding reproducibility of protocols are problematic as they hinder translation to good manufacturing practice (GMP), thus reducing clinical efficacy and uptake. Pluripotent cultures require standardization to ensure that input material is consistent prior to differentiation, as inconsistency of input cells creates end-product variation. To improve protocols, developers first must understand the cells they are working with and their related culture dynamics. This innovative work highlights key conditions required for optimized and cost-effective bioprocesses compared to generic protocols typically implemented. This entailed investigating conditions affecting growth, metabolism, and phenotype dynamics to ensure cell quality is appropriate for use. Results revealed critical process parameters (CPPs) including feeding regime and seeding density impact critical quality attributes (CQAs) including specific metabolic rate (SMR) and specific growth rate (SGR). This implied that process understanding, and control is essential to maintain key cell characteristics, reduce process variation and retain CQAs. Examination of cell dynamics and CPPs permitted the formation of a defined protocol for culturing H9 hESCs. The authors recommend that H9 seeding densities of 20,000 cells/cm2, four-day cultures or three-day cultures following a recovery passage from cryopreservation and 100% medium exchange after 48 hours are optimal. These parameters gave ~SGR of 0.018 hour-1 ± 1.5x10-3 over three days and cell viabilities ≥95%±0.4, while producing cells which highly expressed pluripotent and proliferation markers, Oct3/4 (>99% positive) and Ki-67 (>99% positive).
Collapse
Affiliation(s)
- J W T Kusena
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| | - M Shariatzadeh
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| | - R J Thomas
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| | - S L Wilson
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough University, Loughborough, Leicestershire, UK
| |
Collapse
|
11
|
Sun Y, Feng L, Liang L, Stacey GN, Wang C, Wang Y, Hu B. Neuronal cell-based medicines from pluripotent stem cells: Development, production, and preclinical assessment. Stem Cells Transl Med 2021; 10 Suppl 2:S31-S40. [PMID: 34724724 PMCID: PMC8560198 DOI: 10.1002/sctm.20-0522] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 04/21/2021] [Accepted: 06/06/2021] [Indexed: 12/14/2022] Open
Abstract
Brain degeneration and damage is difficult to cure due to the limited endogenous repair capability of the central nervous system. Furthermore, drug development for treatment of diseases of the central nervous system remains a major challenge. However, it now appears that using human pluripotent stem cell-derived neural cells to replace degenerating cells provides a promising cell-based medicine for rejuvenation of brain function. Accordingly, a large number of studies have carried out preclinical assessments, which have involved different neural cell types in several neurological diseases. Recent advances in animal models identify the transplantation of neural derivatives from pluripotent stem cells as a promising path toward the clinical application of cell therapies [Stem Cells Transl Med 2019;8:681-693; Drug Discov Today 2019;24:992-999; Nat Med 2019;25:1045-1053]. Some groups are moving toward clinical testing in humans. However, the difficulty in selection of valuable critical quality criteria for cell products and the lack of functional assays that could indicate suitability for clinical effect continue to hinder neural cell-based medicine development [Biologicals 2019;59:68-71]. In this review, we summarize the current status of preclinical studies progress in this area and outline the biological characteristics of neural cells that have been used in new developing clinical studies. We also discuss the requirements for translation of stem cell-derived neural cells in examples of stem cell-based clinical therapy.
Collapse
Affiliation(s)
- Yun Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People's Republic of China
| | - Lin Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Lingmin Liang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Glyn N Stacey
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- International Stem Cell Banking Initiative, Barley, Hertfordshire, UK
| | - Chaoqun Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
12
|
Han F, Liu Y, Huang J, Zhang X, Wei C. Current Approaches and Molecular Mechanisms for Directly Reprogramming Fibroblasts Into Neurons and Dopamine Neurons. Front Aging Neurosci 2021; 13:738529. [PMID: 34658841 PMCID: PMC8515543 DOI: 10.3389/fnagi.2021.738529] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/27/2021] [Indexed: 12/30/2022] Open
Abstract
Parkinson's disease is mainly caused by specific degeneration of dopaminergic neurons (DA neurons) in the substantia nigra of the middle brain. Over the past two decades, transplantation of neural stem cells (NSCs) from fetal brain-derived neural stem cells (fNSCs), human embryonic stem cells (hESCs), and induced pluripotent stem cells (iPSCs) has been shown to improve the symptoms of motor dysfunction in Parkinson's disease (PD) animal models and PD patients significantly. However, there are ethical concerns with fNSCs and hESCs and there is an issue of rejection by the immune system, and the iPSCs may involve tumorigenicity caused by the integration of the transgenes. Recent studies have shown that somatic fibroblasts can be directly reprogrammed to NSCs, neurons, and specific dopamine neurons. Directly induced neurons (iN) or induced DA neurons (iDANs) from somatic fibroblasts have several advantages over iPSC cells. The neurons produced by direct transdifferentiation do not pass through a pluripotent state. Therefore, direct reprogramming can generate patient-specific cells, and it can overcome the safety problems of rejection by the immune system and teratoma formation related to hESCs and iPSCs. However, there are some critical issues such as the low efficiency of direct reprogramming, biological functions, and risks from the directly converted neurons, which hinder their clinical applications. Here, the recent progress in methods, mechanisms, and future challenges of directly reprogramming somatic fibroblasts into neurons or dopamine neurons were summarized to speed up the clinical translation of these directly converted neural cells to treat PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabin Han
- Innovation Institute for Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shenzhen Research Institute of Shandong University, Jinan, China.,The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Yanming Liu
- Shenzhen Research Institute of Shandong University, Jinan, China.,The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| | - Jin Huang
- Laboratory of Basic Medical Research, Medical Centre of PLA Strategic Support Force, Beijing, China
| | - Xiaoping Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanfei Wei
- The Institute for Tissue Engineering and Regenerative Medicine, Liaocheng University/Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
13
|
|
14
|
Dhoke NR, Kim H, Selvaraj S, Azzag K, Zhou H, Oliveira NAJ, Tungtur S, Ortiz-Cordero C, Kiley J, Lu QL, Bang AG, Perlingeiro RCR. A universal gene correction approach for FKRP-associated dystroglycanopathies to enable autologous cell therapy. Cell Rep 2021; 36:109360. [PMID: 34260922 PMCID: PMC8327854 DOI: 10.1016/j.celrep.2021.109360] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/13/2021] [Accepted: 06/17/2021] [Indexed: 01/24/2023] Open
Abstract
Mutations in the fukutin-related protein (FKRP) gene result in a broad spectrum of muscular dystrophy (MD) phenotypes, including the severe Walker-Warburg syndrome (WWS). Here, we develop a gene-editing approach that replaces the entire mutant open reading frame with the wild-type sequence to universally correct all FKRP mutations. We apply this approach to correct FKRP mutations in induced pluripotent stem (iPS) cells derived from patients displaying broad clinical severity. Our findings show rescue of functional α-dystroglycan (α-DG) glycosylation in gene-edited WWS iPS cell-derived myotubes. Transplantation of gene-corrected myogenic progenitors in the FKRPP448L-NSG mouse model gives rise to myofiber and satellite cell engraftment and, importantly, restoration of α-DG functional glycosylation in vivo. These findings suggest the potential feasibility of using CRISPR-Cas9 technology in combination with patient-specific iPS cells for the future development of autologous cell transplantation for FKRP-associated MDs.
Collapse
Affiliation(s)
- Neha R Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Hyunkee Kim
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Haowen Zhou
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Nelio A J Oliveira
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sudheer Tungtur
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Carolina Ortiz-Cordero
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James Kiley
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Qi Long Lu
- McColl-Lockwood Laboratory for Muscular Dystrophy Research, Cannon Research Center, Carolinas Medical Center, Atrium Health, Charlotte, NC, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
15
|
Aguila J, Cheng S, Kee N, Cao M, Wang M, Deng Q, Hedlund E. Spatial RNA Sequencing Identifies Robust Markers of Vulnerable and Resistant Human Midbrain Dopamine Neurons and Their Expression in Parkinson's Disease. Front Mol Neurosci 2021; 14:699562. [PMID: 34305528 PMCID: PMC8297217 DOI: 10.3389/fnmol.2021.699562] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/08/2021] [Indexed: 01/26/2023] Open
Abstract
Defining transcriptional profiles of substantia nigra pars compacta (SNc) and ventral tegmental area (VTA) dopamine neurons is critical to understanding their differential vulnerability in Parkinson’s Disease (PD). Here, we determine transcriptomes of human SNc and VTA dopamine neurons using LCM-seq on a large sample cohort. We apply a bootstrapping strategy as sample input to DESeq2 and identify 33 stably differentially expressed genes (DEGs) between these two subpopulations. We also compute a minimal sample size for identification of stable DEGs, which highlights why previous reported profiles from small sample sizes display extensive variability. Network analysis reveal gene interactions unique to each subpopulation and highlight differences in regulation of mitochondrial stability, apoptosis, neuronal survival, cytoskeleton regulation, extracellular matrix modulation as well as synapse integrity, which could explain the relative resilience of VTA dopamine neurons. Analysis of PD tissues showed that while identified stable DEGs can distinguish the subpopulations also in disease, the SNc markers SLIT1 and ATP2A3 were down-regulated and thus appears to be biomarkers of disease. In summary, our study identifies human SNc and VTA marker profiles, which will be instrumental for studies aiming to modulate dopamine neuron resilience and to validate cell identity of stem cell-derived dopamine neurons.
Collapse
Affiliation(s)
- Julio Aguila
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Shangli Cheng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Nigel Kee
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ming Cao
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Menghan Wang
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Hedlund
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| |
Collapse
|
16
|
Human stem cells harboring a suicide gene improve the safety and standardisation of neural transplants in Parkinsonian rats. Nat Commun 2021; 12:3275. [PMID: 34045451 PMCID: PMC8160354 DOI: 10.1038/s41467-021-23125-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 04/14/2021] [Indexed: 12/28/2022] Open
Abstract
Despite advancements in human pluripotent stem cells (hPSCs) differentiation protocols to generate appropriate neuronal progenitors suitable for transplantation in Parkinson's disease, resultant grafts contain low proportions of dopamine neurons. Added to this is the tumorigenic risk associated with the potential presence of incompletely patterned, proliferative cells within grafts. Here, we utilised a hPSC line carrying a FailSafeTM suicide gene (thymidine kinase linked to cyclinD1) to selectively ablate proliferative cells in order to improve safety and purity of neural transplantation in a Parkinsonian model. The engineered FailSafeTM hPSCs demonstrated robust ventral midbrain specification in vitro, capable of forming neural grafts upon transplantation. Activation of the suicide gene within weeks after transplantation, by ganciclovir administration, resulted in significantly smaller grafts without affecting the total yield of dopamine neurons, their capacity to innervate the host brain or reverse motor deficits at six months in a rat Parkinsonian model. Within ganciclovir-treated grafts, other neuronal, glial and non-neural populations (including proliferative cells), were significantly reduced-cell types that may pose adverse or unknown influences on graft and host function. These findings demonstrate the capacity of a suicide gene-based system to improve both the standardisation and safety of hPSC-derived grafts in a rat model of Parkinsonism.
Collapse
|
17
|
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]
|
18
|
Adler AF, Cardoso T, Nolbrant S, Mattsson B, Hoban DB, Jarl U, Wahlestedt JN, Grealish S, Björklund A, Parmar M. hESC-Derived Dopaminergic Transplants Integrate into Basal Ganglia Circuitry in a Preclinical Model of Parkinson's Disease. Cell Rep 2020; 28:3462-3473.e5. [PMID: 31553914 PMCID: PMC6899556 DOI: 10.1016/j.celrep.2019.08.058] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/30/2019] [Accepted: 08/19/2019] [Indexed: 12/20/2022] Open
Abstract
Cell replacement is currently being explored as a therapeutic approach for neurodegenerative disease. Using stem cells as a source, transplantable progenitors can now be generated under conditions compliant with clinical application in patients. In this study, we elucidate factors controlling target-appropriate innervation and circuitry integration of human embryonic stem cell (hESC)-derived grafts after transplantation to the adult brain. We show that cell-intrinsic factors determine graft-derived axonal innervation, whereas synaptic inputs from host neurons primarily reflect the graft location. Furthermore, we provide evidence that hESC-derived dopaminergic grafts transplanted in a long-term preclinical rat model of Parkinson’s disease (PD) receive synaptic input from subtypes of host cortical, striatal, and pallidal neurons that are known to regulate the function of endogenous nigral dopamine neurons. This refined understanding of how graft neurons integrate with host circuitry will be important for the design of clinical stem-cell-based replacement therapies for PD, as well as for other neurodegenerative diseases. Pattern of graft-derived innervation is determined by phenotype of grafted cells Synaptic inputs from host-to-graft depend on location of graft Intrastriatal dopaminergic grafts receive correct excitatory and inhibitory host inputs Individual host neurons provide inputs to both dopaminergic grafts and the host nigra
Collapse
Affiliation(s)
- Andrew F Adler
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden; Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Tiago Cardoso
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden; Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Sara Nolbrant
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden; Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Bengt Mattsson
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Deirdre B Hoban
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden; Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Ulla Jarl
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Jenny Nelander Wahlestedt
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden; Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
| | - Shane Grealish
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Anders Björklund
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 22184 Lund, Sweden; Lund Stem Cell Center, Lund University, 22184 Lund, Sweden.
| |
Collapse
|
19
|
Jang SE, Qiu L, Chan LL, Tan EK, Zeng L. Current Status of Stem Cell-Derived Therapies for Parkinson's Disease: From Cell Assessment and Imaging Modalities to Clinical Trials. Front Neurosci 2020; 14:558532. [PMID: 33177975 PMCID: PMC7596695 DOI: 10.3389/fnins.2020.558532] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/17/2020] [Indexed: 12/23/2022] Open
Abstract
Curative therapies or treatments reversing the progression of Parkinson’s disease (PD) have attracted considerable interest in the last few decades. PD is characterized by the gradual loss of dopaminergic (DA) neurons and decreased striatal dopamine levels. Current challenges include optimizing neuroprotective strategies, developing personalized drug therapy, and minimizing side effects from the long-term prescription of pharmacological drugs used to relieve short-term motor symptoms. Transplantation of DA cells into PD patients’ brains to replace degenerated DA has the potential to change the treatment paradigm. Herein, we provide updates on current progress in stem cell-derived DA neuron transplantation as a therapeutic alternative for PD. We briefly highlight cell sources for transplantation and focus on cell assessment methods such as identification of genetic markers, single-cell sequencing, and imaging modalities used to access cell survival and function. More importantly, we summarize clinical reports of patients who have undergone cell-derived transplantation in PD to better perceive lessons that can be drawn from past and present clinical outcomes. Modifying factors include (1) source of the stem cells, (2) quality of the stem cells, (3) age of the patient, (4) stage of disease progression at the time of cell therapy, (5) surgical technique/practices, and (6) the use of immunosuppression. We await the outcomes of joint efforts in clinical trials around the world such as NYSTEM and CiRA to further guide us in the selection of the most suitable parameters for cell-based neurotransplantation in PD.
Collapse
Affiliation(s)
- Se Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Ling Ling Chan
- Department of Diagnostic Radiology, Singapore General Hospital, Singapore, Singapore.,Neuroscience & Behavioral Disorders Program, Duke University and National University of Singapore (DUKE-NUS), Graduate Medical School, Singapore, Singapore
| | - Eng-King Tan
- Neuroscience & Behavioral Disorders Program, Duke University and National University of Singapore (DUKE-NUS), Graduate Medical School, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore General Hospital Campus, Singapore, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore.,Neuroscience & Behavioral Disorders Program, Duke University and National University of Singapore (DUKE-NUS), Graduate Medical School, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Novena Campus, Singapore, Singapore
| |
Collapse
|
20
|
Fonseca AC, Melchels FPW, Ferreira MJS, Moxon SR, Potjewyd G, Dargaville TR, Kimber SJ, Domingos M. Emulating Human Tissues and Organs: A Bioprinting Perspective Toward Personalized Medicine. Chem Rev 2020; 120:11128-11174. [PMID: 32937071 PMCID: PMC7645917 DOI: 10.1021/acs.chemrev.0c00342] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Indexed: 02/06/2023]
Abstract
The lack of in vitro tissue and organ models capable of mimicking human physiology severely hinders the development and clinical translation of therapies and drugs with higher in vivo efficacy. Bioprinting allow us to fill this gap and generate 3D tissue analogues with complex functional and structural organization through the precise spatial positioning of multiple materials and cells. In this review, we report the latest developments in terms of bioprinting technologies for the manufacturing of cellular constructs with particular emphasis on material extrusion, jetting, and vat photopolymerization. We then describe the different base polymers employed in the formulation of bioinks for bioprinting and examine the strategies used to tailor their properties according to both processability and tissue maturation requirements. By relating function to organization in human development, we examine the potential of pluripotent stem cells in the context of bioprinting toward a new generation of tissue models for personalized medicine. We also highlight the most relevant attempts to engineer artificial models for the study of human organogenesis, disease, and drug screening. Finally, we discuss the most pressing challenges, opportunities, and future prospects in the field of bioprinting for tissue engineering (TE) and regenerative medicine (RM).
Collapse
Affiliation(s)
- Ana Clotilde Fonseca
- Centre
for Mechanical Engineering, Materials and Processes, Department of
Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Polo II, 3030-790 Coimbra, Portugal
| | - Ferry P. W. Melchels
- Institute
of Biological Chemistry, Biophysics and Bioengineering, School of
Engineering and Physical Sciences, Heriot-Watt
University, Edinburgh EH14 4AS, U.K.
| | - Miguel J. S. Ferreira
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| | - Samuel R. Moxon
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Geoffrey Potjewyd
- Division
of Neuroscience and Experimental Psychology, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Tim R. Dargaville
- Institute
of Health and Biomedical Innovation, Science and Engineering Faculty, Queensland University of Technology, Queensland 4001, Australia
| | - Susan J. Kimber
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, U.K.
| | - Marco Domingos
- Department
of Mechanical, Aerospace and Civil Engineering, School of Engineering,
Faculty of Science and Engineering, The
University of Manchester, Manchester M13 9PL, U.K.
| |
Collapse
|
21
|
Chen Y, Kunath T, Simpson J, Homer N, Sylantyev S. Synaptic signalling in a network of dopamine neurons: what prevents proper intercellular crosstalk? FEBS Lett 2020; 594:3272-3292. [PMID: 33073864 DOI: 10.1002/1873-3468.13910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 01/09/2023]
Abstract
Human embryonic stem cell (hESC)-derived midbrain dopamine (DA) neurons stand out as a cell source for transplantation with their sustainability and consistency superior to the formerly used fetal tissues. However, multiple studies of DA neurons in culture failed to register action potential (AP) generation upon synaptic input. To test whether this is due to deficiency of NMDA receptor (NMDAR) coagonists released from astroglia, we studied the functional properties of neural receptors in hESC-derived DA neuronal cultures. We find that, apart from an insufficient amount of coagonists, lack of interneuronal crosstalk is caused by hypofunction of synaptic NMDARs due to their direct inhibition by synaptically released DA. This inhibitory tone is independent of DA receptors and affects the NMDAR coagonist binding site.
Collapse
Affiliation(s)
- Yixi Chen
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, University of Edinburgh, Edinburgh, UK.,UK Centre for Mammalian Synthetic Biology, University of Edinburgh, Edinburgh, UK
| | - Tilo Kunath
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, University of Edinburgh, Edinburgh, UK.,UK Centre for Mammalian Synthetic Biology, University of Edinburgh, Edinburgh, UK
| | - Joanna Simpson
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Natalie Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
22
|
Paniza T, Deshpande M, Wang N, O’Neil R, Zuccaro MV, Smith ME, Madireddy A, James D, Ecker J, Rosenwaks Z, Egli D, Gerhardt J. Pluripotent stem cells with low differentiation potential contain incompletely reprogrammed DNA replication. J Cell Biol 2020; 219:e201909163. [PMID: 32673399 PMCID: PMC7480103 DOI: 10.1083/jcb.201909163] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/26/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Reprogrammed pluripotent stem cells (PSCs) are valuable for research and potentially for cell replacement therapy. However, only a fraction of reprogrammed PSCs are developmentally competent. Genomic stability and accurate DNA synthesis are fundamental for cell development and critical for safety. We analyzed whether defects in DNA replication contribute to genomic instability and the diverse differentiation potentials of reprogrammed PSCs. Using a unique single-molecule approach, we visualized DNA replication in isogenic PSCs generated by different reprogramming approaches, either somatic cell nuclear transfer (NT-hESCs) or with defined factors (iPSCs). In PSCs with lower differentiation potential, DNA replication was incompletely reprogrammed, and genomic instability increased during replicative stress. Reprogramming of DNA replication did not correlate with DNA methylation. Instead, fewer replication origins and a higher frequency of DNA breaks in PSCs with incompletely reprogrammed DNA replication were found. Given the impact of error-free DNA synthesis on the genomic integrity and differentiation proficiency of PSCs, analyzing DNA replication may be a useful quality control tool.
Collapse
Affiliation(s)
- Theodore Paniza
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY
| | - Madhura Deshpande
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY
| | - Ning Wang
- Department of Pediatrics, Columbia University, New York, NY
| | - Ryan O’Neil
- Plant Molecular and Cellular Biology Laboratory, Salk Institute, La Jolla, CA
| | - Michael V. Zuccaro
- Department of Pediatrics, Columbia University, New York, NY
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | | | - Advaitha Madireddy
- Department of Pediatric Hematology/Oncology, Rutgers University, New Brunswick, NJ
| | - Daylon James
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY
| | - Joseph Ecker
- Plant Molecular and Cellular Biology Laboratory, Salk Institute, La Jolla, CA
| | - Zev Rosenwaks
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY
| | - Dieter Egli
- Department of Pediatrics, Columbia University, New York, NY
| | - Jeannine Gerhardt
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY
| |
Collapse
|
23
|
Ásgrímsdóttir ES, Arenas E. Midbrain Dopaminergic Neuron Development at the Single Cell Level: In vivo and in Stem Cells. Front Cell Dev Biol 2020; 8:463. [PMID: 32733875 PMCID: PMC7357704 DOI: 10.3389/fcell.2020.00463] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that predominantly affects dopaminergic (DA) neurons of the substantia nigra. Current treatment options for PD are symptomatic and typically involve the replacement of DA neurotransmission by DA drugs, which relieve the patients of some of their motor symptoms. However, by the time of diagnosis, patients have already lost about 70% of their substantia nigra DA neurons and these drugs offer only temporary relief. Therefore, cell replacement therapy has garnered much interest as a potential treatment option for PD. Early studies using human fetal tissue for transplantation in PD patients provided proof of principle for cell replacement therapy, but they also highlighted the ethical and practical difficulties associated with using human fetal tissue as a cell source. In recent years, advancements in stem cell research have made human pluripotent stem cells (hPSCs) an attractive source of material for cell replacement therapy. Studies on how DA neurons are specified and differentiated in the developing mouse midbrain have allowed us to recapitulate many of the positional and temporal cues needed to generate DA neurons in vitro. However, little is known about the developmental programs that govern human DA neuron development. With the advent of single-cell RNA sequencing (scRNA-seq) and bioinformatics, it has become possible to analyze precious human samples with unprecedented detail and extract valuable high-quality information from large data sets. This technology has allowed the systematic classification of cell types present in the human developing midbrain along with their gene expression patterns. By studying human development in such an unbiased manner, we can begin to elucidate human DA neuron development and determine how much it differs from our knowledge of the rodent brain. Importantly, this molecular description of the function of human cells has become and will increasingly be a reference to define, evaluate, and engineer cell types for PD cell replacement therapy and disease modeling.
Collapse
Affiliation(s)
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Muñoz A, Lopez-Lopez A, Labandeira CM, Labandeira-Garcia JL. Interactions Between the Serotonergic and Other Neurotransmitter Systems in the Basal Ganglia: Role in Parkinson's Disease and Adverse Effects of L-DOPA. Front Neuroanat 2020; 14:26. [PMID: 32581728 PMCID: PMC7289026 DOI: 10.3389/fnana.2020.00026] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. However, other non-dopaminergic neuronal systems such as the serotonergic system are also involved. Serotonergic dysfunction is associated with non-motor symptoms and complications, including anxiety, depression, dementia, and sleep disturbances. This pathology reduces patient quality of life. Interaction between the serotonergic and other neurotransmitters systems such as dopamine, noradrenaline, glutamate, and GABA controls the activity of striatal neurons and are particularly interesting for understanding the pathophysiology of PD. Moreover, serotonergic dysfunction also causes motor symptoms. Interestingly, serotonergic neurons play an important role in the effects of L-DOPA in advanced PD stages. Serotonergic terminals can convert L-DOPA to dopamine, which mediates dopamine release as a "false" transmitter. The lack of any autoregulatory feedback control in serotonergic neurons to regulate L-DOPA-derived dopamine release contributes to the appearance of L-DOPA-induced dyskinesia (LID). This mechanism may also be involved in the development of graft-induced dyskinesias (GID), possibly due to the inclusion of serotonin neurons in the grafted tissue. Consistent with this, the administration of serotonergic agonists suppressed LID. In this review article, we summarize the interactions between the serotonergic and other systems. We also discuss the role of the serotonergic system in LID and if therapeutic approaches specifically targeting this system may constitute an effective strategy in PD.
Collapse
Affiliation(s)
- Ana Muñoz
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Deptartment of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Andrea Lopez-Lopez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Deptartment of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Carmen M Labandeira
- Department of Clinical Neurology, Hospital Alvaro Cunqueiro, University Hospital Complex, Vigo, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Deptartment of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| |
Collapse
|
25
|
Henchcliffe C, Sarva H. Restoring Function to Dopaminergic Neurons: Progress in the Development of Cell-Based Therapies for Parkinson's Disease. CNS Drugs 2020; 34:559-577. [PMID: 32472450 DOI: 10.1007/s40263-020-00727-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is escalating interest in cell-based therapies to restore lost dopamine inputs in Parkinson's disease. This is based upon the rationale that implanting dopamine progenitors into the striatum can potentially improve dopamine-responsive motor symptoms. A rich body of data describing clinical trials of previous cell transplantation exists. These have included multiple cell sources for transplantation including allogeneic (human embryonic mesencephalic tissue, retinal pigment epithelial cells) and autologous (carotid body, adrenal medullary tissue) cells, as well as xenotransplantation. However, there are multiple limitations related to these cell sources, including availability of adequate numbers of cells for transplant, heterogeneity within cells transplanted, imprecisely defined mechanisms of action, and poor cell survival after transplantation in some cases. Nonetheless, evidence has accrued from a subset of trials to support the rationale for such a regenerative approach. Recent rapid advances in stem cell technology may now overcome these prior limitations. For example, dopamine neuron precursor cells for transplant can be generated from induced pluripotent cells and human embryonic stem cells. The benefits of these innovative approaches include: the possibility of scalability; a high degree of quality control; and improved understanding of mechanisms of action with rigorous preclinical testing. In this review, we focus on the potential for cell-based therapies in Parkinson's disease to restore the function of dopaminergic neurons, we critically review previous attempts to harness such strategies, we discuss potential benefits and predicted limitations, and we address how previous roadblocks may be overcome to bring a cell-based approach to the clinic.
Collapse
Affiliation(s)
- Claire Henchcliffe
- Department of Neurology, Weill Medical College of Cornell University, 428 East 72nd Street, Suite 400, New York, NY, 10021, USA.
| | - Harini Sarva
- Department of Neurology, Weill Medical College of Cornell University, 428 East 72nd Street, Suite 400, New York, NY, 10021, USA
| |
Collapse
|
26
|
Björklund A, Parmar M. Neuronal Replacement as a Tool for Basal Ganglia Circuitry Repair: 40 Years in Perspective. Front Cell Neurosci 2020; 14:146. [PMID: 32547369 PMCID: PMC7272540 DOI: 10.3389/fncel.2020.00146] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/30/2020] [Indexed: 01/07/2023] Open
Abstract
The ability of new neurons to promote repair of brain circuitry depends on their capacity to re-establish afferent and efferent connections with the host. In this review article, we give an overview of past and current efforts to restore damaged connectivity in the adult mammalian brain using implants of fetal neuroblasts or stem cell-derived neuronal precursors, with a focus on strategies aimed to repair damaged basal ganglia circuitry induced by lesions that mimic the pathology seen in humans affected by Parkinson’s or Huntington’s disease. Early work performed in rodents showed that neuroblasts obtained from striatal primordia or fetal ventral mesencephalon can become anatomically and functionally integrated into lesioned striatal and nigral circuitry, establish afferent and efferent connections with the lesioned host, and reverse the lesion-induced behavioral impairments. Recent progress in the generation of striatal and nigral progenitors from pluripotent stem cells have provided compelling evidence that they can survive and mature in the lesioned brain and re-establish afferent and efferent axonal connectivity with a remarkable degree of specificity. The studies of cell-based circuitry repair are now entering a new phase. The introduction of genetic and virus-based techniques for brain connectomics has opened entirely new possibilities for studies of graft-host integration and connectivity, and the access to more refined experimental techniques, such as chemo- and optogenetics, has provided new powerful tools to study the capacity of grafted neurons to impact the function of the host brain. Progress in this field will help to guide the efforts to develop therapeutic strategies for cell-based repair in Huntington’s and Parkinson’s disease and other neurodegenerative conditions involving damage to basal ganglia circuitry.
Collapse
Affiliation(s)
- Anders Björklund
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
27
|
Osborn TM, Hallett PJ, Schumacher JM, Isacson O. Advantages and Recent Developments of Autologous Cell Therapy for Parkinson's Disease Patients. Front Cell Neurosci 2020; 14:58. [PMID: 32317934 PMCID: PMC7147334 DOI: 10.3389/fncel.2020.00058] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/27/2020] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s Disease (PD) is a progressive degenerative disease characterized by tremor, bradykinesia, rigidity and postural instability. There are approximately 7–10 million PD patients worldwide. Currently, there are no biomarkers available or pharmaceuticals that can halt the dopaminergic neuron degeneration. At the time of diagnosis about 60% of the midbrain dopamine (mDA) neurons have already degenerated, resulting in a depletion of roughly 70% of striatal dopamine (DA) levels and synapses. Symptomatic treatment (e.g., with L-dopa) can initially restore DA levels and motor function, but with time often lead to side-effects like dyskinesia. Deep-brain-stimulation can alleviate these side-effects and some of the motor symptoms but requires repeat procedures and adds limitations for the patients. Restoration of dopaminergic synapses using neuronal cell replacement therapy has shown benefit in clinical studies using cells from fetal ventral midbrain. This approach, if done correctly, increases DA levels and restores synapses, allowing biofeedback regulation between the grafted cells and the host brain. Drawbacks are that it is not scalable for a large patient population and the patients require immunosuppression. Stem cells differentiated in vitro to mDA neurons or progenitors have shown promise in animal studies and is a scalable approach that allows for cryopreservation of transplantable cells and rigorous quality control prior to transplantation. However, all allogeneic grafts require immunosuppression. HLA-donor-matching, reduces, but does not completely eliminate, the need for immunosuppression, and is currently investigated in a clinical trial for PD in Japan. Since immune compatibility is very important in all areas of transplantation, these approaches may ultimately be of less benefit to the patients than an autologous approach. By using the patient’s own somatic cells, reprogrammed to induced pluripotent stem cells (iPSCs) and differentiated to mDA neurons immunosuppression is not required, and may also present with several biological and functional advantages in the patients, as described in this article. The proof-of-principle of autologous iPSC mDA restoration of function has been shown in parkinsonian non-human primates (NHPs), and this can now be investigated in clinical trials in addition to the allogeneic and HLA-matched approaches. In this review, we focus on the autologous approach of cell therapy for PD.
Collapse
Affiliation(s)
- Teresia M Osborn
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, Belmont, MA, United States
| | - Penelope J Hallett
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, Belmont, MA, United States
| | - James M Schumacher
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, Belmont, MA, United States
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, Belmont, MA, United States
| |
Collapse
|
28
|
Jefri M, Bell S, Peng H, Hettige N, Maussion G, Soubannier V, Wu H, Silveira H, Theroux JF, Moquin L, Zhang X, Aouabed Z, Krishnan J, O'Leary LA, Antonyan L, Zhang Y, McCarty V, Mechawar N, Gratton A, Schuppert A, Durcan TM, Fon EA, Ernst C. Stimulation of L-type calcium channels increases tyrosine hydroxylase and dopamine in ventral midbrain cells induced from somatic cells. Stem Cells Transl Med 2020; 9:697-712. [PMID: 32154672 PMCID: PMC7214648 DOI: 10.1002/sctm.18-0180] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/03/2020] [Indexed: 01/29/2023] Open
Abstract
Making high‐quality dopamine (DA)‐producing cells for basic biological or small molecule screening studies is critical for the development of novel therapeutics for disorders of the ventral midbrain. Currently, many ventral midbrain assays have low signal‐to‐noise ratio due to low levels of cellular DA and the rate‐limiting enzyme of DA synthesis, tyrosine hydroxylase (TH), hampering discovery efforts. Using intensively characterized ventral midbrain cells derived from human skin, which demonstrate calcium pacemaking activity and classical electrophysiological properties, we show that an L‐type calcium agonist can significantly increase TH protein levels and DA content and release. Live calcium imaging suggests that it is the immediate influx of calcium occurring simultaneously in all cells that drives this effect. Genome‐wide expression profiling suggests that L‐type calcium channel stimulation has a significant effect on specific genes related to DA synthesis and affects expression of L‐type calcium receptor subunits from the CACNA1 and CACNA2D families. Together, our findings provide an advance in the ability to increase DA and TH levels to improve the accuracy of disease modeling and small molecule screening for disorders of the ventral midbrain, including Parkinson's disease.
Collapse
Affiliation(s)
- Malvin Jefri
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Scott Bell
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Huashan Peng
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Nuwan Hettige
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Gilles Maussion
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Vincent Soubannier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Hanrong Wu
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Heika Silveira
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Jean-Francois Theroux
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Luc Moquin
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Xin Zhang
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Zahia Aouabed
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Jeyashree Krishnan
- Institute for Computational Biomedicine, Aachen University, Aachen, Germany
| | - Liam A O'Leary
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | | | - Ying Zhang
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Vincent McCarty
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Naguib Mechawar
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Alain Gratton
- Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| | - Andreas Schuppert
- Institute for Computational Biomedicine, Aachen University, Aachen, Germany
| | - Thomas M Durcan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Carl Ernst
- Psychiatric Genetics Group, McGill University, Montreal, Quebec, Canada.,Department of Psychiatry, McGill University and Douglas Hospital Research Institute, Montreal, Quebec, Canada
| |
Collapse
|
29
|
Cherkashova EA, Leonov GE, Namestnikova DD, Solov'eva AA, Gubskii IL, Bukharova TB, Gubskii LV, Goldstein DV, Yarygin KN. Methods of Generation of Induced Pluripotent Stem Cells and Their Application for the Therapy of Central Nervous System Diseases. Bull Exp Biol Med 2020; 168:566-573. [PMID: 32157511 DOI: 10.1007/s10517-020-04754-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Indexed: 12/12/2022]
Abstract
The use of induced pluripotent stem cells (IPSC) is a promising approach to the therapy of CNS diseases. The undeniable advantage of IPSC technology is the possibility of obtaining practically all types of somatic cells for autologous transplantation bypassing bioethical problems. The review presents integrative and non-integrative methods for obtaining IPSC and the ways of their in vitro and in vivo application for the study and treatment of neurological diseases.
Collapse
Affiliation(s)
- E A Cherkashova
- Federal Center for Cerebrovascular Pathology and Stroke, Ministry of Health of Russian Federation, Moscow, Russia
| | - G E Leonov
- N. P. Bochkov Research Center for Medical Genetics, Moscow, Russia.
| | - D D Namestnikova
- N. I. Pirogov Russian National Research Medical University, Ministry of Health of Russian Federation, Moscow, Russia
| | - A A Solov'eva
- Federal Center for Cerebrovascular Pathology and Stroke, Ministry of Health of Russian Federation, Moscow, Russia
| | - I L Gubskii
- Federal Center for Cerebrovascular Pathology and Stroke, Ministry of Health of Russian Federation, Moscow, Russia
| | - T B Bukharova
- N. P. Bochkov Research Center for Medical Genetics, Moscow, Russia
| | - L V Gubskii
- Federal Center for Cerebrovascular Pathology and Stroke, Ministry of Health of Russian Federation, Moscow, Russia.,N. I. Pirogov Russian National Research Medical University, Ministry of Health of Russian Federation, Moscow, Russia
| | - D V Goldstein
- N. P. Bochkov Research Center for Medical Genetics, Moscow, Russia
| | - K N Yarygin
- V. N. Orekhovich Research Institute of Biomedical Chemistry, Moscow, Russia.,Russian Medical Academy for Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
30
|
Parmar M, Grealish S, Henchcliffe C. The future of stem cell therapies for Parkinson disease. Nat Rev Neurosci 2020; 21:103-115. [DOI: 10.1038/s41583-019-0257-7] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2019] [Indexed: 01/07/2023]
|
31
|
Selvaraj S, Dhoke NR, Kiley J, Mateos-Aierdi AJ, Tungtur S, Mondragon-Gonzalez R, Killeen G, Oliveira VKP, López de Munain A, Perlingeiro RCR. Gene Correction of LGMD2A Patient-Specific iPSCs for the Development of Targeted Autologous Cell Therapy. Mol Ther 2019; 27:2147-2157. [PMID: 31501033 PMCID: PMC6904833 DOI: 10.1016/j.ymthe.2019.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 08/18/2019] [Accepted: 08/21/2019] [Indexed: 01/25/2023] Open
Abstract
Limb girdle muscular dystrophy type 2A (LGMD2A), caused by mutations in the Calpain 3 (CAPN3) gene, is an incurable autosomal recessive disorder that results in muscle wasting and loss of ambulation. To test the feasibility of an autologous induced pluripotent stem cell (iPSC)-based therapy for LGMD2A, here we applied CRISPR-Cas9-mediated genome editing to iPSCs from three LGMD2A patients to enable correction of mutations in the CAPN3 gene. Using a gene knockin approach, we genome edited iPSCs carrying three different CAPN3 mutations, and we demonstrated the rescue of CAPN3 protein in myotube derivatives in vitro. Transplantation of gene-corrected LGMD2A myogenic progenitors in a novel mouse model combining immunodeficiency and a lack of CAPN3 resulted in muscle engraftment and rescue of the CAPN3 mRNA. Thus, we provide here proof of concept for the integration of genome editing and iPSC technologies to develop a novel autologous cell therapy for LGMD2A.
Collapse
MESH Headings
- Animals
- Calpain/physiology
- Cell- and Tissue-Based Therapy/methods
- Cells, Cultured
- Humans
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Male
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/physiology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophies, Limb-Girdle/genetics
- Muscular Dystrophies, Limb-Girdle/pathology
- Muscular Dystrophies, Limb-Girdle/therapy
- Mutation
- Transplantation, Autologous
Collapse
Affiliation(s)
- Sridhar Selvaraj
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Neha R Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - James Kiley
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alba Judith Mateos-Aierdi
- Neurosciences Department, Biodonostia Research Institute-University of the Basque Country UPV-EHU, San Sebastián 20014, Spain; CIBERNED, Institute Carlos III, Madrid 28029, Spain
| | - Sudheer Tungtur
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ricardo Mondragon-Gonzalez
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), 07360 Ciudad de México, Mexico
| | - Grace Killeen
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vanessa K P Oliveira
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Adolfo López de Munain
- Neurosciences Department, Biodonostia Research Institute-University of the Basque Country UPV-EHU, San Sebastián 20014, Spain; CIBERNED, Institute Carlos III, Madrid 28029, Spain
| | - Rita C R Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
32
|
Balestrino R, Schapira A. Parkinson disease. Eur J Neurol 2019; 27:27-42. [DOI: 10.1111/ene.14108] [Citation(s) in RCA: 382] [Impact Index Per Article: 76.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Affiliation(s)
- R. Balestrino
- Department of Neuroscience University of Turin Turin Italy
| | - A.H.V. Schapira
- Department of Clinical and Movement Neurosciences UCL Queen Square Institute of Neurology London UK
| |
Collapse
|
33
|
Leitner D, Ramamoorthy M, Dejosez M, Zwaka TP. Immature mDA neurons ameliorate motor deficits in a 6-OHDA Parkinson's disease mouse model and are functional after cryopreservation. Stem Cell Res 2019; 41:101617. [PMID: 31731178 DOI: 10.1016/j.scr.2019.101617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/05/2019] [Accepted: 10/10/2019] [Indexed: 01/12/2023] Open
Abstract
Parkinson's disease is associated with the loss of dopaminergic neurons in the midbrain. Clinical studies investigating replacement of these neurons with in vitro-generated neurons are currently underway. However, this approach has been limited by difficulties in scaling up on-demand production of midbrain dopaminergic (mDA) neurons from pluripotent stem cells. Cryo-preservation may offer a solution, as it allows for banking of quality controlled mDA neurons. In this study, we tested different freezing conditions and found that optimal cryopreservation of immature human mDA neurons at an early differentiation time point was achieved in STEM-CELLBANKER medium using a controlled freezing program.
Collapse
Affiliation(s)
- Dominique Leitner
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai Icahn School of Medicine, New York, NY 10029, United States; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Huffington Foundation Center for Cell-Based Research in Parkinson's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Mahesh Ramamoorthy
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai Icahn School of Medicine, New York, NY 10029, United States; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Huffington Foundation Center for Cell-Based Research in Parkinson's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Marion Dejosez
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai Icahn School of Medicine, New York, NY 10029, United States; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Huffington Foundation Center for Cell-Based Research in Parkinson's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Thomas P Zwaka
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai Icahn School of Medicine, New York, NY 10029, United States; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Huffington Foundation Center for Cell-Based Research in Parkinson's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
34
|
Kusena JWT, Thomas RJ, McCall MJ, Wilson SL. From protocol to product: ventral midbrain dopaminergic neuron differentiation for the treatment of Parkinson's disease. Regen Med 2019; 14:1057-1069. [DOI: 10.2217/rme-2019-0076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Current cell therapy product limitations include the need for in-depth product understanding to ensure product potency, safety and purity. New technologies require development and validation to address issues of production scale-up to meet clinical need; assays are required for process control, validation and release. Prior to clinical realization, an understanding of production processes is required to implement process changes that are essential for process control. Identification of key parameters forms the basis of process tolerances, allowing for validated, adaptive manufacturing processes. This enables greater process control and yield while withstanding regulatory scrutiny. This report summaries key milestones in specifically for ventral midbrain dopaminergic neuroprogenitor differentiation and key translational considerations and recommendations to enable successful, robust and reproducible current cell therapy product-manufacturing.
Collapse
Affiliation(s)
- James WT Kusena
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough, Leicestershire, LE11 3TU, UK
| | - Robert J Thomas
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough, Leicestershire, LE11 3TU, UK
| | - Mark J McCall
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough, Leicestershire, LE11 3TU, UK
| | - Samantha L Wilson
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough, Leicestershire, LE11 3TU, UK
| |
Collapse
|
35
|
Henchcliffe C, Parmar M. Repairing the Brain: Cell Replacement Using Stem Cell-Based Technologies. JOURNAL OF PARKINSONS DISEASE 2019; 8:S131-S137. [PMID: 30584166 PMCID: PMC6311366 DOI: 10.3233/jpd-181488] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Current approaches to cell replacement therapy in Parkinson's disease are strongly focused on the dopamine system, with the view that restoring dopaminergic inputs in a localized and physiologic manner will provide superior benefits in terms of effect and longevity compared with oral medication. Experience using transplants of fetal tissue containing dopaminergic cell precursors has provided valuable proof that the approach is feasible, and that engrafted cells can survive and function over many years. However, multiple drawbacks and procedural complications are recognized in using fetal cells. Recent strides in stem cell technology now make it possible to overcome some of the barriers associated with fetal tissue. In particular the generation of high numbers of specific cell types, such as dopaminergic neurons, from stem cells means that quality, consistency, activity, and safety can be more thoroughly determined prior to transplantation, thus providing hope for more robust outcomes. These cells are also predicted to provide benefit without leading to the graft-induced dyskinesia that led to morbidity in a subset of individuals who underwent fetal mesencephalic cell and tissue grafting in the 1990s. In thinking about developing such novel therapeutics, the choice of starting material has also expanded, with the availability of multiple human embryonic stem cell lines, as well as the possibilities for producing induced pluripotent cells, or neuronal cells from a patient's own tissue. In this article, we speculate on how rapidly expanding knowledge and technical possibilities may impact on stem cell-based therapies for cell replacement in Parkinson's disease over the next two decades.
Collapse
Affiliation(s)
- Claire Henchcliffe
- Department of Neurology, Weill Cornell Medical College, and Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York NY, USA
| | - Malin Parmar
- Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, BMC, Lund, Sweden
| |
Collapse
|
36
|
De Luca M, Aiuti A, Cossu G, Parmar M, Pellegrini G, Robey PG. Advances in stem cell research and therapeutic development. Nat Cell Biol 2019; 21:801-811. [PMID: 31209293 DOI: 10.1038/s41556-019-0344-z] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 05/09/2019] [Indexed: 12/12/2022]
Abstract
Despite many reports of putative stem-cell-based treatments in genetic and degenerative disorders or severe injuries, the number of proven stem cell therapies has remained small. In this Review, we survey advances in stem cell research and describe the cell types that are currently being used in the clinic or are close to clinical trials. Finally, we analyse the scientific rationale, experimental approaches, caveats and results underpinning the clinical use of such stem cells.
Collapse
Affiliation(s)
- Michele De Luca
- Center for Regenerative Medicine "Stefano Ferrari", Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget) and Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Graziella Pellegrini
- Center for Regenerative Medicine "Stefano Ferrari", Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Pamela Gehron Robey
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
37
|
Affiliation(s)
- Helen M Blau
- From the Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA (H.M.B.); and the Department of Medicine, Harvard Medical School, Boston (G.Q.D.)
| | - George Q Daley
- From the Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA (H.M.B.); and the Department of Medicine, Harvard Medical School, Boston (G.Q.D.)
| |
Collapse
|
38
|
Sebastian S, Hourd P, Chandra A, Williams DJ, Medcalf N. The management of risk and investment in cell therapy process development: a case study for neurodegenerative disease. Regen Med 2019; 14:465-488. [PMID: 31210581 DOI: 10.2217/rme-2018-0081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cell-based therapies must achieve clinical efficacy and safety with reproducible and cost-effective manufacturing. This study addresses process development issues using the exemplar of a human pluripotent stem cell-based dopaminergic neuron cell therapy product. Early identification and correction of risks to product safety and the manufacturing process reduces the expensive and time-consuming bridging studies later in development. A New Product Introduction map was used to determine the developmental requirements specific to the product. Systematic Risk Analysis is exemplified here. Expected current value-based prioritization guides decisions about the sequence of process studies and whether and if an early abandonment of further research is appropriate. The application of the three tools enabled prioritization of the development studies.
Collapse
Affiliation(s)
- Sujith Sebastian
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Paul Hourd
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Amit Chandra
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - David J Williams
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Nicholas Medcalf
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical & Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| |
Collapse
|
39
|
Cell therapy for Parkinson′s disease is coming of age: current challenges and future prospects with a focus on immunomodulation. Gene Ther 2019; 27:6-14. [DOI: 10.1038/s41434-019-0077-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/12/2019] [Accepted: 03/20/2019] [Indexed: 12/17/2022]
|
40
|
Chen W, Huang Q, Ma S, Li M. Progress in Dopaminergic Cell Replacement and Regenerative Strategies for Parkinson's Disease. ACS Chem Neurosci 2019; 10:839-851. [PMID: 30346716 DOI: 10.1021/acschemneuro.8b00389] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is a chronic progressive neurodegenerative disorder symptomatically characterized by resting tremor, rigidity, bradykinesia, and gait impairment. These motor deficits suffered by PD patients primarily result from selective dysfunction or loss of dopaminergic neurons of the substantia nigra pars compacta (SNpc). Most of the existing therapies for PD are based on the replacement of dopamine, which is symptomatically effective in the early stage but becomes increasingly less effective and is accompanied by serious side effects in the advanced stages of the disease. Currently, there are no strategies to slow neuronal degeneration or prevent the progression of PD. Thus, the prospect of regenerating functional dopaminergic neurons is very attractive. Over the last few decades, significant progress has been made in the development of dopaminergic regenerative strategies for curing PD. The most promising approach seems to be cell-replacement therapy (CRT) using human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs), which are unlimitedly available and have gained much success in preclinical trials. Despite the challenges, stem cell-based CRT will make significant steps toward the clinic in the coming decade. Alternatively, direct lineage reprogramming, especially in situ direct conversion of glia cells to induced neurons, which exhibits some advantages including no ethical concerns, no risk of tumor formation, and even no need for transplantation, has gained much attention recently. Evoking the endogenous regeneration ability of neural stem cells (NSCs) is an idyllic method of dopaminergic neuroregeneration which remains highly controversial. Here, we review many of these advances, highlighting areas and strategies that might be particularly suited to the development of regenerative approaches that restore dopaminergic function in PD.
Collapse
Affiliation(s)
- Weizhao Chen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Qiaoying Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan Road 2, Guangzhou 510080, China
| |
Collapse
|
41
|
Irion S. Cell Therapies for Parkinson's Disease. Clin Transl Sci 2019; 12:95-97. [PMID: 30771274 PMCID: PMC6440559 DOI: 10.1111/cts.12612] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/16/2018] [Indexed: 12/27/2022] Open
|
42
|
Duru LN, Quan Z, Qazi TJ, Qing H. Stem cells technology: a powerful tool behind new brain treatments. Drug Deliv Transl Res 2018; 8:1564-1591. [PMID: 29916013 DOI: 10.1007/s13346-018-0548-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cell research has recently become a hot research topic in biomedical research due to the foreseen unlimited potential of stem cells in tissue engineering and regenerative medicine. For many years, medicine has been facing intense challenges, such as an insufficient number of organ donations that is preventing clinicians to fulfill the increasing needs. To try and overcome this regrettable matter, research has been aiming at developing strategies to facilitate the in vitro culture and study of stem cells as a tool for tissue regeneration. Meanwhile, new developments in the microfluidics technology brought forward emerging cell culture applications that are currently allowing for a better chemical and physical control of cellular microenvironment. This review presents the latest developments in stem cell research that brought new therapies to the clinics and how the convergence of the microfluidics technology with stem cell research can have positive outcomes on the fields of regenerative medicine and high-throughput screening. These advances will bring new translational solutions for drug discovery and will upgrade in vitro cell culture to a new level of accuracy and performance. We hope this review will provide new insights into the understanding of new brain treatments from the perspective of stem cell technology especially regarding regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Lucienne N Duru
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhenzhen Quan
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Talal Jamil Qazi
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Beijing, China. .,Beijing Key Laboratory of Separation and Analysis in Biomedical and Pharmaceuticals, Department of Biomedical Engineering, School of Life Science, Beijing Institute of Technology, 5 South Zhongguancun Street, Haidian District, Beijing, 100081, China.
| |
Collapse
|
43
|
Fleifel D, Rahmoon MA, AlOkda A, Nasr M, Elserafy M, El-Khamisy SF. Recent advances in stem cells therapy: A focus on cancer, Parkinson's and Alzheimer's. J Genet Eng Biotechnol 2018; 16:427-432. [PMID: 30733756 PMCID: PMC6354001 DOI: 10.1016/j.jgeb.2018.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/29/2018] [Accepted: 09/09/2018] [Indexed: 02/07/2023]
Abstract
Stem cells serve as potential therapeutics due to their high proliferative capacity, low immunogenic reactivity and their differentiating capabilities. Several pre-clinical and early-stage clinical studies are carried out to treat genetic diseases, cancers and neurodegenerative disorders with promising preliminary results. However, there are still many challenges that scientists are trying to overcome such as the unclear expression profile of stem cells in vivo, the homing of stem cells to the site of injury and their potential immune-reactivity. Prospective research lies in gene editing of autologous stem cells in vitro and safe injection of these modified cells back into patients. Here, we review the clinical trials executed using stem cell therapy in an attempt to cure challenging diseases like cancer, Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Dalia Fleifel
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, 6th of October City, Giza 12578, Egypt
| | - Mai Atef Rahmoon
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, 6th of October City, Giza 12578, Egypt
| | - Abdelrahman AlOkda
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, 6th of October City, Giza 12578, Egypt
| | - Mostafa Nasr
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, 6th of October City, Giza 12578, Egypt
| | - Menattallah Elserafy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, 6th of October City, Giza 12578, Egypt
| | - Sherif F. El-Khamisy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Ahmed Zewail Road, October Gardens, 6th of October City, Giza 12578, Egypt
- Krebs Institute, Department of Molecular Biology and Biotechnology, Firth Court, University of Sheffield, S10 2TN Sheffield, UK
| |
Collapse
|
44
|
Lane EL. L-DOPA for Parkinson's disease-a bittersweet pill. Eur J Neurosci 2018; 49:384-398. [PMID: 30118169 DOI: 10.1111/ejn.14119] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 01/02/2023]
Abstract
3,4-dihydroxy-L-phenylalanine (L-DOPA) is the gold standard treatment for Parkinson's disease. It has earned that title through its highly effective treatment of some of the motor symptoms in the early stages of the disease but it is a far from perfect drug. The inevitable long-term treatment that comes with this chronic neurodegenerative condition raises the risk significantly of the development of motor fluctuations including disabling L-DOPA-induced dyskinesia. Being unsurpassed as a therapy means that understanding the mechanisms of dyskinesia priming and induction is vital to the search for therapies to treat these side effects and allow optimal use of L-DOPA. However, L-DOPA use may also have consequences (positive or negative) for the development of other interventions, such as cell transplantation, which are designed to treat or repair the ailing brain. This review looks at the issues around the use of L-DOPA with a focus on its potential impact on advanced reparative interventions.
Collapse
Affiliation(s)
- Emma L Lane
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
45
|
Parmar M, Torper O, Drouin-Ouellet J. Cell-based therapy for Parkinson's disease: A journey through decades toward the light side of the Force. Eur J Neurosci 2018; 49:463-471. [PMID: 30099795 PMCID: PMC6519227 DOI: 10.1111/ejn.14109] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/09/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
Abstract
This review describes the history, development, and evolution of cell‐based replacement therapy for Parkinson's disease (PD), from the first pioneering trials with fetal ventral midbrain progenitors to future trials using stem cells as well as reprogrammed cells. In the spirit of Tom Isaacs, the review takes parallels to the storyline of Star Wars, including the temptations from the dark side and the continuous fight for the light side of the Force. It is subdivided into headings based on the original movies, spanning from A New Hope to the Last Jedi.
Collapse
Affiliation(s)
- Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Olof Torper
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Janelle Drouin-Ouellet
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
46
|
|
47
|
Novel Epigenetic Techniques Provided by the CRISPR/Cas9 System. Stem Cells Int 2018; 2018:7834175. [PMID: 30123293 PMCID: PMC6079388 DOI: 10.1155/2018/7834175] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 02/04/2018] [Accepted: 03/27/2018] [Indexed: 12/26/2022] Open
Abstract
Epigenetics classically refers to the inheritable changes of hereditary information without perturbing DNA sequences. Understanding mechanisms of how epigenetic factors contribute to inheritable phenotype changes and cell identity will pave the way for us to understand diverse biological processes. In recent years, the emergence of CRISPR/Cas9 technology has provided us with new routes to the epigenetic field. In this review, novel epigenetic techniques utilizing the CRISPR/Cas9 system are the main contents to be discussed, including epigenome editing, temporal and spatial control of epigenetic effectors, noncoding RNA manipulation, chromatin in vivo imaging, and epigenetic element screening.
Collapse
|
48
|
Abstract
ABSTRACT
Treating neurodegenerative diseases with cell transplantation has been within reach since the first pioneering clinical trials in which dopamine neuron progenitors from the fetal brain were transplanted to individuals with Parkinson's disease. However, the use of fetal tissue is problematic in terms of low availability and high variability, and it is also associated with ethical concerns that vary between countries. For decades, the field has therefore investigated new scalable source of therapeutic cells from stem cells or via reprogramming. Now it is possible to generate authentic midbrain dopaminergic neurons from pluripotent stem cells and clinical trials using such cells are rapidly approaching.
Collapse
Affiliation(s)
- Malin Parmar
- Department of Experimental Medical Science, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, Lund University, BMC A11, S-221 84 Lund, Sweden
| |
Collapse
|