1
|
Yi LX, Woon HR, Saw G, Zeng L, Tan EK, Zhou ZD. Induced pluripotent stem cell-related approaches to generate dopaminergic neurons for Parkinson's disease. Neural Regen Res 2025; 20:3193-3206. [PMID: 39665833 PMCID: PMC11881713 DOI: 10.4103/nrr.nrr-d-24-00771] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/25/2024] [Accepted: 10/23/2024] [Indexed: 12/13/2024] Open
Abstract
The progressive loss of dopaminergic neurons in affected patient brains is one of the pathological features of Parkinson's disease, the second most common human neurodegenerative disease. Although the detailed pathogenesis accounting for dopaminergic neuron degeneration in Parkinson's disease is still unclear, the advancement of stem cell approaches has shown promise for Parkinson's disease research and therapy. The induced pluripotent stem cells have been commonly used to generate dopaminergic neurons, which has provided valuable insights to improve our understanding of Parkinson's disease pathogenesis and contributed to anti-Parkinson's disease therapies. The current review discusses the practical approaches and potential applications of induced pluripotent stem cell techniques for generating and differentiating dopaminergic neurons from induced pluripotent stem cells. The benefits of induced pluripotent stem cell-based research are highlighted. Various dopaminergic neuron differentiation protocols from induced pluripotent stem cells are compared. The emerging three-dimension-based brain organoid models compared with conventional two-dimensional cell culture are evaluated. Finally, limitations, challenges, and future directions of induced pluripotent stem cell-based approaches are analyzed and proposed, which will be significant to the future application of induced pluripotent stem cell-related techniques for Parkinson's disease.
Collapse
Affiliation(s)
| | | | | | - Li Zeng
- National Neuroscience Institute, Singapore
- Department of Neurology, Singapore General Hospital, Singapore
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore
| | - Eng King Tan
- National Neuroscience Institute, Singapore
- Department of Neurology, Singapore General Hospital, Singapore
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore
| | - Zhi Dong Zhou
- National Neuroscience Institute, Singapore
- Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore
| |
Collapse
|
2
|
Daadi MM, Snyder EY. Exercise your graft - An important lesson for cell replacement therapy for Parkinson's disease. Exp Neurol 2025; 385:115085. [PMID: 39631719 DOI: 10.1016/j.expneurol.2024.115085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/24/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Parkinson's disease (PD) is a complex multisystem, chronic and so far, incurable disease affecting millions of people worldwide. With the continuing need for better therapeutic options for PD, there is a global renewed interest in cell replacement therapy due to progress in using pluripotent stem cells as an unlimited source of dopaminergic (DA) neurons for cell transplantation. Despite the significant progress made, obstacles remain that interfere with the restoration of functional circuits by DA grafts. The functional connectivity between DA grafts and host cells may be enhanced by adjunctive therapies, such as physical activity. Exercise modalities, such as use of treadmill, enhance neuroplasticity and improve motor and cognitive functions in PD patients. The patients are able to re-learn movement and adjust their posture, which, in turn, results in short term-reduced rigidity and improved stride length and cadence. By stabilizing selected active inputs and eliminating inactive ones, activity-dependent mechanisms fine-tune new neural circuits for optimal connection and physiological function. This communication will review the mechanisms and synergies between cell replacement therapy and physical and cognitive training to enhance induced pluripotent stem cell-mediated functional reinnervation of the striatum in PD.
Collapse
Affiliation(s)
- Marcel M Daadi
- Department of Cell Systems & Anatomy, Long School of Medicine, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA; Department of Radiology, Long School of Medicine, University of Texas Health at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229, USA.
| | - Evan Y Snyder
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA; Sanford Burnham Prebys Medical Discovery Institute, Center for Stem Cells & Regenerative Medicine, La Jolla, CA 92037, USA
| |
Collapse
|
3
|
Kim MS, Yoon S, Choi J, Kim YJ, Lee G. Stem Cell-Based Approaches in Parkinson's Disease Research. Int J Stem Cells 2025; 18:21-36. [PMID: 38449089 PMCID: PMC11867902 DOI: 10.15283/ijsc23169] [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: 10/21/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative condition characterized by the loss of midbrain dopaminergic neurons, leading to motor symptoms. While current treatments provide limited relief, they don't alter disease progression. Stem cell technology, involving patient-specific stem cell-derived neurons, offers a promising avenue for research and personalized regenerative therapies. This article reviews the potential of stem cell-based research in PD, summarizing ongoing efforts, their limitations, and introducing innovative research models. The integration of stem cell technology and advanced models promises to enhance our understanding and treatment strategies for PD.
Collapse
Affiliation(s)
- Min Seong Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Subeen Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Jiwoo Choi
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Yong Jun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
- KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, Korea
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Mercado NM, Szarowicz C, Stancati JA, Sortwell CE, Boezwinkle SA, Collier TJ, Caulfield ME, Steece-Collier K. Advancing age and the rs6265 BDNF SNP are permissive to graft-induced dyskinesias in parkinsonian rats. NPJ Parkinsons Dis 2024; 10:163. [PMID: 39179609 PMCID: PMC11344059 DOI: 10.1038/s41531-024-00771-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024] Open
Abstract
The rs6265 single nucleotide polymorphism (SNP) in the gene for brain-derived neurotrophic factor is a common variant that alters therapeutic outcomes for individuals with Parkinson's disease (PD). We previously investigated the effects of this SNP on the experimental therapeutic approach of neural grafting, demonstrating that young adult parkinsonian rats carrying the variant Met allele exhibited enhanced graft function compared to wild-type rats and also exclusively developed aberrant graft-induced dyskinesias (GID). Aging is the primary risk factor for PD and reduces graft efficacy. Here we investigated whether aging interacts with this SNP to further alter cell transplantation outcomes. We hypothesized that aging would reduce enhancement of graft function associated with this genetic variant and exacerbate GID in all grafted subjects. Unexpectedly, beneficial graft function was maintained in aged rs6265 subjects. However, aging was permissive to GID induction, regardless of genotype, with the greatest incidence and severity found in rs6265-expressing animals.
Collapse
Affiliation(s)
- Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Carlye Szarowicz
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, 49503, USA
| | - Samuel A Boezwinkle
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Timothy J Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, 49503, USA
| | - Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, 49503, USA.
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
5
|
Zampar S, Di Gregorio SE, Grimmer G, Watts JC, Ingelsson M. "Prion-like" seeding and propagation of oligomeric protein assemblies in neurodegenerative disorders. Front Neurosci 2024; 18:1436262. [PMID: 39161653 PMCID: PMC11330897 DOI: 10.3389/fnins.2024.1436262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
Intra- or extracellular aggregates of proteins are central pathogenic features in most neurodegenerative disorders. The accumulation of such proteins in diseased brains is believed to be the end-stage of a stepwise aggregation of misfolded monomers to insoluble cross-β fibrils via a series of differently sized soluble oligomers/protofibrils. Several studies have shown how α-synuclein, amyloid-β, tau and other amyloidogenic proteins can act as nucleating particles and thereby share properties with misfolded forms, or strains, of the prion protein. Although the roles of different protein assemblies in the respective aggregation cascades remain unclear, oligomers/protofibrils are considered key pathogenic species. Numerous observations have demonstrated their neurotoxic effects and a growing number of studies have indicated that they also possess seeding properties, enabling their propagation within cellular networks in the nervous system. The seeding behavior of oligomers differs between the proteins and is also affected by various factors, such as size, shape and epitope presentation. Here, we are providing an overview of the current state of knowledge with respect to the "prion-like" behavior of soluble oligomers for several of the amyloidogenic proteins involved in neurodegenerative diseases. In addition to providing new insight into pathogenic mechanisms, research in this field is leading to novel diagnostic and therapeutic opportunities for neurodegenerative diseases.
Collapse
Affiliation(s)
- Silvia Zampar
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Sonja E. Di Gregorio
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Gustavo Grimmer
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Joel C. Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Martin Ingelsson
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Public Health/Geriatrics, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Park TY, Jeon J, Cha Y, Kim KS. Past, present, and future of cell replacement therapy for parkinson's disease: a novel emphasis on host immune responses. Cell Res 2024; 34:479-492. [PMID: 38777859 PMCID: PMC11217403 DOI: 10.1038/s41422-024-00971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Parkinson's disease (PD) stands as the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence continues to rise with the aging global population. Central to the pathophysiology of PD is the specific degeneration of midbrain dopamine neurons (mDANs) in the substantia nigra. Consequently, cell replacement therapy (CRT) has emerged as a promising treatment approach, initially supported by various open-label clinical studies employing fetal ventral mesencephalic (fVM) cells. Despite the initial favorable results, fVM cell therapy has intrinsic and logistical limitations that hinder its transition to a standard treatment for PD. Recent efforts in the field of cell therapy have shifted its focus towards the utilization of human pluripotent stem cells, including human embryonic stem cells and induced pluripotent stem cells, to surmount existing challenges. However, regardless of the transplantable cell sources (e.g., xenogeneic, allogeneic, or autologous), the poor and variable survival of implanted dopamine cells remains a major obstacle. Emerging evidence highlights the pivotal role of host immune responses following transplantation in influencing the survival of implanted mDANs, underscoring an important area for further research. In this comprehensive review, building upon insights derived from previous fVM transplantation studies, we delve into the functional ramifications of host immune responses on the survival and efficacy of grafted dopamine cells. Furthermore, we explore potential strategic approaches to modulate the host immune response, ultimately aiming for optimal outcomes in future clinical applications of CRT for PD.
Collapse
Affiliation(s)
- Tae-Yoon Park
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Jeha Jeon
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Young Cha
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA.
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA.
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard Medical School, Belmont, MA, USA.
| |
Collapse
|
7
|
Bogomiakova ME, Bogomazova AN, Lagarkova MA. Dysregulation of Immune Tolerance to Autologous iPSCs and Their Differentiated Derivatives. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:799-816. [PMID: 38880643 DOI: 10.1134/s0006297924050031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/21/2023] [Accepted: 02/13/2024] [Indexed: 06/18/2024]
Abstract
Induced pluripotent stem cells (iPSCs), capable of differentiating into any cell type, are a promising tool for solving the problem of donor organ shortage. In addition, reprogramming technology makes it possible to obtain a personalized, i.e., patient-specific, cell product transplantation of which should not cause problems related to histocompatibility of the transplanted tissues and organs. At the same time, inconsistent information about the main advantage of autologous iPSC-derivatives - lack of immunogenicity - still casts doubt on the possibility of using such cells beyond immunosuppressive therapy protocols. This review is devoted to immunogenic properties of the syngeneic and autologous iPSCs and their derivatives, as well as to the reasons for dysregulation of their immune tolerance.
Collapse
Affiliation(s)
- Margarita E Bogomiakova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia.
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Alexandra N Bogomazova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
8
|
Daadi EW, Daadi ES, Oh T, Li M, Kim J, Daadi MM. Combining physical & cognitive training with iPSC-derived dopaminergic neuron transplantation promotes graft integration & better functional outcome in parkinsonian marmosets. Exp Neurol 2024; 374:114694. [PMID: 38272159 DOI: 10.1016/j.expneurol.2024.114694] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024]
Abstract
Parkinson's disease (PD) is a relentlessly progressive and currently incurable neurodegenerative disease with significant unmet medical needs. Since PD stems from the degeneration of midbrain dopaminergic (DA) neurons in a defined brain location, PD patients are considered optimal candidates for cell replacement therapy. Clinical trials for cell transplantation in PD are beginning to re-emerge worldwide with a new focus on induced pluripotent stem cells (iPSCs) as a source of DA neurons since they can be derived from adult somatic cells and produced in large quantities under current good manufacturing practices. However, for this therapeutic strategy to be realized as a viable clinical option, fundamental translational challenges need to be addressed including the manufacturing process, purity and efficacy of the cells, the method of delivery, the extent of host reinnervation and the impact of patient-centered adjunctive interventions. In this study we report on the impact of physical and cognitive training (PCT) on functional recovery in the nonhuman primate (NHP) model of PD after cell transplantation. We observed that at 6 months post-transplant, the PCT group returned to normal baseline in their daily activity measured by actigraphy, significantly improved in their sensorimotor and cognitive tasks, and showed enhanced synapse formation between grafted cells and host cells. We also describe a robust, simple, efficient, scalable, and cost-effective manufacturing process of engraftable DA neurons derived from iPSCs. This study suggests that integrating PCT with cell transplantation therapy could promote optimal graft functional integration and better outcome for patients with PD.
Collapse
Affiliation(s)
- Etienne W Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
| | - Elyas S Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
| | - Thomas Oh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
| | - Mingfeng Li
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA; Department of Cell Systems & Anatomy, Long School of Medicine, University of Texas Health at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA
| | - Marcel M Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA; Department of Cell Systems & Anatomy, Long School of Medicine, University of Texas Health at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA; Department of Radiology, Long School of Medicine, University of Texas Health at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA.
| |
Collapse
|
9
|
Gima S, Oe K, Nishimura K, Ohgita T, Ito H, Kimura H, Saito H, Takata K. Host-to-graft propagation of inoculated α-synuclein into transplanted human induced pluripotent stem cell-derived midbrain dopaminergic neurons. Regen Ther 2024; 25:229-237. [PMID: 38283940 PMCID: PMC10818157 DOI: 10.1016/j.reth.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/30/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction Cell therapeutic clinical trials using fetal mesencephalic tissue provided a proof-of-concept for regenerative therapy in patients with Parkinson's disease. Postmortem studies of patients with fetal grafts revealed that α-synuclein+ Lewy body (LB)-like inclusions emerged in long-term transplantation and might worsen clinical outcomes even if the grafts survived and innervated in the recipients. Various studies aimed at addressing whether host-derived α-synuclein could be transferred to the grafted neurons to assess α-synuclein+ inclusion appearance in the grafts. However, determining whether α-synuclein in the grafted neurons has been propagated from the host is difficult due to the intrinsic α-synuclein expression. Methods We induced midbrain dopaminergic (mDA) neurons from human induced pluripotent stem cells (hiPSCs) and transplanted them into the striatum of immunodeficient rats. The recombinant human α-synuclein preformed fibrils (PFFs) were inoculated into the cerebral cortex after transplantation of SNCA-/- hiPSC-derived mDA neural progenitors into the striatum of immunodeficient rats to evaluate the host-to-graft propagation of human α-synuclein PFFs. Additionally, we examined the incorporation of human α-synuclein PFFs into SNCA-/- hiPSC-derived mDA neurons using in vitro culture system. Results We detected human α-synuclein-immunoreactivity in SNCA-/- hiPSC-derived mDA neurons that lacked endogenous α-synuclein expression in vitro. Additionally, we observed host-to-graft α-synuclein propagation into the grafted SNCA-/- hiPSC-derived mDA neurons. Conclusion We have successfully proven that intracerebral inoculated α-synuclein PFFs are propagated and incorporated from the host into grafted SNCA-/- hiPSC-derived mDA neurons. Our results contribute toward the basic understanding of the molecular mechanisms related to LB-like α-synuclein deposit formation in grafted mDA neurons.
Collapse
Affiliation(s)
- Serina Gima
- Joint Research Laboratory, Division of Integrated Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Kazuya Oe
- Joint Research Laboratory, Division of Integrated Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Kaneyasu Nishimura
- Joint Research Laboratory, Division of Integrated Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
- Laboratory of Functional Brain Circuit Construction, Graduate School of Brain Science, Doshisha University, Kyotanabe 610-0394, Japan
| | - Takashi Ohgita
- Center for Instrumental Analysis, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Haruka Ito
- Joint Research Laboratory, Division of Integrated Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
- Division of Probe Chemistry for Disease Analysis/Central Institute for Radioisotope Science, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa 920-8640, Japan
| | - Hiroyuki Saito
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Kazuyuki Takata
- Joint Research Laboratory, Division of Integrated Pharmaceutical Science, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| |
Collapse
|
10
|
Zhang L, Yang H. Research progress of neural stem cells as a source of dopaminergic neurons for cell therapy in Parkinson's disease. Mol Biol Rep 2024; 51:347. [PMID: 38400887 DOI: 10.1007/s11033-024-09294-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, the most characteristic pathological feature is the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compactus (SNpc) of the mesencephalon, along with reduced dopamine content in the striatum. Researchers have been searching for drugs and therapies to treat PD in decades. However, no approach could stop the progression of the disease, and even some of them caused adverse clinical side effects. PD has a well-defined lesion. Therefore, it is considered to be one of the most curable central nervous system diseases by cell replacement treatment. Fetal ventral mesencephalic tissue transplantation has been used to treat patients with PD and obtained positive treatment results. However, ethical issues, such as limited donor tissue, and side effects including graft-induced dyskinesias, limit its clinical applications. Neural stem cell (NSC) transplantation is a viable therapy choice because it possesses multipotency, self-renewal ability, and differentiation into DA neurons, which may substitute for lost DA neurons and slow down the neurodegenerative process in PD. Studies that investigated the delivery of NSCs by using animal models of PD revealed survival, migration, and even amelioration of behavioral deficits. Here, the research progress of NSCs or NSC-derived DA neurons in treating PD was reviewed, and the practicability of present manufacturing processes for clinical testing was considered. This review is expected to offer ideas for practical strategies to solve the present technical and biological problems related to the clinical application of NSCs in PD.
Collapse
Affiliation(s)
- Lingling Zhang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, 555 East Youyi Road, Beilin District, Xi'an, 710054, China.
| | - Hao Yang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, 555 East Youyi Road, Beilin District, Xi'an, 710054, China
| |
Collapse
|
11
|
Cooper O, Hallett P, Isacson O. Upstream lipid and metabolic systems are potential causes of Alzheimer's disease, Parkinson's disease and dementias. FEBS J 2024; 291:632-645. [PMID: 36165619 PMCID: PMC10040476 DOI: 10.1111/febs.16638] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022]
Abstract
Brain health requires circuits, cells and molecular pathways to adapt when challenged and to promptly reset once the challenge has resolved. Neurodegeneration occurs when adaptability becomes confined, causing challenges to overwhelm neural circuitry. Studies of rare and common neurodegenerative diseases suggest that the accumulation of lipids can compromise circuit adaptability. Using microglia as an example, we review data that suggest increased lipid concentrations cause dysfunctional inflammatory responses to immune challenges, leading to Alzheimer's disease, Parkinson's disease and dementia. We highlight current approaches to treat lipid metabolic and clearance pathways and identify knowledge gaps towards restoring adaptive homeostasis in individuals who are at-risk of losing cognition.
Collapse
Affiliation(s)
- Oliver Cooper
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Penny Hallett
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| |
Collapse
|
12
|
Cunha AB, Schuelke C, Mesri A, Ruud SK, Aizenshtadt A, Ferrari G, Heiskanen A, Asif A, Keller SS, Ramos-Moreno T, Kalvøy H, Martínez-Serrano A, Krauss S, Emnéus J, Sampietro M, Martinsen ØG. Development of a Smart Wireless Multisensor Platform for an Optogenetic Brain Implant. SENSORS (BASEL, SWITZERLAND) 2024; 24:575. [PMID: 38257668 PMCID: PMC11154348 DOI: 10.3390/s24020575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
Implantable cell replacement therapies promise to completely restore the function of neural structures, possibly changing how we currently perceive the onset of neurodegenerative diseases. One of the major clinical hurdles for the routine implementation of stem cell therapies is poor cell retention and survival, demanding the need to better understand these mechanisms while providing precise and scalable approaches to monitor these cell-based therapies in both pre-clinical and clinical scenarios. This poses significant multidisciplinary challenges regarding planning, defining the methodology and requirements, prototyping and different stages of testing. Aiming toward an optogenetic neural stem cell implant controlled by a smart wireless electronic frontend, we show how an iterative development methodology coupled with a modular design philosophy can mitigate some of these challenges. In this study, we present a miniaturized, wireless-controlled, modular multisensor platform with fully interfaced electronics featuring three different modules: an impedance analyzer, a potentiostat and an optical stimulator. We show the application of the platform for electrical impedance spectroscopy-based cell monitoring, optical stimulation to induce dopamine release from optogenetically modified neurons and a potentiostat for cyclic voltammetry and amperometric detection of dopamine release. The multisensor platform is designed to be used as an opto-electric headstage for future in vivo animal experiments.
Collapse
Affiliation(s)
- André B. Cunha
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
| | - Christin Schuelke
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, P.O. Box 1110 Blindern, 0317 Oslo, Norway; (A.A.); (S.K.)
| | - Alireza Mesri
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy; (A.M.); (G.F.); (M.S.)
| | - Simen K. Ruud
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
| | - Aleksandra Aizenshtadt
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, P.O. Box 1110 Blindern, 0317 Oslo, Norway; (A.A.); (S.K.)
| | - Giorgio Ferrari
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy; (A.M.); (G.F.); (M.S.)
| | - Arto Heiskanen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (A.H.); (A.A.); (J.E.)
| | - Afia Asif
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (A.H.); (A.A.); (J.E.)
| | - Stephan S. Keller
- National Centre for Nano Fabrication and Characterization, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Tania Ramos-Moreno
- Lund Stem Cell Center, Division of Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, 22184 Lund, Sweden;
| | - Håvard Kalvøy
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway;
| | - Alberto Martínez-Serrano
- Department of Molecular Neurobiology, Center of Molecular Biology ‘Severo Ochoa’, Universidad Autónoma de Madrid, Calle Nicolás Cabrera 1, 28049 Madrid, Spain;
| | - Stefan Krauss
- Hybrid Technology Hub—Centre of Excellence, Institute of Basic Medical Sciences, P.O. Box 1110 Blindern, 0317 Oslo, Norway; (A.A.); (S.K.)
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950, 0424 Oslo, Norway
| | - Jenny Emnéus
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (A.H.); (A.A.); (J.E.)
| | - Marco Sampietro
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy; (A.M.); (G.F.); (M.S.)
| | - Ørjan G. Martinsen
- Department of Physics, University of Oslo, Sem Sælands vei 24, 0371 Oslo, Norway; (A.B.C.); (C.S.); (S.K.R.)
- Department of Clinical and Biomedical Engineering, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway;
| |
Collapse
|
13
|
Lindvall O. History of cellular grafting for central nervous system repair-A clinical perspective. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:15-40. [PMID: 39341652 DOI: 10.1016/b978-0-323-90120-8.00011-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
As late as in the 1970s, the evidence supporting that brain function might be restored by replacing dead cells by transplantation of new healthy cells was scarce in experimental animals and lacking in humans. Repairing the human brain was regarded as completely unrealistic by clinicians. Fifty years later, the situation is very different, and cellular grafting has reached patient application in several conditions affecting the CNS. The clinical studies performed so far have shown that cellular grafts can survive, grow, and function also in the diseased adult human brain. However, no proven treatment based on cell transplantation is currently available for any brain disorder. Here, the history of cellular grafting is described from a clinical perspective, including some of the preclinical work that has formed the basis for its translation to patient application. The focus is on cell transplantation for Parkinson disease, which in many ways is paving the way for this field of research. The chapter gives an account of the scientific milestones, the ups and downs, as well as the positive and negative reactions from the scientific and clinical community, and how this research field despite many obstacles has continued to move forward over more than four decades.
Collapse
Affiliation(s)
- Olle Lindvall
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden; Division of Neurology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Sun J, Zhang W, Wei ZZ, Song X, Jian L, Jiang F, Wang S, Li H, Zhang Y, Tuo H. Mesenchymal stromal cell biotherapy for Parkinson's disease premotor symptoms. Chin Neurosurg J 2023; 9:28. [PMID: 37833807 PMCID: PMC10571301 DOI: 10.1186/s41016-023-00338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 07/30/2023] [Indexed: 10/15/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with motor deficits due to nigrostriatal dopamine depletion and with the non-motor/premotor symptoms (NMS) such as anxiety, cognitive dysfunction, depression, hyposmia, and sleep disorders. NMS is presented in at least one-fifth of the patients with PD. With the histological information being investigated, stem cells are shown to provide neurotrophic supports and cellular replacement in the damaging brain areas under PD conditions. Pathological change of progressive PD includes degeneration and loss of dopaminergic neurons in the substantia nigra of the midbrain. The current stem cell beneficial effect addresses dopamine boost for the striatal neurons and gliovascular mechanisms as competing for validated PD drug targets. In addition, there are clinical interventions for improving the patient's NMS and targeting their autonomic dysfunction, dementia, mood disorders, or sleep problems. In our and many others' research using brain injury models, multipotent mesenchymal stromal cells demonstrate an additional and unique ability to alleviate depressive-like behaviors, independent of an accelerated motor recovery. Intranasal delivery of the stem cells is discussed for it is extensively tested in rodent animal models of neurological and psychiatric disorders. In this review, we attempt to discuss the repairing potentials of transplanted cells into parkinsonism pathological regions of motor deficits and focus on preventive and treatment effects. From new approaches in the PD biological therapy, it is believed that it can as well benefit patients against PD-NMS.
Collapse
Affiliation(s)
- Jinmei Sun
- Clinical Diagnosis and Treatment Center for Parkinson's Disease, Beijing Friendship Hospital, Beijing, China
- Laboratories of Biological Therapeutic Medical Technology, Department of Neurology, Beijing Friendship Hospital Center for Neurological Disorders, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Neuroscience Institute, Beijing, China
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
- Beijing Tropical Medicine Research Institute, Beijing, China
| | - Wei Zhang
- Laboratories of Biological Therapeutic Medical Technology, Department of Neurology, Beijing Friendship Hospital Center for Neurological Disorders, Capital Medical University, Beijing, China.
| | - Zheng Zachory Wei
- Clinical Diagnosis and Treatment Center for Parkinson's Disease, Beijing Friendship Hospital, Beijing, China
- Laboratories of Biological Therapeutic Medical Technology, Department of Neurology, Beijing Friendship Hospital Center for Neurological Disorders, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Neuroscience Institute, Beijing, China
| | - Xiaopeng Song
- McLean Imaging Center, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Liu Jian
- Laboratories of Biological Therapeutic Medical Technology, Department of Neurology, Beijing Friendship Hospital Center for Neurological Disorders, Capital Medical University, Beijing, China
- Beijing Tropical Medicine Research Institute, Beijing, China
| | - Feng Jiang
- Neuroscience Research Institute, Peking University, Beijing, China
- Casstar, Zhongguancun No.1 Global Key & Core Technology (AI) Innovation Center, Beijing, China
| | - Shuanglin Wang
- Department of Critical Care Medicine, Airport Hospital of Tianjin Medical University General Hospital, Tianjin, China
- Department of Cardiovascular Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Haibo Li
- Department of Critical Care Medicine, Airport Hospital of Tianjin Medical University General Hospital, Tianjin, China
- Department of Cardiovascular Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Department of Biochemistry and Cell Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Yongbo Zhang
- Laboratories of Biological Therapeutic Medical Technology, Department of Neurology, Beijing Friendship Hospital Center for Neurological Disorders, Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Neuroscience Institute, Beijing, China
| | - Houzhen Tuo
- Clinical Diagnosis and Treatment Center for Parkinson's Disease, Beijing Friendship Hospital, Beijing, China.
- Laboratories of Biological Therapeutic Medical Technology, Department of Neurology, Beijing Friendship Hospital Center for Neurological Disorders, Capital Medical University, Beijing, China.
- National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Neuroscience Institute, Beijing, China.
| |
Collapse
|
15
|
Wang F, Sun Z, Peng D, Gianchandani S, Le W, Boltze J, Li S. Cell-therapy for Parkinson's disease: a systematic review and meta-analysis. J Transl Med 2023; 21:601. [PMID: 37679754 PMCID: PMC10483810 DOI: 10.1186/s12967-023-04484-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Cell-based strategies focusing on replacement or protection of dopaminergic neurons have been considered as a potential approach to treat Parkinson's disease (PD) for decades. However, despite promising preclinical results, clinical trials on cell-therapy for PD reported mixed outcomes and a thorough synthesis of these findings is lacking. We performed a systematic review and meta-analysis to evaluate cell-therapy for PD patients. METHODS We systematically identified all clinical trials investigating cell- or tissue-based therapies for PD published before July 2023. Out of those, studies reporting transplantation of homogenous cells (containing one cell type) were included in meta-analysis. The mean difference or standardized mean difference in quantitative neurological scale scores before and after cell-therapy was analyzed to evaluate treatment effects. RESULTS The systematic literature search revealed 106 articles. Eleven studies reporting data from 11 independent trials (210 patients) were eligible for meta-analysis. Disease severity and motor function evaluation indicated beneficial effects of homogenous cell-therapy in the 'off' state at 3-, 6-, 12-, or 24-month follow-ups, and for motor function even after 36 months. Most of the patients were levodopa responders (61.6-100% in different follow-ups). Cell-therapy was also effective in improving the daily living activities in the 'off' state of PD patients. Cells from diverse sources were used and multiple transplantation modes were applied. Autografts did not improve functional outcomes, while allografts exhibited beneficial effects. Encouragingly, both transplantation into basal ganglia and to areas outside the basal ganglia were effective to reduce disease severity. Some trials reported adverse events potentially related to the surgical procedure. One confirmed and four possible cases of graft-induced dyskinesia were reported in two trials included in this meta-analysis. CONCLUSIONS This meta-analysis provides preliminary evidence for the beneficial effects of homogenous cell-therapy for PD, potentially to the levodopa responders. Allogeneic cells were superior to autologous cells, and the effective transplantation sites are not limited to the basal ganglia. PROSPERO registration number: CRD42022369760.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Zhengwu Sun
- Department of Clinical Pharmacy, Central Hospital of Dalian University of Technology, Dalian, China
| | - Daoyong Peng
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Shikha Gianchandani
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Weidong Le
- Institute of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial Hospital, Chengdu, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Beijing, 100038, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
16
|
Kim J, Inbo H, Kim HS, Kim W, Jang SJ, Min K, Kim SH, Bae SH, Jeong YH, Kim B, Kim C, Schwarz SC, Schwarz J, Cho KG, Chung SS, Moon J. First Clinical Report on the Treatment of Parkinson's Disease with Fetal Midbrain Precursor Cells. Mov Disord 2023; 38:589-603. [PMID: 36692025 DOI: 10.1002/mds.29316] [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: 07/28/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Because human fetal ventral mesencephalic tissue grafts provide promising results in ameliorating Parkinson's disease-implicated motor dysfunctions, human fetal midbrain-derived dopamine neuronal precursor cells are considered good candidates for cell-based therapy for Parkinson's disease in that large quantities of cells can be supplied through a good manufacturing practice-compliant system. OBJECTIVE We conducted a prospective, phase I/IIa, dose-escalation, open-label "first-in-human" clinical trial with fetal neural precursor cells to assess their safety and therapeutic efficacy in patients with idiopathic Parkinson's disease. METHODS Fifteen patients were assigned to receive three different doses of cells (4 × 106 , 12 × 106 , and 40 × 106 cells) and completed a 12-month follow-up. The primary outcome was safety, by measuring the presence of grade 3 or higher cells according to National Cancer Institute guidelines and any contaminated cells. Secondary outcomes assessed motor and neurocognitive function, as well as the level of dopamine transporters, by positron emission tomography-computed tomography. RESULTS Although a pronation-supination and hand/arm movement performance was remarkably enhanced in all three groups (all P < 0.05), the medium- and high-dose-treated groups exhibited significant improvement in Unified Parkinson's Disease Rating Scale Part III only up to 26.16% and 40%, respectively, at 12 months after transplantation without any serious clinical complications or graft-induced dyskinesia in all patients. However, the motor improvements did not correlate with increase in the dopamine transporter on positron emission tomography images. CONCLUSIONS Our results primarily demonstrate the safety and plausible dose-dependent efficacy of human fetal midbrain-derived dopamine neuronal precursor cells for idiopathic Parkinson's disease. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Joopyoung Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Han Inbo
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Hyun Sook Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - WonChan Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Su Jin Jang
- Department of Nuclear Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Kyunghoon Min
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang Heum Kim
- Department of Neuroradiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Hun Bae
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| | - Yun-Hwa Jeong
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| | - Borah Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Chul Kim
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| | - Sigrid C Schwarz
- Department of Neurology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,German Center for Neurodegenerative Diseases, Technical University Munich, Munich, Germany
| | - Johannes Schwarz
- Department of Neurology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,Geriatric Hospital Haag, Haag, Germany
| | - Kyung Gi Cho
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Sup Chung
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Jisook Moon
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
17
|
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
|
18
|
Sekiya H, Tsuji A, Hashimoto Y, Takata M, Koga S, Nishida K, Futamura N, Kawamoto M, Kohara N, Dickson DW, Kowa H, Toda T. Discrepancy between distribution of alpha-synuclein oligomers and Lewy-related pathology in Parkinson's disease. Acta Neuropathol Commun 2022; 10:133. [PMID: 36068646 PMCID: PMC9450240 DOI: 10.1186/s40478-022-01440-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
The pathological hallmarks of Parkinson’s disease (PD) are α-synuclein (αSYN)-positive inclusions referred to as Lewy bodies and Lewy neurites, collectively referred to as Lewy-related pathology (LRP). LRP is thought to propagate in an ascending manner throughout the brain as the disease progresses. LRP is visible with histologic methods and is thought to represent a later stage of the disease process, while αSYN oligomers, which are not visible with routine histologic methods, are considered earlier. There is increasing evidence to suggest that αSYN oligomers may be more toxic than visible LRP. Detecting αSYN oligomers requires special techniques, and their distribution and association with clinical features are important research objectives. In this report, we describe the distribution of αSYN oligomers in multiple cortical and subcortical regions of PD using a proximity ligation assay (PLA). We observe widespread distribution of αSYN oligomers with PLA and more restricted distribution of LRP with αSYN immunohistochemistry. The distribution of αSYN oligomers differed from LRP in that αSYN oligomer burden was significantly greater in the neocortex, while LRP was greater in vulnerable subcortical regions, including the brainstem. We also found that cognitive impairment was associated with αSYN oligomers in the hippocampus. These results suggest that αSYN oligomers may be widely distributed in PD early in the disease process and that they may contribute to cognitive impairment in PD.
Collapse
Affiliation(s)
- Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA. .,Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan. .,Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.
| | - Asato Tsuji
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yuki Hashimoto
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Mariko Takata
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Shunsuke Koga
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Katsuya Nishida
- Department of Neurology, National Hospital Organization Hyogo-Chuo Hospital, Sanda, Hyogo, Japan
| | - Naonobu Futamura
- Department of Neurology, National Hospital Organization Hyogo-Chuo Hospital, Sanda, Hyogo, Japan
| | - Michi Kawamoto
- Department of Neurology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Nobuo Kohara
- Department of Neurology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Hisatomo Kowa
- Division of Neurology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Hyogo, Japan
| | - Tatsushi Toda
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan. .,Department of Neurology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
19
|
Gordon J, Lockard G, Monsour M, Alayli A, Choudhary H, Borlongan CV. Sequestration of Inflammation in Parkinson's Disease via Stem Cell Therapy. Int J Mol Sci 2022; 23:ijms231710138. [PMID: 36077534 PMCID: PMC9456021 DOI: 10.3390/ijms231710138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease is the second most common neurodegenerative disease. Insidious and progressive, this disorder is secondary to the gradual loss of dopaminergic signaling and worsening neuroinflammation, affecting patients’ motor capabilities. Gold standard treatment includes exogenous dopamine therapy in the form of levodopa–carbidopa, or surgical intervention with a deep brain stimulator to the subcortical basal ganglia. Unfortunately, these therapies may ironically exacerbate the already pro-inflammatory environment. An alternative approach may involve cell-based therapies. Cell-based therapies, whether endogenous or exogenous, often have anti-inflammatory properties. Alternative strategies, such as exercise and diet modifications, also appear to play a significant role in facilitating endogenous and exogenous stem cells to induce an anti-inflammatory response, and thus are of unique interest to neuroinflammatory conditions including Parkinson’s disease. Treating patients with current gold standard therapeutics and adding adjuvant stem cell therapy, alongside the aforementioned lifestyle modifications, may ideally sequester inflammation and thus halt neurodegeneration.
Collapse
Affiliation(s)
- Jonah Gordon
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Gavin Lockard
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Molly Monsour
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Adam Alayli
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Hassan Choudhary
- Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Correspondence:
| |
Collapse
|
20
|
Xu P, He H, Gao Q, Zhou Y, Wu Z, Zhang X, Sun L, Hu G, Guan Q, You Z, Zhang X, Zheng W, Xiong M, Chen Y. Human midbrain dopaminergic neuronal differentiation markers predict cell therapy outcome in a Parkinson's disease model. J Clin Invest 2022; 132:156768. [PMID: 35700056 PMCID: PMC9282930 DOI: 10.1172/jci156768] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-based replacement therapy holds great promise in treating Parkinson's disease (PD). However, the heterogeneity of hPSC-derived donor cells and the low yield of midbrain dopaminergic (mDA) neurons after transplantation hinder its broad clinical application. Here, we depicted the single-cell molecular landscape during mDA neuron differentiation. We found that this process recapitulated the development of multiple but adjacent fetal brain regions including ventral midbrain, isthmus, and ventral hindbrain, resulting in heterogenous donor cell population. We reconstructed the differentiation trajectory of mDA lineage and identified CLSTN2 and PTPRO as specific surface markers of mDA progenitors, which were predictive of mDA neuron differentiation and could facilitate highly enriched mDA neurons (up to 80%) following progenitor sorting and transplantation. Marker sorted progenitors exhibited higher therapeutic potency in correcting motor deficits of PD mice. Different marker sorted grafts had a strikingly consistent cellular composition, in which mDA neurons were enriched, while off-target neuron types were mostly depleted, suggesting stable graft outcomes. Our study provides a better understanding of cellular heterogeneity during mDA neuron differentiation, and establishes a strategy to generate highly purified donor cells to achieve stable and predictable therapeutic outcomes, raising the prospect of hPSC-based PD cell replacement therapies.
Collapse
Affiliation(s)
- Peibo Xu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Hui He
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qinqin Gao
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yingying Zhou
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ziyan Wu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Zhang
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Linyu Sun
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Gang Hu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qian Guan
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zhiwen You
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xinyue Zhang
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wenping Zheng
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Man Xiong
- Institute State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yuejun Chen
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
21
|
Wilson H, de Natale ER, Politis M. Concise Review: Recent advances in neuroimaging techniques to assist clinical trials on cell-based therapies in neurodegenerative diseases. Stem Cells 2022; 40:724-735. [PMID: 35671344 DOI: 10.1093/stmcls/sxac039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/17/2022] [Indexed: 11/14/2022]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are progressive disorders for which a curative therapy is still lacking. Cell-based therapy aims at replacing dysfunctional cellular populations by repairing damaged tissue and by enriching the microenvironment of selective brain areas, and thus constitutes a promising disease-modifying treatment of neurodegenerative diseases. Scientific research has engineered a wide range of human-derived cellular populations to help overcome some of the logistical, safety, and ethical issues associated with this approach. Open-label studies and clinical trials in human participants have employed neuroimaging techniques, such as positron emission tomography (PET) and magnetic resonance imaging (MRI), to assess the success of the transplantation, to evaluate the functional integration of the implanted tissue into the host environment and to understand the pathophysiological changes associated with the therapy. Neuroimaging has constituted an outcome measure of large, randomized clinical trials, and has given answers to clarify the pathophysiology underlying some of the complications linked with this therapy. Novel PET radiotracers and MRI sequences for the staging of neurodegenerative diseases and to study alterations at molecular level significantly expands the translational potential of neuroimaging to assist pre-clinical and clinical research on cell-based therapy in these disorders. This concise review summarizes the current use of neuroimaging in human studies of cell-based replacement therapy and focuses on future application of PET and MRI techniques to evaluate the pathophysiology and treatment efficacy, as well as to aid patient selection and as an outcome measure to improve treatment success.
Collapse
Affiliation(s)
- Heather Wilson
- Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK
| | | | - Marios Politis
- Neurodegeneration Imaging Group, University of Exeter Medical School, London, UK
| |
Collapse
|
22
|
Rosser AE, Busse ME, Gray WP, Badin RA, Perrier AL, Wheelock V, Cozzi E, Martin UP, Salado-Manzano C, Mills LJ, Drew C, Goldman SA, Canals JM, Thompson LM. Translating cell therapies for neurodegenerative diseases: Huntington's disease as a model disorder. Brain 2022; 145:1584-1597. [PMID: 35262656 PMCID: PMC9166564 DOI: 10.1093/brain/awac086] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/29/2022] [Accepted: 02/06/2022] [Indexed: 11/17/2022] Open
Abstract
There has been substantial progress in the development of regenerative medicine strategies for CNS disorders over the last decade, with progression to early clinical studies for some conditions. However, there are multiple challenges along the translational pipeline, many of which are common across diseases and pertinent to multiple donor cell types. These include defining the point at which the preclinical data are sufficiently compelling to permit progression to the first clinical studies; scaling-up, characterization, quality control and validation of the cell product; design, validation and approval of the surgical device; and operative procedures for safe and effective delivery of cell product to the brain. Furthermore, clinical trials that incorporate principles of efficient design and disease-specific outcomes are urgently needed (particularly for those undertaken in rare diseases, where relatively small cohorts are an additional limiting factor), and all processes must be adaptable in a dynamic regulatory environment. Here we set out the challenges associated with the clinical translation of cell therapy, using Huntington's disease as a specific example, and suggest potential strategies to address these challenges. Huntington's disease presents a clear unmet need, but, importantly, it is an autosomal dominant condition with a readily available gene test, full genetic penetrance and a wide range of associated animal models, which together mean that it is a powerful condition in which to develop principles and test experimental therapeutics. We propose that solving these challenges in Huntington's disease would provide a road map for many other neurological conditions. This white paper represents a consensus opinion emerging from a series of meetings of the international translational platforms Stem Cells for Huntington's Disease and the European Huntington's Disease Network Advanced Therapies Working Group, established to identify the challenges of cell therapy, share experience, develop guidance and highlight future directions, with the aim to expedite progress towards therapies for clinical benefit in Huntington's disease.
Collapse
Affiliation(s)
- Anne E. Rosser
- Cardiff University Neuroscience and Mental Health Research Institute, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
- Cardiff University Brain Repair Group, School of Biosciences, Life Sciences Building, Cardiff CF10 3AX, UK
- Brain Repair and Intracranial Neurotherapeutics (B.R.A.I.N.) Biomedical Research Unit, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4EP, UK
| | - Monica E. Busse
- Cardiff University Centre for Trials Research, College of Biomedical and Life Sciences Cardiff University, 4th Floor Neuadd Meirionnydd, Heath Park, Cardiff CF14 4YS, UK
| | - William P. Gray
- Cardiff University Neuroscience and Mental Health Research Institute, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
- Brain Repair and Intracranial Neurotherapeutics (B.R.A.I.N.) Biomedical Research Unit, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4EP, UK
- University Hospital of Wales Healthcare NHS Trust, Department of Neurosurgery, Cardiff CF14 4XW, UK
| | - Romina Aron Badin
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: mécanismes, thérapies, imagerie, 92265 Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Anselme L. Perrier
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: mécanismes, thérapies, imagerie, 92265 Fontenay-aux-Roses, France
- Université Paris-Saclay, CEA, Molecular Imaging Research Center, 92265 Fontenay-aux-Roses, France
| | - Vicki Wheelock
- University of California Davis, Department of Neurology, 95817 Sacramento, CA, USA
| | - Emanuele Cozzi
- Transplant Immunology Unit, Department of Cardiac, Thoracic and Vascular Sciences, Padua University Hospital—Ospedale Giustinianeo, Padova, Italy
| | - Unai Perpiña Martin
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, and Creatio-Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
| | - Cristina Salado-Manzano
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, and Creatio-Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
| | - Laura J. Mills
- Cardiff University Centre for Trials Research, College of Biomedical and Life Sciences Cardiff University, 4th Floor Neuadd Meirionnydd, Heath Park, Cardiff CF14 4YS, UK
| | - Cheney Drew
- Cardiff University Centre for Trials Research, College of Biomedical and Life Sciences Cardiff University, 4th Floor Neuadd Meirionnydd, Heath Park, Cardiff CF14 4YS, UK
| | - Steven A. Goldman
- Centre for Translational Neuromedicine, University of Rochester, 14642 Rochester, NY, USA
- University of Copenhagen Faculty of Health and Medical Sciences, DK-2200 Kobenhavn, Denmark
| | - Josep M. Canals
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedical Sciences, and Creatio-Production and Validation Center of Advanced Therapies, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
| | - Leslie M. Thompson
- University of California Irvine, Department of Psychiatry and Human Behaviour, Department of Neurobiology and Behavior and the Sue and Bill Gross Stem Cell Center, 92697 Irvine, CA, USA
| |
Collapse
|
23
|
Wang J, Tian Y, Shi X, Feng Z, Jiang L, Hao Y. Safety and Efficacy of Cell Transplantation on Improving Motor Symptoms in Patients With Parkinson’s Disease: A Meta-Analysis. Front Hum Neurosci 2022; 16:849069. [PMID: 35601911 PMCID: PMC9120834 DOI: 10.3389/fnhum.2022.849069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/28/2022] [Indexed: 12/04/2022] Open
Abstract
Background The past four decades have seen the growing use of tissue or cell transplants in Parkinson’s disease (PD) treatment. Parkinson’s cell therapy is a promising new treatment; however, efficacy of cell transplantation for Parkinson’s disease are entirely unclear. Objective To conduct a meta-analysis and a systematic review of the efficacy of cell therapy in patients with PD. Methods A systematic literature review and meta-analysis of 10 studies were performed to assess the efficacy of cell therapy in Parkinson’s patients. To achieve this, we compared the change in Unified Parkinson’s Disease Rating Scale (UPDRS) II and III scale scores to baseline and assessed the incidence of transplant-related adverse events. The MINORS score and the I2 index were applied to evaluate the quality of studies between-study heterogeneity, respectively. Results The literature search yielded 10 articles (n = 120). The improvement in motor function based on the UPDRSIII assessment was −14.044 (95% CI: −20.761, −7.327) (p < 0.001), whereas improvement in daily living ability based on the UPDRSII assessment was −5.661 (95% CI: −7.632, −3.689) (p < 0.001). Conclusion The present findings demonstrate important clues on the therapeutic effect of cell therapy in alleviating motor impairment and daily living ability in PD patients.
Collapse
Affiliation(s)
- Jiaming Wang
- Department of Neurosurgery, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Yu Tian
- Department of Neurosurgery, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Xin Shi
- Department of Neurosurgery, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, China
| | - Zhaohai Feng
- Department of Neurosurgery, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Lei Jiang
- Department of Neurosurgery, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Yujun Hao
- Department of Neurosurgery, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| |
Collapse
|
24
|
Chavarría C, Ivagnes R, Souza JM. Extracellular Alpha-Synuclein: Mechanisms for Glial Cell Internalization and Activation. Biomolecules 2022; 12:655. [PMID: 35625583 PMCID: PMC9138387 DOI: 10.3390/biom12050655] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022] Open
Abstract
Alpha-synuclein (α-syn) is a small protein composed of 140 amino acids and belongs to the group of intrinsically disordered proteins. It is a soluble protein that is highly expressed in neurons and expressed at low levels in glial cells. The monomeric protein aggregation process induces the formation of oligomeric intermediates and proceeds towards fibrillar species. These α-syn conformational species have been detected in the extracellular space and mediate consequences on surrounding neurons and glial cells. In particular, higher-ordered α-syn aggregates are involved in microglial and oligodendrocyte activation, as well as in the induction of astrogliosis. These phenomena lead to mitochondrial dysfunction, reactive oxygen and nitrogen species formation, and the induction of an inflammatory response, associated with neuronal cell death. Several receptors participate in cell activation and/or in the uptake of α-syn, which can vary depending on the α-syn aggregated state and cell types. The receptors involved in this process are of outstanding relevance because they may constitute potential therapeutic targets for the treatment of PD and related synucleinopathies. This review article focuses on the mechanism associated with extracellular α-syn uptake in glial cells and the consequent glial cell activation that contributes to the neuronal death associated with synucleinopathies.
Collapse
Affiliation(s)
| | | | - José M. Souza
- Departamento de Bioquímica, Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, 11400 Montevideo, Uruguay; (C.C.); (R.I.)
| |
Collapse
|
25
|
Better Outcomes with Intranigral versus Intrastriatal Cell Transplantation: Relevance for Parkinson’s Disease. Cells 2022; 11:cells11071191. [PMID: 35406755 PMCID: PMC8997951 DOI: 10.3390/cells11071191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
Intrastriatal embryonic ventral mesencephalon grafts have been shown to integrate, survive, and reinnervate the host striatum in clinical settings and in animal models of Parkinson’s disease. However, this ectopic location does not restore the physiological loops of the nigrostriatal pathway and promotes only moderate behavioral benefits. Here, we performed a direct comparison of the potential benefits of intranigral versus intrastriatal grafts in animal models of Parkinson’s disease. We report that intranigral grafts promoted better survival of dopaminergic neurons and that only intranigral grafts induced recovery of fine motor skills and normalized cortico-striatal responses. The increase in the number of toxic activated glial cells in host tissue surrounding the intrastriatal graft, as well as within the graft, may be one of the causes of the increased cell death observed in the intrastriatal graft. Homotopic localization of the graft and the subsequent physiological cell rewiring of the basal ganglia may be a key factor in successful and beneficial cell transplantation procedures.
Collapse
|
26
|
Markovic M, Yeapuri P, Namminga KL, Lu Y, Saleh M, Olson KE, Gendelman HE, Mosley RL. Interleukin-2 expands neuroprotective regulatory T cells in Parkinson's disease. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2022; 1:43-50. [PMID: 38407500 PMCID: PMC9254387 DOI: 10.1515/nipt-2022-0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022]
Abstract
Background Pharmacological approaches that boost neuroprotective regulatory T cell (Treg) number and function lead to neuroprotective activities in neurodegenerative disorders. Objectives We investigated whether low-dose interleukin 2 (IL-2) expands Treg populations and protects nigrostriatal dopaminergic neurons in a model of Parkinson's disease (PD). Methods IL-2 at 2.5 × 104 IU/dose/mouse was administered for 5 days. Lymphocytes were isolated and phenotype determined by flow cytometric analyses. To 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) intoxicated mice, 0.5 × 106 of enriched IL-2-induced Tregs were adoptively transferred to assess the effects on nigrostriatal neuron survival. Results IL-2 increased frequencies of CD4+CD25+CD127lowFoxP3+ Tregs that express ICOS and CD39 in blood and spleen. Adoptive transfer of IL-2-induced Tregs to MPTP-treated recipients increased tyrosine hydroxylase (TH)+ nigral dopaminergic neuronal bodies by 51% and TH+ striatal termini by 52% compared to control MPTP-treated animal controls. Conclusions IL-2 expands numbers of neuroprotective Tregs providing a vehicle for neuroprotection of nigrostriatal dopaminergic neurons in a pre-clinical PD model.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, 68198Omaha, NE, USA
| | - Pravin Yeapuri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, 68198Omaha, NE, USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, 68198Omaha, NE, USA
| | - Maamoon Saleh
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, 68198Omaha, NE, USA
| | - Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, 68198Omaha, NE, USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, 68198Omaha, NE, USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, 68198Omaha, NE, USA
| |
Collapse
|
27
|
Chau MJ, Quintero JE, Monje PV, Voss SR, Welleford AS, Gerhardt GA, van Horne CG. Using a Transection Paradigm to Enhance the Repair Mechanisms of an Investigational Human Cell Therapy. Cell Transplant 2022; 31:9636897221123515. [PMID: 36169034 PMCID: PMC9523845 DOI: 10.1177/09636897221123515] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022] Open
Abstract
One promising strategy in cell therapies for Parkinson's disease (PD) is to harness a patient's own cells to provide neuroprotection in areas of the brain affected by neurodegeneration. No treatment exists to replace cells in the brain. Thus, our goal has been to support sick neurons and slow neurodegeneration by transplanting living repair tissue from the peripheral nervous system into the substantia nigra of those with PD. Our group has pioneered the transplantation of transection-activated sural nerve fascicles into the brain of human subjects with PD. Our experience in sural nerve transplantation has supported the safety and feasibility of this approach. As part of a paradigm to assess the reparative properties of human sural nerve following a transection injury, we collected nerve tissue approximately 2 weeks after sural nerve transection for immunoassays from 15 participants, and collected samples from two additional participants for single nuclei RNA sequencing. We quantified the expression of key neuroprotective and select anti-apoptotic genes along with their corresponding protein levels using immunoassays. The single nuclei data clustered into 10 distinctive groups defined on the basis of previously published cell type-specific genes. Transection-induced reparative peripheral nerve tissue showed RNA expression of neuroprotective factors and anti-apoptotic factors across multiple cell types after nerve injury induction. Key proteins of interest (BDNF, GDNF, beta-NGF, PDGFB, and VEGF) were upregulated in reparative tissue. These results provide insight on this repair tissue's utility as a neuroprotective cell therapy.
Collapse
Affiliation(s)
- Monica J. Chau
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jorge E. Quintero
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Paula V. Monje
- Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephen Randal Voss
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew S. Welleford
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Greg A. Gerhardt
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Craig G. van Horne
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
28
|
Grosso Jasutkar H, Oh SE, Mouradian MM. Therapeutics in the Pipeline Targeting α-Synuclein for Parkinson's Disease. Pharmacol Rev 2022; 74:207-237. [PMID: 35017177 PMCID: PMC11034868 DOI: 10.1124/pharmrev.120.000133] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder and the fastest growing neurologic disease in the world, yet no disease-modifying therapy is available for this disabling condition. Multiple lines of evidence implicate the protein α-synuclein (α-Syn) in the pathogenesis of PD, and as such, there is intense interest in targeting α-Syn for potential disease modification. α-Syn is also a key pathogenic protein in other synucleionpathies, most commonly dementia with Lewy bodies. Thus, therapeutics targeting this protein will have utility in these disorders as well. Here we discuss the various approaches that are being investigated to prevent and mitigate α-Syn toxicity in PD, including clearing its pathologic aggregates from the brain using immunization strategies, inhibiting its misfolding and aggregation, reducing its expression level, enhancing cellular clearance mechanisms, preventing its cell-to-cell transmission within the brain and perhaps from the periphery, and targeting other proteins associated with or implicated in PD that contribute to α-Syn toxicity. We also discuss the therapeutics in the pipeline that harness these strategies. Finally, we discuss the challenges and opportunities for the field in the discovery and development of therapeutics for disease modification in PD. SIGNIFICANCE STATEMENT: PD is the second most common neurodegenerative disorder, for which disease-modifying therapies remain a major unmet need. A large body of evidence points to α-synuclein as a key pathogenic protein in this disease as well as in dementia with Lewy bodies, making it of leading therapeutic interest. This review discusses the various approaches being investigated and progress made to date toward discovering and developing therapeutics that would slow and stop progression of these disabling diseases.
Collapse
Affiliation(s)
- Hilary Grosso Jasutkar
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - Stephanie E Oh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| | - M Maral Mouradian
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Biomedical and Health Sciences, Piscataway, New Jersey
| |
Collapse
|
29
|
Barker RA. The Origins of the Protein Spread in Parkinson's Disease. Mov Disord Clin Pract 2021; 8:1194-1197. [PMID: 34765685 DOI: 10.1002/mdc3.13314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/05/2021] [Indexed: 11/10/2022] Open
Affiliation(s)
- Roger A Barker
- Wellcome-MRC Stem Cell institute and Department of Neurology, Addenbrooke's Hospital University of Cambridge Cambridge United Kingdom
| |
Collapse
|
30
|
Nandakumar S, Shahani P, Datta I, Pal R. Interventional Strategies for Parkinson Disease: Can Neural Precursor Cells Forge a Path Ahead? ACS Chem Neurosci 2021; 12:3785-3794. [PMID: 34628850 DOI: 10.1021/acschemneuro.1c00525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neural precursor cells (NPCs), derived from pluripotent stem cells (PSCs), with their unique ability to generate multiple neuronal and glial cell types are extremely useful for understanding biological mechanisms in normal and diseased states. However, generation of specific neuronal subtypes (mature) from NPCs in large numbers adequate for cell therapy is challenging due to lack of a thorough understanding of the cues that govern their differentiation. Interestingly, neural stem cells (NSCs) themselves are in consideration for therapy given their potency to form different neural cell types, release of trophic factors, and immunomodulatory effects that confer neuroprotection. With the recent COVID-19 outbreak and its accompanying neurological indications, the immunomodulatory role of NSCs may gain additional significance in the prevention of disease progression in vulnerable populations. In this regard, small-molecule mediated NPC generation from PSCs via NSC formation has become an important strategy that ensures consistency and robustness of the process. The development of the mammalian brain occurs along the rostro-caudal axis, and the establishment of anterior identity is an early event. Wnt signaling, along with fibroblast growth factor and retinoic acid, acts as a caudalization signal. Further, the increasing amount of epigenetic data available from human fetal brain development has enhanced both our understanding of and ability to experimentally manipulate these developmental regulatory programs in vitro. However, the impact on homing and engraftment after transplantation and subsequently on therapeutic efficacy of NPCs based on their derivation strategy is not yet clear. Another formidable challenge in cell replacement therapy for neurodegenerative disorders is the mode of delivery. In this Perspective, we discuss these core ideas with insights from our preliminary studies exploring the role of PSC-derived NPCs in rat models of MPTP-induced Parkinson's disease following intranasal injections.
Collapse
Affiliation(s)
- Swapna Nandakumar
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, Karnataka, India
| | - Pradnya Shahani
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru 560029, Karnataka, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru 560029, Karnataka, India
| | - Rajarshi Pal
- Eyestem Research, Centre for Cellular and Molecular Platforms (C-CAMP), Bengaluru 560065, Karnataka, India
| |
Collapse
|
31
|
Gibson R, Dalvi SP, Dalvi PS. DJ-1 and Parkinson's disease. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
32
|
Tomishima M, Kirkeby A. Bringing Advanced Therapies for Parkinson's Disease to the Clinic: The Scientist's Perspective. JOURNAL OF PARKINSONS DISEASE 2021; 11:S135-S140. [PMID: 34250954 PMCID: PMC8543246 DOI: 10.3233/jpd-212685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
After many years of preclinical development, cell and gene therapies have advanced from research tools in the lab to clinical-grade products for patients, and today they constitute more than a quarter of all new Phase I clinical trials for Parkinson’s disease. Whereas efficacy has been convincingly proven for many of these products in preclinical models, the field is now entering a new phase where the functionality and safety of these products will need to stand the test in clinical trials. If successful, these new products can have the potential to provide patients with a one-time administered treatment which may alleviate them from daily symptomatic dopaminergic medication.
Collapse
Affiliation(s)
| | - Agnete Kirkeby
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Wallenberg Center for Molecular Medicine (WCMM) and Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
33
|
Burns TC, Quinones-Hinojosa A. Regenerative medicine for neurological diseases-will regenerative neurosurgery deliver? BMJ 2021; 373:n955. [PMID: 34162530 DOI: 10.1136/bmj.n955] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regenerative medicine aspires to transform the future practice of medicine by providing curative, rather than palliative, treatments. Healing the central nervous system (CNS) remains among regenerative medicine's most highly prized but formidable challenges. "Regenerative neurosurgery" provides access to the CNS or its surrounding structures to preserve or restore neurological function. Pioneering efforts over the past three decades have introduced cells, neurotrophins, and genes with putative regenerative capacity into the CNS to combat neurodegenerative, ischemic, and traumatic diseases. In this review we critically evaluate the rationale, paradigms, and translational progress of regenerative neurosurgery, harnessing access to the CNS to protect, rejuvenate, or replace cell types otherwise irreversibly compromised by neurological disease. We discuss the evidence surrounding fetal, somatic, and pluripotent stem cell derived implants to replace endogenous neuronal and glial cell types and provide trophic support. Neurotrophin based strategies via infusions and gene therapy highlight the motivation to preserve neuronal circuits, the complex fidelity of which cannot be readily recreated. We specifically highlight ongoing translational efforts in Parkinson's disease, amyotrophic lateral sclerosis, stroke, and spinal cord injury, using these to illustrate the principles, challenges, and opportunities of regenerative neurosurgery. Risks of associated procedures and novel neurosurgical trials are discussed, together with the ethical challenges they pose. After decades of efforts to develop and refine necessary tools and methodologies, regenerative neurosurgery is well positioned to advance treatments for refractory neurological diseases. Strategic multidisciplinary efforts will be critical to harness complementary technologies and maximize mechanistic feedback, accelerating iterative progress toward cures for neurological diseases.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
34
|
Eskenazi D, Malave L, Mingote S, Yetnikoff L, Ztaou S, Velicu V, Rayport S, Chuhma N. Dopamine Neurons That Cotransmit Glutamate, From Synapses to Circuits to Behavior. Front Neural Circuits 2021; 15:665386. [PMID: 34093138 PMCID: PMC8170480 DOI: 10.3389/fncir.2021.665386] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/16/2021] [Indexed: 11/21/2022] Open
Abstract
Discovered just over 20 years ago, dopamine neurons have the ability to cotransmit both dopamine and glutamate. Yet, the functional roles of dopamine neuron glutamate cotransmission and their implications for therapeutic use are just emerging. This review article encompasses the current body of evidence investigating the functions of dopamine neurons of the ventral midbrain that cotransmit glutamate. Since its discovery in dopamine neuron cultures, further work in vivo confirmed dopamine neuron glutamate cotransmission across species. From there, growing interest has led to research related to neural functioning including roles in synaptic signaling, development, and behavior. Functional connectome mapping reveals robust connections in multiple forebrain regions to various cell types, most notably to cholinergic interneurons in both the medial shell of the nucleus accumbens and the lateral dorsal striatum. Glutamate markers in dopamine neurons reach peak levels during embryonic development and increase in response to various toxins, suggesting dopamine neuron glutamate cotransmission may serve neuroprotective roles. Findings from behavioral analyses reveal prominent roles for dopamine neuron glutamate cotransmission in responses to psychostimulants, in positive valence and cognitive systems and for subtle roles in negative valence systems. Insight into dopamine neuron glutamate cotransmission informs the pathophysiology of neuropsychiatric disorders such as addiction, schizophrenia and Parkinson Disease, with therapeutic implications.
Collapse
Affiliation(s)
- Daniel Eskenazi
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Lauren Malave
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY, United States
| | - Leora Yetnikoff
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, United States
- CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, United States
| | - Samira Ztaou
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Vlad Velicu
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| | - Nao Chuhma
- Department of Psychiatry, Columbia University, New York, NY, United States
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, United States
| |
Collapse
|
35
|
Li JY, Li W. Postmortem Studies of Fetal Grafts in Parkinson's Disease: What Lessons Have We Learned? Front Cell Dev Biol 2021; 9:666675. [PMID: 34055800 PMCID: PMC8155361 DOI: 10.3389/fcell.2021.666675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/06/2021] [Indexed: 12/28/2022] Open
Abstract
Neural transplantation is a potential therapeutic method for Parkinson’s disease (PD). Fetal dopaminergic (DA) neurons have been important transplantation cell sources in the history of replacement therapy for PD. Several decades of preclinical animal experiments and clinical trials using fetal DA neuron transplantation in PD therapy have shown not only promising results but also problems. In order to reveal possible factors influencing the clinical outcomes, we reviewed fetal DA neuron transplantation therapies from 1970s to present, with a special focus on postmortem studies. Firstly, we gave a general description of the clinical outcomes and neuroanatomy of grafted cases; secondly, we summarized the main available postmortem studies, including the cell survival, reinnervation, and pathology development. In the end, we further discussed the link between function and structure of the grafts, seeking for the possible factors contributing to a functional graft. With our review, we hope to provide references for future transplantation trials from a histological point of view.
Collapse
Affiliation(s)
- Jia-Yi Li
- Laboratory of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, China.,Neural Plasticity and Repair Unit, Wallenberg Neuroscience Centre, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Wen Li
- Laboratory of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, China.,Neural Plasticity and Repair Unit, Wallenberg Neuroscience Centre, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
36
|
Persistent dyskinesias in patients with fetal tissue transplantation for Parkinson disease. NPJ PARKINSONS DISEASE 2021; 7:38. [PMID: 33893319 PMCID: PMC8065148 DOI: 10.1038/s41531-021-00183-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 02/05/2021] [Indexed: 01/24/2023]
Abstract
Cell transplants are being developed for patients with Parkinson disease (PD) who have insufficient benefit with standard medical treatment. We describe the clinical features of five patients who developed persistent dyskinesias after fetal dopaminergic tissue transplantation. All had levodopa-induced dyskinesias preoperatively. We implanted fetal mesencephalic dopaminergic tissue into the putamina bilaterally in 34 patients with advanced PD. They were not immunosuppressed. Five of 34 patients (15%) developed troublesome choreic or dystonic dyskinesias that persisted despite lowering or discontinuing medications. Attempts to treat the involuntary movements with amantadine, clozapine, anticholinergics, dopamine depletors and other medicines had limited success. Metyrosine eliminated dyskinesias but led to the parkinsonian “off” state. Increasing the dose of levodopa worsened the dyskinesias. Three patients required placement of pallidal stimulators, bilaterally in two and unilaterally in one patient who had only contralateral dyskinesias. The two with the bilateral stimulators had improvement in dyskinesias. The patient with the unilateral pallidal stimulator had a substantial reduction of the dyskinesias, but attempts to treat residual “off” symptoms with levodopa were limited by worsening dyskinesias. Although the number of patients developing these persistent dyskinesias was small, these five patients had dramatic improvement after transplant. As a group, they had milder Parkinson signs at baseline and improved to the point of having minimal parkinsonism, with reduction or elimination of levodopa therapy prior to developing persistent dyskinesias. These involuntary movements establish the principle that fetal dopaminergic tissue transplants can mimic the effects of levodopa, not only in reducing bradykinesia, but also in provoking dyskinesias.
Collapse
|
37
|
Brás IC, Outeiro TF. Alpha-Synuclein: Mechanisms of Release and Pathology Progression in Synucleinopathies. Cells 2021; 10:cells10020375. [PMID: 33673034 PMCID: PMC7917664 DOI: 10.3390/cells10020375] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The accumulation of misfolded alpha-synuclein (aSyn) throughout the brain, as Lewy pathology, is a phenomenon central to Parkinson’s disease (PD) pathogenesis. The stereotypical distribution and evolution of the pathology during disease is often attributed to the cell-to-cell transmission of aSyn between interconnected brain regions. The spreading of conformationally distinct aSyn protein assemblies, commonly referred as strains, is thought to result in a variety of clinically and pathologically heterogenous diseases known as synucleinopathies. Although tremendous progress has been made in the field, the mechanisms involved in the transfer of these assemblies between interconnected neural networks and their role in driving PD progression are still unclear. Here, we present an update of the relevant discoveries supporting or challenging the prion-like spreading hypothesis. We also discuss the importance of aSyn strains in pathology progression and the various putative molecular mechanisms involved in cell-to-cell protein release. Understanding the pathways underlying aSyn propagation will contribute to determining the etiology of PD and related synucleinopathies but also assist in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Inês C. Brás
- Center for Biostructural Imaging of Neurodegeneration, Department of Experimental Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Tiago F. Outeiro
- Center for Biostructural Imaging of Neurodegeneration, Department of Experimental Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany;
- Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany
- Faculty of Medical Sciences, Translational and Clinical Research Institute, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
- Scientific Employee with a Honorary Contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075 Göttingen, Germany
- Correspondence: ; Tel.: +49-(0)-551-391-3544; Fax: +49-(0)-551-392-2693
| |
Collapse
|
38
|
Yu X, Persillet M, Zhang L, Zhang Y, Xiuping S, Li X, Ran G, Breger LS, Dovero S, Porras G, Dehay B, Bezard E, Qin C. Evaluation of blood flow as a route for propagation in experimental synucleinopathy. Neurobiol Dis 2021; 150:105255. [PMID: 33421564 DOI: 10.1016/j.nbd.2021.105255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
In Parkinson's disease, synucleinopathy is hypothesized to spread from the enteric nervous system, via the vagus nerve, to the central nervous system. Recent evidences collected in non-human primates challenge however the hypothesis of a transmission of α-synuclein (α-syn) pathology through the vagus nerve. Would the hypothesis whereby the bloodstream acts as a route for long-distance transmission of pathological α-syn hold true, an inter-individual transmission of synucleinopathy could occur via blood contact. Here, we used a parabiosis approach to join the circulatory systems of wild type and GFP transgenic C57BL/6 J mice, for which one of the partners parabiont received a stereotaxic intranigral injection of patient-derived α-syn aggregates. While the Lewy Body-receiving mice exhibited a loss of dopamine neurons and an increase in nigral S129 phosphorylated α-syn immunoreactivity, their parabiotic bloodstream-sharing partners did not show any trend for a lesion or change in S129 phosphorylated-α-syn levels. Altogether, our study suggests that, in the patient-derived α-synuclein aggregates-injected mouse model and within the selected time frame, the disease is not "transmitted" through the bloodstream.
Collapse
Affiliation(s)
- Xuan Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | | | - Ling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Yu Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Sun Xiuping
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Xianglei Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Gao Ran
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | | | - Sandra Dovero
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Gregory Porras
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Benjamin Dehay
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Erwan Bezard
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China; Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China.
| |
Collapse
|
39
|
Forloni G, La Vitola P, Cerovic M, Balducci C. Inflammation and Parkinson's disease pathogenesis: Mechanisms and therapeutic insight. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:175-202. [PMID: 33453941 DOI: 10.1016/bs.pmbts.2020.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
After Alzheimer's disease, Parkinson's disease is the most frequent neurodegenerative disorder. Although numerous treatments have been developed to control the disease symptomatology, with some successes, an efficacious therapy affecting the causes of PD is still a goal to pursue. The genetic evidence and the identification of α-synuclein as the main component of intracellular Lewy bodies, the neuropathological hallmark of PD and related disorders, have changed the approach to these disorders. More recently, the detrimental role of α-synuclein has been further extended to explain the wide spread of cerebral pathology through its oligomers. To emphasize the central pathogenic role of these soluble aggregates, we have defined synucleinopathies and other neurodegenerative disorders associated with protein misfolding as oligomeropathies. Another common element in the pathogenesis of oligomeropathies is the role played by inflammation, both at the peripheral and cerebral levels. In the brain parenchyma, inflammatory reaction has been considered an obvious consequence of neuronal degeneration, but recent observations indicate a direct contribution of glial alteration in the early phase of the disease. Furthermore, systemic inflammation also influences the development of neuronal dysfunction caused by specific elements, β amyloid, α-synuclein, tau or prion. However, each disorder has its own specific pathological process and within the same pathological condition, it is possible to find inter-individual differences. This heterogeneity might explain the difficulties developing efficacious therapeutic approaches, even though the possibility of intervention is supported by robust biological evidence. We have recently demonstrated that peripheral inflammation can amplify the neuronal dysfunction induced by α-synuclein oligomers and the neuropathological consequences observed in a Parkinson's disease model. In both cases, activation of microglia was incremented by the "double hit" process, compared to the single treatment. In contrast, astrocyte activation was attenuated and these cells appeared damaged when chronic inflammation was combined with α-synuclein exposure. This evidence might indicate a more specific anti-inflammatory strategy rather than the generic anti-inflammatory treatment.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Pietro La Vitola
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Milica Cerovic
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Claudia Balducci
- Biology of Neurodegenerative Diseases, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
40
|
The BDNF Val66Met polymorphism (rs6265) enhances dopamine neuron graft efficacy and side-effect liability in rs6265 knock-in rats. Neurobiol Dis 2020; 148:105175. [PMID: 33188920 PMCID: PMC7855552 DOI: 10.1016/j.nbd.2020.105175] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 01/10/2023] Open
Abstract
Prevalent in approximately 20% of the worldwide human population, the
rs6265 (also called ‘Val66Met’) single nucleotide polymorphism
(SNP) in the gene for brain-derived neurotrophic factor (BDNF)
is a common genetic variant that can alter therapeutic responses in individuals
with Parkinson’s disease (PD). Possession of the variant Met allele
results in decreased activity-dependent release of BDNF. Given the resurgent
worldwide interest in neural transplantation for PD and the biological relevance
of BDNF, the current studies examined the effects of the rs6265 SNP on
therapeutic efficacy and side-effect development following primary dopamine (DA)
neuron transplantation. Considering the significant reduction in BDNF release
associated with rs6265, we hypothesized that rs6265-mediated dysfunctional BDNF
signaling contributes to the limited clinical benefit observed in a
subpopulation of PD patients despite robust survival of grafted DA neurons, and
further, that this mutation contributes to the development of aberrant
graft-induced dyskinesias (GID). To this end, we generated a CRISPR knock-in rat
model of the rs6265 BDNF SNP to examine for the first time the
influence of a common genetic polymorphism on graft survival, functional
efficacy, and side-effect liability, comparing these parameters between
wild-type (Val/Val) rats and those homozygous for the variant Met allele
(Met/Met). Counter to our hypothesis, the current research indicates that
Met/Met rats show enhanced graft-associated therapeutic efficacy and a
paradoxical enhancement of graft-derived neurite outgrowth compared to wild-type
rats. However, consistent with our hypothesis, we demonstrate that the rs6265
genotype in the host rat is strongly linked to development of GID, and that this
behavioral phenotype is significantly correlated with neurochemical signatures
of atypical glutamatergic neurotransmission by grafted DA neurons.
Collapse
|
41
|
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: 19] [Impact Index Per Article: 3.8] [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
|
42
|
Rouaud T, Corbillé AG, Leclair-Visonneau L, de Guilhem de Lataillade A, Lionnet A, Preterre C, Damier P, Derkinderen P. Pathophysiology of Parkinson's disease: Mitochondria, alpha-synuclein and much more…. Rev Neurol (Paris) 2020; 177:260-271. [PMID: 33032797 DOI: 10.1016/j.neurol.2020.07.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a complex, age-related, neurodegenerative disease whose pathogenesis remains incompletely understood. Here, we give an overview of the progress that has been made over the past four decades in our understanding of this disorder. We review the role of mitochondria, environmental toxicants, alpha-synuclein and neuroinflammation in the development of PD. We also discuss more recent data from genetics, which strongly support the endosomal-lysosomal pathways and mitophagy as being central to PD. Finally, we discuss the emerging role of the gut-brain axis as a modulator of PD progression. This article is intended to provide a comprehensive, general and practical review of PD pathogenesis for the general neurologist.
Collapse
Affiliation(s)
- T Rouaud
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - A-G Corbillé
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | | | | | - A Lionnet
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - C Preterre
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - P Damier
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - P Derkinderen
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| |
Collapse
|
43
|
Xiong M, Tao Y, Gao Q, Feng B, Yan W, Zhou Y, Kotsonis TA, Yuan T, You Z, Wu Z, Xi J, Haberman A, Graham J, Block J, Zhou W, Chen Y, Zhang SC. Human Stem Cell-Derived Neurons Repair Circuits and Restore Neural Function. Cell Stem Cell 2020; 28:112-126.e6. [PMID: 32966778 DOI: 10.1016/j.stem.2020.08.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/11/2020] [Accepted: 08/21/2020] [Indexed: 02/02/2023]
Abstract
Although cell transplantation can rescue motor defects in Parkinson's disease (PD) models, whether and how grafts functionally repair damaged neural circuitry in the adult brain is not known. We transplanted hESC-derived midbrain dopamine (mDA) or cortical glutamate neurons into the substantia nigra or striatum of a mouse PD model and found extensive graft integration with host circuitry. Axonal pathfinding toward the dorsal striatum was determined by the identity of the grafted neurons, and anatomical presynaptic inputs were largely dependent on graft location, whereas inhibitory versus excitatory input was dictated by the identity of grafted neurons. hESC-derived mDA neurons display A9 characteristics and restore functionality of the reconstructed nigrostriatal circuit to mediate improvements in motor function. These results indicate similarity in cell-type-specific pre- and post-synaptic integration between transplant-reconstructed circuit and endogenous neural networks, highlighting the capacity of hPSC-derived neuron subtypes for specific circuit repair and functional restoration in the adult brain.
Collapse
Affiliation(s)
- Man Xiong
- Institute of Pediatrics, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Yezheng Tao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Program in Neuroscience & Behavioral Disorders, Duke-NUS Medical School, 169857 Singapore, Singapore
| | - Qinqin Gao
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ban Feng
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Yan
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yingying Zhou
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Thomas A Kotsonis
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Tingli Yuan
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhiwen You
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Ziyan Wu
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiajie Xi
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Julia Graham
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jasper Block
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wenhao Zhou
- Institute of Pediatrics, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Yuejun Chen
- Institute of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| | - Su-Chun Zhang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Program in Neuroscience & Behavioral Disorders, Duke-NUS Medical School, 169857 Singapore, Singapore
| |
Collapse
|
44
|
Heras-Garvin A, Stefanova N. From Synaptic Protein to Prion: The Long and Controversial Journey of α-Synuclein. Front Synaptic Neurosci 2020; 12:584536. [PMID: 33071772 PMCID: PMC7536368 DOI: 10.3389/fnsyn.2020.584536] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Since its discovery 30 years ago, α-synuclein (α-syn) has been one of the most studied proteins in the field of neuroscience. Dozens of groups worldwide have tried to reveal not only its role in the CNS but also in other organs. α-syn has been linked to several processes essential in brain homeostasis such as neurotransmitter release, synaptic function, and plasticity. However, despite the efforts made in this direction, the main function of α-syn is still unknown. Moreover, α-syn became a protein of interest for neurologists and neuroscientists when mutations in its gene were found associated with Parkinson's disease (PD) and even more when α-syn protein deposits were observed in the brain of PD, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA) patients. At present, the abnormal accumulation of α-syn constitutes one of the pathological hallmarks of these disorders, also referred to as α-synucleinopathies, and it is used for post-mortem diagnostic criteria. Whether α-syn aggregation is cause or consequence of the pathogenic events underlying α-synucleinopathies remains unclear and under discussion. Recently, different in vitro and in vivo studies have shown the ability of pathogenic α-syn to spread between cells, not only within the CNS but also from peripheral locations such as the gut, salivary glands, and through the olfactory network into the CNS, inducing abnormal misfolding of endogenous α-syn and leading to neurodegeneration and motor and cognitive impairment in animal models. Thus, it has been suggested that α-syn should be considered a prion protein. Here we present an update of what we know about α-syn function, aggregation and spreading, and its role in neurodegeneration. We also discuss the rationale and findings supporting the hypothetical prion nature of α-syn, its weaknesses, and future perspectives for research and the development of disease-modifying therapies.
Collapse
Affiliation(s)
- Antonio Heras-Garvin
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
45
|
Tomov N. Glial cells in intracerebral transplantation for Parkinson's disease. Neural Regen Res 2020; 15:1173-1178. [PMID: 31960796 PMCID: PMC7047789 DOI: 10.4103/1673-5374.270296] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/02/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
In the last few decades, intracerebral transplantation has grown from a dubious neuroscientific topic to a plausible modality for treatment of neurological disorders. The possibility for cell replacement opens a new field of perspectives in the therapy of neurodegenerative disorders, ischemia, and neurotrauma, with the most lessons learned from intracerebral transplantation in Parkinson's disease. Multiple animal studies and a few small-scale clinical trials have proven the concept of intracerebral grafting, but still have to provide a uniform and highly efficient approach to the procedure, suitable for clinical application. The success of intracerebral transplantation is highly dependent on the integration of the grafted cells with the host brain. In this process, glial cells are clearly more than passive bystanders. They provide transplanted cells with mechanical support, trophics, mediate synapse formation, and participate in graft vascularization. At the same time, glial cells mediate scarring, graft rejection, and neuroinflammation, which can be detrimental. We can use this information to try to understand the mechanisms behind the glial reaction to intracerebral transplantation. Recognizing and utilizing glial reactivity can move translational research forward and provide an insight not only to post-transplantation events but also to mechanisms of neuronal death and degeneration. Knowledge about glial reactivity to transplanted cells could also be a key for optimization of transplantation protocols, which ultimately should contribute to greater patient benefit.
Collapse
Affiliation(s)
- Nikola Tomov
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012 Bern, Switzerland
| |
Collapse
|
46
|
Dehay B, Bezard E. Intrastriatal injection of alpha-synuclein fibrils induces Parkinson-like pathology in macaques. Brain 2020; 142:3321-3322. [PMID: 31665750 DOI: 10.1093/brain/awz329] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This scientific commentary refers to ‘Intrastriatal alpha-synuclein fibrils in monkeys: spreading, imaging and neuropathological changes’, by Chu et al. (doi:10.1093/brain/awz296).
Collapse
Affiliation(s)
- Benjamin Dehay
- Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France.,Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Erwan Bezard
- Université de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France.,Centre National de la Recherche Scientifique Unité Mixte de Recherche 5293, Institut des Maladies Neurodégénératives, Bordeaux, France
| |
Collapse
|
47
|
Bourdenx M, Nioche A, Dovero S, Arotcarena ML, Camus S, Porras G, Thiolat ML, Rougier NP, Prigent A, Aubert P, Bohic S, Sandt C, Laferrière F, Doudnikoff E, Kruse N, Mollenhauer B, Novello S, Morari M, Leste-Lasserre T, Trigo-Damas I, Goillandeau M, Perier C, Estrada C, Garcia-Carrillo N, Recasens A, Vaikath NN, El-Agnaf OMA, Herrero MT, Derkinderen P, Vila M, Obeso JA, Dehay B, Bezard E. Identification of distinct pathological signatures induced by patient-derived α-synuclein structures in nonhuman primates. SCIENCE ADVANCES 2020; 6:eaaz9165. [PMID: 32426502 PMCID: PMC7220339 DOI: 10.1126/sciadv.aaz9165] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/06/2020] [Indexed: 06/11/2023]
Abstract
Dopaminergic neuronal cell death, associated with intracellular α-synuclein (α-syn)-rich protein aggregates [termed "Lewy bodies" (LBs)], is a well-established characteristic of Parkinson's disease (PD). Much evidence, accumulated from multiple experimental models, has suggested that α-syn plays a role in PD pathogenesis, not only as a trigger of pathology but also as a mediator of disease progression through pathological spreading. Here, we have used a machine learning-based approach to identify unique signatures of neurodegeneration in monkeys induced by distinct α-syn pathogenic structures derived from patients with PD. Unexpectedly, our results show that, in nonhuman primates, a small amount of singular α-syn aggregates is as toxic as larger amyloid fibrils present in the LBs, thus reinforcing the need for preclinical research in this species. Furthermore, our results provide evidence supporting the true multifactorial nature of PD, as multiple causes can induce a similar outcome regarding dopaminergic neurodegeneration.
Collapse
Affiliation(s)
- M. Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - A. Nioche
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- Institut Jean Nicod, Département d’études cognitives, ENS, EHESS, PSL Research University, 75005 Paris, France
- Institut Jean Nicod, Département d’études cognitives, CNRS, UMR 8129, Paris, France
| | - S. Dovero
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - M.-L. Arotcarena
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - S. Camus
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - G. Porras
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - M.-L. Thiolat
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - N. P. Rougier
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- INRIA Bordeaux Sud-Ouest, 33405 Talence, France
| | - A. Prigent
- INSERM, U1235, Nantes F-44035, France
- Nantes University, Nantes F-44035, France
- CHU Nantes, Department of Neurology, Nantes F-44093, France
| | - P. Aubert
- INSERM, U1235, Nantes F-44035, France
- Nantes University, Nantes F-44035, France
- CHU Nantes, Department of Neurology, Nantes F-44093, France
| | - S. Bohic
- EA-7442 Rayonnement Synchrotron et Recherche Medicale, RSRM, University of Grenoble Alpes, 38000 Grenoble, France
| | - C. Sandt
- SMIS beamline, Synchrotron SOLEIL, l’orme des merisiers, 91192 Gif sur Yvette, France
| | - F. Laferrière
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - E. Doudnikoff
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - N. Kruse
- Paracelsus-Elena-Klinik, Kassel, Germany
- University Medical Center Goettingen, Institute of Neuropathology, Goettingen, Germany
| | - B. Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany
- University Medical Center Goettingen, Institute of Neuropathology, Goettingen, Germany
| | - S. Novello
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
- Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - M. Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
- Neuroscience Center and National Institute of Neuroscience, University of Ferrara, via Fossato di Mortara 17-19, 44121 Ferrara, Italy
| | - T. Leste-Lasserre
- INSERM, Neurocentre Magendie, U1215, Physiopathologie de la Plasticité Neuronale, F-33000 Bordeaux, France
| | - I. Trigo-Damas
- HM CINAC, HM Puerta del Sur and CEU–San Pablo University Madrid, E-28938 Mostoles, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - M. Goillandeau
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - C. Perier
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute (VHIR)–Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - C. Estrada
- Clinical and Experimental Neuroscience Unit, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Institute of Research on Aging (IUIE), School of Medicine, University of Murcia, 30100 Murcia, Spain
| | - N. Garcia-Carrillo
- Centro Experimental en Investigaciones Biomédica (CEIB), Universidad de Murcia, Murcia, Spain
| | - A. Recasens
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute (VHIR)–Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - N. N. Vaikath
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar
| | - O. M. A. El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Education City, Qatar
| | - M. T. Herrero
- Clinical and Experimental Neuroscience Unit, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, Campus Mare Nostrum, 30100 Murcia, Spain
- Institute of Research on Aging (IUIE), School of Medicine, University of Murcia, 30100 Murcia, Spain
| | - P. Derkinderen
- INSERM, U1235, Nantes F-44035, France
- Nantes University, Nantes F-44035, France
- CHU Nantes, Department of Neurology, Nantes F-44093, France
| | - M. Vila
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- Neurodegenerative Diseases Research Group, Vall d’Hebron Research Institute (VHIR)–Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Autonomous University of Barcelona (UAB), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - J. A. Obeso
- HM CINAC, HM Puerta del Sur and CEU–San Pablo University Madrid, E-28938 Mostoles, Spain
- Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - B. Dehay
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| | - E. Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, F-33000 Bordeaux, France
| |
Collapse
|
48
|
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: 29] [Impact Index Per Article: 5.8] [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
|
49
|
Muddapu VR, Dharshini SAP, Chakravarthy VS, Gromiha MM. Neurodegenerative Diseases - Is Metabolic Deficiency the Root Cause? Front Neurosci 2020; 14:213. [PMID: 32296300 PMCID: PMC7137637 DOI: 10.3389/fnins.2020.00213] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/26/2020] [Indexed: 01/31/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer, Parkinson, Huntington, and amyotrophic lateral sclerosis, are a prominent class of neurological diseases currently without a cure. They are characterized by an inexorable loss of a specific type of neurons. The selective vulnerability of specific neuronal clusters (typically a subcortical cluster) in the early stages, followed by the spread of the disease to higher cortical areas, is a typical pattern of disease progression. Neurodegenerative diseases share a range of molecular and cellular pathologies, including protein aggregation, mitochondrial dysfunction, glutamate toxicity, calcium load, proteolytic stress, oxidative stress, neuroinflammation, and aging, which contribute to neuronal death. Efforts to treat these diseases are often limited by the fact that they tend to address any one of the above pathological changes while ignoring others. Lack of clarity regarding a possible root cause that underlies all the above pathologies poses a significant challenge. In search of an integrative theory for neurodegenerative pathology, we hypothesize that metabolic deficiency in certain vulnerable neuronal clusters is the common underlying thread that links many dimensions of the disease. The current review aims to present an outline of such an integrative theory. We present a new perspective of neurodegenerative diseases as metabolic disorders at molecular, cellular, and systems levels. This helps to understand a common underlying mechanism of the many facets of the disease and may lead to more promising disease-modifying therapeutic interventions. Here, we briefly discuss the selective metabolic vulnerability of specific neuronal clusters and also the involvement of glia and vascular dysfunctions. Any failure in satisfaction of the metabolic demand by the neurons triggers a chain of events that precipitate various manifestations of neurodegenerative pathology.
Collapse
Affiliation(s)
- Vignayanandam Ravindernath Muddapu
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - S. Akila Parvathy Dharshini
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - V. Srinivasa Chakravarthy
- Laboratory for Computational Neuroscience, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - M. Michael Gromiha
- Protein Bioinformatics Lab, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
50
|
Gonzalez-Rodriguez P, Zampese E, Surmeier DJ. Selective neuronal vulnerability in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2020; 252:61-89. [PMID: 32247375 DOI: 10.1016/bs.pbr.2020.02.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, disabling millions worldwide. Despite the imperative PD poses, at present, there is no cure or means of slowing progression. This gap is attributable to our incomplete understanding of the factors driving pathogenesis. Research over the past several decades suggests that both cell-autonomous and non-cell autonomous processes contribute to the neuronal dysfunction underlying PD symptoms. The thesis of this review is that an intersection of these processes governs the pattern of pathology in PD. Studies of substantia nigra pars compacta (SNc) dopaminergic neurons, whose loss is responsible for the core motor symptoms of PD, suggest that they have a combination of traits-a long, highly branched axon, autonomous activity, and elevated mitochondrial oxidant stress-that predispose them to non-cell autonomous drivers of pathogenesis, like misfolded forms of alpha-synuclein (α-SYN) and inflammation. The literature surrounding these issues will be briefly summarized, and the translational implications of an intersectional hypothesis of PD pathogenesis discussed.
Collapse
Affiliation(s)
| | - Enrico Zampese
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - D James Surmeier
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| |
Collapse
|