1
|
Kim MS, Kim H, Lee G. Precision Medicine in Parkinson's Disease Using Induced Pluripotent Stem Cells. Adv Healthc Mater 2024; 13:e2303041. [PMID: 38269602 DOI: 10.1002/adhm.202303041] [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: 09/11/2023] [Revised: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Parkinson's disease (PD) is one of the most devastating neurological diseases; however, there is no effective cure yet. The availability of human induced pluripotent stem cells (iPSCs) provides unprecedented opportunities to understand the pathogenic mechanism and identification of new therapy for PD. Here a new model system of PD, including 2D human iPSC-derived midbrain dopaminergic (mDA) neurons, 3D iPSC-derived midbrain organoids (MOs) with cellular complexity, and more advanced microphysiological systems (MPS) with 3D organoids, is introduced. It is believed that successful integrations and applications of iPSC, organoid, and MPS technologies can bring new insight on PD's pathogenesis that will lead to more effective treatments for this debilitating disease.
Collapse
Affiliation(s)
- Min Seong Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hyesoo Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
2
|
Catlin JP, Tooley CES. Exploring potential developmental origins of common neurodegenerative disorders. Biochem Soc Trans 2024; 52:1035-1044. [PMID: 38661189 PMCID: PMC11440815 DOI: 10.1042/bst20230422] [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: 09/11/2023] [Revised: 03/12/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
In the United States, it is now estimated that 6.7 million people over the age of 65 are afflicted by Alzheimer's disease (AD), over 1 million people are living with Parkinson's disease (PD), and over 200 000 have or are at risk for developing Huntington's disease (HD). All three of these neurodegenerative diseases result in the ultimate death of distinct neuronal subtypes, and it is widely thought that age-related damage is the single biggest contributing factor to this neuronal death. However, recent studies are now suggesting that developmental defects during early neurogenesis could also play a role in the pathology of neurodegenerative diseases. Loss or overexpression of proteins associated with HD, PD, and AD also result in embryonic phenotypes but whether these developmental defects slowly unmask over time and contribute to age-related neurodegeneration remains highly debated. Here, we discuss known links between embryonic neurogenesis and neurodegenerative disorders (including common signaling pathways), potential compensatory mechanisms that could delay presentation of neurodegenerative disorders, and the types of model systems that could be used to study these links in vivo.
Collapse
Affiliation(s)
- James P. Catlin
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Christine E. Schaner Tooley
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
3
|
Zhao HH, Haddad G. Brain organoid protocols and limitations. Front Cell Neurosci 2024; 18:1351734. [PMID: 38572070 PMCID: PMC10987830 DOI: 10.3389/fncel.2024.1351734] [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: 12/06/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
Stem cell-derived organoid technology is a powerful tool that revolutionizes the field of biomedical research and extends the scope of our understanding of human biology and diseases. Brain organoids especially open an opportunity for human brain research and modeling many human neurological diseases, which have lagged due to the inaccessibility of human brain samples and lack of similarity with other animal models. Brain organoids can be generated through various protocols and mimic whole brain or region-specific. To provide an overview of brain organoid technology, we summarize currently available protocols and list several factors to consider before choosing protocols. We also outline the limitations of current protocols and challenges that need to be solved in future investigation of brain development and pathobiology.
Collapse
Affiliation(s)
- Helen H. Zhao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
| | - Gabriel Haddad
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
- The Rady Children's Hospital, San Diego, CA, United States
| |
Collapse
|
4
|
Rosh I, Tripathi U, Hussein Y, Rike WA, Djamus J, Shklyar B, Manole A, Houlden H, Winkler J, Gage FH, Stern S. Synaptic dysfunction and extracellular matrix dysregulation in dopaminergic neurons from sporadic and E326K-GBA1 Parkinson's disease patients. NPJ Parkinsons Dis 2024; 10:38. [PMID: 38374278 PMCID: PMC10876637 DOI: 10.1038/s41531-024-00653-x] [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: 05/21/2023] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with both genetic and sporadic origins. In this study, we investigated the electrophysiological properties, synaptic activity, and gene expression differences in dopaminergic (DA) neurons derived from induced pluripotent stem cells (iPSCs) of healthy controls, sporadic PD (sPD) patients, and PD patients with E326K-GBA1 mutations. Our results demonstrate reduced sodium currents and synaptic activity in DA neurons derived from PD patients with E326K-GBA1 mutations, suggesting a potential contribution to PD pathophysiology. We also observed distinct electrophysiological alterations in sPD DA neurons, which included a decrease in synaptic currents. RNA sequencing analysis revealed unique dysregulated pathways in sPD neurons and E326K-GBA1 neurons, further supporting the notion that molecular mechanisms driving PD may differ between PD patients. In agreement with our previous reports, Extracellular matrix and Focal adhesion pathways were among the top dysregulated pathways in DA neurons from sPD patients and from patients with E326K-GBA1 mutations. Overall, our study further confirms that impaired synaptic activity is a convergent functional phenotype in DA neurons derived from PD patients across multiple genetic mutations as well as sPD. At the transcriptome level, we find that the brain extracellular matrix is highly involved in PD pathology across multiple PD-associated mutations as well as sPD.
Collapse
Affiliation(s)
- Idan Rosh
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Wote Amelo Rike
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Jose Djamus
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Boris Shklyar
- Bioimaging Unit, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Andreea Manole
- Laboratory of Genetics, Gage, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Henry Houlden
- UCL Queen Square Institute of Neurology, University College London, London, England
| | | | - Fred H Gage
- Laboratory of Genetics, Gage, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Shani Stern
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel.
| |
Collapse
|
5
|
Rosety I, Zagare A, Saraiva C, Nickels S, Antony P, Almeida C, Glaab E, Halder R, Velychko S, Rauen T, Schöler HR, Bolognin S, Sauter T, Jarazo J, Krüger R, Schwamborn JC. Impaired neuron differentiation in GBA-associated Parkinson's disease is linked to cell cycle defects in organoids. NPJ Parkinsons Dis 2023; 9:166. [PMID: 38110400 PMCID: PMC10728202 DOI: 10.1038/s41531-023-00616-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023] Open
Abstract
The mechanisms underlying Parkinson's disease (PD) etiology are only partially understood despite intensive research conducted in the field. Recent evidence suggests that early neurodevelopmental defects might play a role in cellular susceptibility to neurodegeneration. To study the early developmental contribution of GBA mutations in PD we used patient-derived iPSCs carrying a heterozygous N370S mutation in the GBA gene. Patient-specific midbrain organoids displayed GBA-PD relevant phenotypes such as reduction of GCase activity, autophagy impairment, and mitochondrial dysfunction. Genome-scale metabolic (GEM) modeling predicted changes in lipid metabolism which were validated with lipidomics analysis, showing significant differences in the lipidome of GBA-PD. In addition, patient-specific midbrain organoids exhibited a decrease in the number and complexity of dopaminergic neurons. This was accompanied by an increase in the neural progenitor population showing signs of oxidative stress-induced damage and premature cellular senescence. These results provide insights into how GBA mutations may lead to neurodevelopmental defects thereby predisposing to PD pathology.
Collapse
Affiliation(s)
- Isabel Rosety
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- OrganoTherapeutics SARL-S, Esch-sur-Alzette, Luxembourg
| | - Alise Zagare
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Claudia Saraiva
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sarah Nickels
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Paul Antony
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catarina Almeida
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Enrico Glaab
- Biomedical Data Science group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Rashi Halder
- Systems Ecology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sergiy Velychko
- Max Planck Institute for Molecular Biomedicine, MPG White Paper Group - Animal Testing in the Max Planck Society, Muenster, Germany
| | - Thomas Rauen
- Max Planck Institute for Molecular Biomedicine, MPG White Paper Group - Animal Testing in the Max Planck Society, Muenster, Germany
| | - Hans R Schöler
- Max Planck Institute for Molecular Biomedicine, MPG White Paper Group - Animal Testing in the Max Planck Society, Muenster, Germany
| | - Silvia Bolognin
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Thomas Sauter
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, 4367, Luxembourg
| | - Javier Jarazo
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- OrganoTherapeutics SARL-S, Esch-sur-Alzette, Luxembourg
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Transversial Translational Medicine, Luxembourg Institute of Health (LIH), 1 A-B rue Thomas Ediison, L-1445, Strassen, Luxembourg
| | - Jens C Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
6
|
Zagare A, Preciat G, Nickels SL, Luo X, Monzel AS, Gomez-Giro G, Robertson G, Jaeger C, Sharif J, Koseki H, Diederich NJ, Glaab E, Fleming RMT, Schwamborn JC. Omics data integration suggests a potential idiopathic Parkinson's disease signature. Commun Biol 2023; 6:1179. [PMID: 37985891 PMCID: PMC10662437 DOI: 10.1038/s42003-023-05548-w] [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: 06/30/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023] Open
Abstract
The vast majority of Parkinson's disease cases are idiopathic. Unclear etiology and multifactorial nature complicate the comprehension of disease pathogenesis. Identification of early transcriptomic and metabolic alterations consistent across different idiopathic Parkinson's disease (IPD) patients might reveal the potential basis of increased dopaminergic neuron vulnerability and primary disease mechanisms. In this study, we combine systems biology and data integration approaches to identify differences in transcriptomic and metabolic signatures between IPD patient and healthy individual-derived midbrain neural precursor cells. Characterization of gene expression and metabolic modeling reveal pyruvate, several amino acid and lipid metabolism as the most dysregulated metabolic pathways in IPD neural precursors. Furthermore, we show that IPD neural precursors endure mitochondrial metabolism impairment and a reduced total NAD pool. Accordingly, we show that treatment with NAD precursors increases ATP yield hence demonstrating a potential to rescue early IPD-associated metabolic changes.
Collapse
Affiliation(s)
- Alise Zagare
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg
| | - German Preciat
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA, Leiden, The Netherlands
| | - Sarah L Nickels
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg
| | - Xi Luo
- School of Medicine, University of Galway, University Rd, Galway, Ireland
| | - Anna S Monzel
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg
| | - Gemma Gomez-Giro
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg
| | - Graham Robertson
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg
| | - Christian Jaeger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg
| | - Jafar Sharif
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, 230-0045, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, 230-0045, Japan
| | - Nico J Diederich
- Centre Hospitalier de Luxembourg (CHL), 4, Rue Nicolas Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg
| | - Ronan M T Fleming
- Metabolomics and Analytics Center, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA, Leiden, The Netherlands
- School of Medicine, University of Galway, University Rd, Galway, Ireland
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts-Fourneaux, 4362, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
7
|
Toh HSY, Choo XY, Sun AX. Midbrain organoids-development and applications in Parkinson's disease. OXFORD OPEN NEUROSCIENCE 2023; 2:kvad009. [PMID: 38596240 PMCID: PMC10913847 DOI: 10.1093/oons/kvad009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/31/2023] [Indexed: 04/11/2024]
Abstract
Human brain development is spatially and temporally complex. Insufficient access to human brain tissue and inadequacy of animal models has limited the study of brain development and neurodegenerative diseases. Recent advancements of brain organoid technology have created novel opportunities to model human-specific neurodevelopment and brain diseases. In this review, we discuss the use of brain organoids to model the midbrain and Parkinson's disease. We critically evaluate the extent of recapitulation of PD pathology by organoids and discuss areas of future development that may lead to the model to become a next-generation, personalized therapeutic strategy for PD and beyond.
Collapse
Affiliation(s)
- Hilary S Y Toh
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
| | - Xin Yi Choo
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
| | - Alfred Xuyang Sun
- Neuroscience & Behavioural Disorders Programme, Duke-NUS Medical School, 8 College Road, Singapore
- National Neuroscience Institute, 11 Jln Tan Tock Seng, Singapore
| |
Collapse
|
8
|
Jiménez-Salvador I, Meade P, Iglesias E, Bayona-Bafaluy P, Ruiz-Pesini E. Developmental origins of Parkinson disease: Improving the rodent models. Ageing Res Rev 2023; 86:101880. [PMID: 36773760 DOI: 10.1016/j.arr.2023.101880] [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: 10/20/2022] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Numerous pesticides are inhibitors of the oxidative phosphorylation system. Oxidative phosphorylation dysfunction adversely affects neurogenesis and often accompanies Parkinson disease. Since brain development occurs mainly in the prenatal period, early exposure to pesticides could alter the development of the nervous system and increase the risk of Parkinson disease. Different rodent models have been used to confirm this hypothesis. However, more precise considerations of the selected strain, the xenobiotic, its mode of administration, and the timing of animal analysis, are necessary to resemble the model to the human clinical condition and obtain more reliable results.
Collapse
Affiliation(s)
- Irene Jiménez-Salvador
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain.
| | - Patricia Meade
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza, 50018 Zaragoza, Spain.
| | - Eldris Iglesias
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Facultad de Ciencias de la Salud, Universidad San Jorge, 50830 Villanueva de Gállego, Zaragoza, Spain.
| | - Pilar Bayona-Bafaluy
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza, 50018 Zaragoza, Spain.
| | - Eduardo Ruiz-Pesini
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009- and 50013 Zaragoza, Spain; Instituto de Investigación Sanitaria (IIS) de Aragón, 50009 Zaragoza, Spain; Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| |
Collapse
|
9
|
Godefroy V, Sezer I, Bouzigues A, Montembeault M, Koban L, Plassmann H, Migliaccio R. Altered delay discounting in neurodegeneration: insight into the underlying mechanisms and perspectives for clinical applications. Neurosci Biobehav Rev 2023; 146:105048. [PMID: 36669749 DOI: 10.1016/j.neubiorev.2023.105048] [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: 09/23/2022] [Revised: 12/12/2022] [Accepted: 01/14/2023] [Indexed: 01/19/2023]
Abstract
Steeper delay discounting (i.e., the extent to which future rewards are perceived as less valuable than immediate ones) has been proposed as a transdiagnostic process across different health conditions, in particular psychiatric disorders. Impulsive decision-making is a hallmark of different neurodegenerative conditions but little is known about delay discounting in the domain of neurodegenerative conditions. We reviewed studies on delay discounting in patients with Parkinson's disease (PD) and in patients with dementia (Alzheimer's disease / AD or frontotemporal dementia / FTD). We proposed that delay discounting could be an early marker of the neurodegenerative process. We developed the idea that altered delay discounting is associated with overlapping but distinct neurocognitive mechanisms across neurodegenerative diseases: dopaminergic-related disorders of reward processing in PD, memory/projection deficits due to medial temporal atrophy in AD, modified reward processing due to orbitofrontal atrophy in FTD. Neurodegeneration could provide a framework to decipher the neuropsychological mechanisms of value-based decision-making. Further, delay discounting could become a marker of interest in clinical practice, in particular for differential diagnosis.
Collapse
Affiliation(s)
- Valérie Godefroy
- FrontLab, INSERM U1127, Institut du cerveau, Hôpital Pitié-Salpêtrière, Paris, France; Marketing Area, INSEAD, Fontainebleau, France; Control-Interoception-Attention Team, Paris Brain Institute (ICM), INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France.
| | - Idil Sezer
- FrontLab, INSERM U1127, Institut du cerveau, Hôpital Pitié-Salpêtrière, Paris, France
| | - Arabella Bouzigues
- FrontLab, INSERM U1127, Institut du cerveau, Hôpital Pitié-Salpêtrière, Paris, France
| | - Maxime Montembeault
- Douglas Research Centre, Montréal, Canada; Department of Psychiatry, McGill University, Montréal, Canada
| | - Leonie Koban
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, Bron, France
| | - Hilke Plassmann
- Marketing Area, INSEAD, Fontainebleau, France; Control-Interoception-Attention Team, Paris Brain Institute (ICM), INSERM U 1127, CNRS UMR 7225, Sorbonne University, Paris, France
| | - Raffaella Migliaccio
- FrontLab, INSERM U1127, Institut du cerveau, Hôpital Pitié-Salpêtrière, Paris, France; Centre de Référence des Démences Rares ou Précoces, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France; Institute of Memory and Alzheimer's Disease, Centre of Excellence of Neurodegenerative Disease, Department of Neurology, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Paris, France.
| |
Collapse
|
10
|
Rummey C, Corben LA, Delatycki M, Wilmot G, Subramony SH, Corti M, Bushara K, Duquette A, Gomez C, Hoyle JC, Roxburgh R, Seeberger L, Yoon G, Mathews K, Zesiewicz T, Perlman S, Lynch DR. Natural History of Friedreich Ataxia: Heterogeneity of Neurologic Progression and Consequences for Clinical Trial Design. Neurology 2022; 99:e1499-e1510. [PMID: 35817567 PMCID: PMC9576299 DOI: 10.1212/wnl.0000000000200913] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The understanding of the natural history of Friedreich ataxia (FRDA) has improved considerably recently, but patterns of neurologic deterioration are not fully clarified, compromising the assessment of the clinical relevance of effects and guidance for study design. The goal of this study was to acknowledge the broad genetic diversity of the population, especially for younger individuals, and to provide analyses stratified by age to guide population selection in future studies. METHODS Based on a large natural history study, the FRDA Clinical Outcome Measures study that at the current data cut enrolled 1,115 participants, followed up for 5,287 yearly visits, we present results from the modified FRDA Rating Scale and its subscores. The secondary outcomes included the patient-reported activities of daily living scale, the timed 25-foot walk, and the 9-hole peg test. Long-term progression was modeled using slope analyses within early-onset, typical-onset, intermediate-onset, and late-onset FRDA. To reflect recruitment in clinical trials, short-term changes were analyzed within age-based subpopulations. All analyses were stratified by ambulation status. RESULTS Long-term progression models stratified by disease severity indicated highly differential disease progression, especially at earlier ages at onset. In the ambulatory phase, decline was driven by axial items assessed by the Upright Stability subscore of the mFARS. The analyses of short-term changes showed slower progression with increasing population age due to decreasing genetic severity. Future clinical studies could reduce population diversity, interpatient variability, and the risk of imbalanced treatment groups by selecting the study population based on the functional capacity (e.g., ambulatory status) and by strict age-based stratification. DISCUSSION The understanding of the diversity within FRDA populations and their patterns of functional decline provides an essential foundation for future clinical trial design including patient selection and facilitates the interpretation of the clinical relevance of progression detected in FRDA.
Collapse
Affiliation(s)
- Christian Rummey
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Louise A Corben
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Martin Delatycki
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - George Wilmot
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Sub H Subramony
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Manuela Corti
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Khalaf Bushara
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Antoine Duquette
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Christopher Gomez
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - J Chad Hoyle
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Richard Roxburgh
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Lauren Seeberger
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Grace Yoon
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Katherine Mathews
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Theresa Zesiewicz
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - Susan Perlman
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA
| | - David R Lynch
- From the Clinical Data Science GmbH (C.R.), Basel, Switzerland; Bruce Lefroy Centre for Genetic Health Research (L.A.C., M.D.), Murdoch Children's Research Institute, Parkville, Victoria; Department of Paediatrics (L.A.C., M.D.), University of Melbourne. Parkville, Victoria, Australia; Emory University (G.W.), Atlanta, GA; Department of Neurology (S.H.S., M.C.), McKnight Brain Institute, Gainesville, FL; University of Minnesota (K.B.), Minneapolis; Service de Neurologie (A.D.), Département de Médecine, Unité de Troubles du Mouvement André-Barbeau, Centre Hospitalier de l'Université de Montréal (CHUM) and CRCHUM, Quebec, Canada; University of Chicago (C.G.), IL; Ohio State University (J.C.H.), Columbus; University of Auckland (R.R.), New Zealand; University of Colorado (L.S.), Denver; Divisions of Neurology and Clinical and Metabolic Genetics (G.Y.), Department of Paediatrics, the Hospital for Sick Children, University of Toronto, Ontario, Canada; University of Iowa (K.M.), Iowa City, Carver College of Medicine, Iowa City, IA; University of South Florida (T.Z.), Tampa; University of California Los Angeles (S.P.); and Division of Neurology (D.R.L.), Children's Hospital of Philadelphia, PA.
| |
Collapse
|
11
|
Langlie J, Mittal R, Finberg A, Bencie NB, Mittal J, Omidian H, Omidi Y, Eshraghi AA. Unraveling pathological mechanisms in neurological disorders: the impact of cell-based and organoid models. Neural Regen Res 2022; 17:2131-2140. [PMID: 35259819 PMCID: PMC9083150 DOI: 10.4103/1673-5374.335836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cell-based models are a promising tool in deciphering the molecular mechanisms underlying the pathogenesis of neurological disorders as well as aiding in the discovery and development of future drug therapies. The greatest challenge is creating cell-based models that encapsulate the vast phenotypic presentations as well as the underlying genotypic etiology of these conditions. In this article, we discuss the recent advancements in cell-based models for understanding the pathophysiology of neurological disorders. We reviewed studies discussing the progression of cell-based models to the advancement of three-dimensional models and organoids that provide a more accurate model of the pathophysiology of neurological disorders in vivo. The better we understand how to create more precise models of the neurological system, the sooner we will be able to create patient-specific models and large libraries of these neurological disorders. While three-dimensional models can be used to discover the linking factors to connect the varying phenotypes, such models will also help to understand the early pathophysiology of these neurological disorders and how they are affected by their environment. The three-dimensional cell models will allow us to create more specific treatments and uncover potentially preventative measures in neurological disorders such as autism spectrum disorder, Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Jake Langlie
- Department of Otolaryngology, Hearing Research and Communication Disorders Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rahul Mittal
- Department of Otolaryngology, Hearing Research and Communication Disorders Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ariel Finberg
- Department of Otolaryngology, Hearing Research and Communication Disorders Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nathalie B Bencie
- Department of Otolaryngology, Hearing Research and Communication Disorders Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jeenu Mittal
- Department of Otolaryngology, Hearing Research and Communication Disorders Laboratory, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hossein Omidian
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Yadollah Omidi
- College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Adrien A Eshraghi
- Department of Otolaryngology, Hearing Research and Communication Disorders Laboratory; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami; Department of Biomedical Engineering, University of Miami, Coral Gables; Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
12
|
Farrow SL, Schierding W, Gokuladhas S, Golovina E, Fadason T, Cooper AA, O’Sullivan JM. Establishing gene regulatory networks from Parkinson's disease risk loci. Brain 2022; 145:2422-2435. [PMID: 35094046 PMCID: PMC9373962 DOI: 10.1093/brain/awac022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 12/02/2021] [Accepted: 12/20/2021] [Indexed: 11/25/2022] Open
Abstract
The latest meta-analysis of genome-wide association studies identified 90 independent variants across 78 genomic regions associated with Parkinson's disease, yet the mechanisms by which these variants influence the development of the disease remains largely elusive. To establish the functional gene regulatory networks associated with Parkinson's disease risk variants, we utilized an approach combining spatial (chromosomal conformation capture) and functional (expression quantitative trait loci) data. We identified 518 genes subject to regulation by 76 Parkinson's variants across 49 tissues, whicih encompass 36 peripheral and 13 CNS tissues. Notably, one-third of these genes were regulated via trans-acting mechanisms (distal; risk locus-gene separated by >1 Mb, or on different chromosomes). Of particular interest is the identification of a novel trans-expression quantitative trait loci-gene connection between rs10847864 and SYNJ1 in the adult brain cortex, highlighting a convergence between familial studies and Parkinson's disease genome-wide association studies loci for SYNJ1 (PARK20) for the first time. Furthermore, we identified 16 neurodevelopment-specific expression quantitative trait loci-gene regulatory connections within the foetal cortex, consistent with hypotheses suggesting a neurodevelopmental involvement in the pathogenesis of Parkinson's disease. Through utilizing Louvain clustering we extracted nine significant and highly intraconnected clusters within the entire gene regulatory network. The nine clusters are enriched for specific biological processes and pathways, some of which have not previously been associated with Parkinson's disease. Together, our results not only contribute to an overall understanding of the mechanisms and impact of specific combinations of Parkinson's disease variants, but also highlight the potential impact gene regulatory networks may have when elucidating aetiological subtypes of Parkinson's disease.
Collapse
Affiliation(s)
- Sophie L Farrow
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - William Schierding
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | | | - Evgeniia Golovina
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Tayaza Fadason
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| | - Antony A Cooper
- Australian Parkinson’s Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent’s Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Justin M O’Sullivan
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
- Australian Parkinson’s Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Brain Research New Zealand, The University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK
| |
Collapse
|
13
|
Prahl JD, Pierce SE, van der Schans EJC, Coetzee GA, Tyson T. The Parkinson's disease variant rs356182 regulates neuronal differentiation independently from alpha-synuclein. Hum Mol Genet 2022; 32:1-14. [PMID: 35866299 PMCID: PMC9837835 DOI: 10.1093/hmg/ddac161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/16/2022] [Accepted: 07/10/2022] [Indexed: 01/25/2023] Open
Abstract
One of the most significant risk variants for Parkinson's disease (PD), rs356182, is located at the PD-associated locus near the alpha-synuclein (α-syn) encoding gene, SNCA. SNCA-proximal variants, including rs356182, are thought to function in PD risk through enhancers via allele-specific regulatory effects on SNCA expression. However, this interpretation discounts the complex activity of genetic enhancers and possible non-conical functions of α-syn. Here we investigated a novel risk mechanism for rs356182. We use CRISPR-Cas9 in LUHMES cells, a model for dopaminergic midbrain neurons, to generate precise hemizygous lesions at rs356182. The PD-protective (A/-), PD-risk (G/-) and wild-type (A/G) clones were neuronally differentiated and then compared transcriptionally and morphologically. Among the affected genes was SNCA, whose expression was promoted by the PD-protective allele (A) and repressed in its absence. In addition to SNCA, hundreds of genes were differentially expressed and associated with neurogenesis and axonogenesis-an effect not typically ascribed to α-syn. We also found that the transcription factor FOXO3 specifically binds to the rs356182 A-allele in differentiated LUHMES cells. Finally, we compared the results from the rs356182-edited cells to our previously published knockouts of SNCA and found only minimal overlap between the sets of significant differentially expressed genes. Together, the data implicate a risk mechanism for rs356182 in which the risk-allele (G) is associated with abnormal neuron development, independent of SNCA expression. We speculate that these pathological effects manifest as a diminished population of dopaminergic neurons during development leading to the predisposition for PD later in life.
Collapse
Affiliation(s)
- Jordan D Prahl
- To whom correspondence should be addressed. Tel: +1 6162345793; Fax: +1 6162345001;
| | - Steven E Pierce
- Department of Neurodegenerative Research, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids MI 49503, USA
| | - Edwin J C van der Schans
- Department of Neurodegenerative Research, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids MI 49503, USA
| | | | | |
Collapse
|
14
|
Prahl J, Coetzee GA. Genetic Elements at the Alpha-Synuclein Locus. Front Neurosci 2022; 16:889802. [PMID: 35898413 PMCID: PMC9309432 DOI: 10.3389/fnins.2022.889802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022] Open
Abstract
Genome-wide association studies have consistently shown that the alpha-synuclein locus is significantly associated with Parkinson's disease. The mechanism by which this locus modulates the disease pathology and etiology remains largely under-investigated. This is due to the assumption that SNCA is the only driver of the functional aspects of several single nucleotide polymorphism (SNP) risk-signals at this locus. Recent evidence has shown that the risk associated with the top GWAS-identified variant within this locus is independent of SNCA expression, calling into question the validity of assigning function to the nearest gene, SNCA. In this review, we examine additional genes and risk variants present at the SNCA locus and how they may contribute to Parkinson's disease. Using the SNCA locus as an example, we hope to demonstrate that deeper and detailed functional validations are required for high impact disease-linked variants.
Collapse
Affiliation(s)
- Jordan Prahl
- Department of Neurodegenerative Disease, Van Andel Institute, Grand Rapids, MI, United States
| | | |
Collapse
|
15
|
Cukier HN, Kim H, Griswold AJ, Codreanu SG, Prince LM, Sherrod SD, McLean JA, Dykxhoorn DM, Ess KC, Hedera P, Bowman AB, Neely MD. Genomic, transcriptomic, and metabolomic profiles of hiPSC-derived dopamine neurons from clinically discordant brothers with identical PRKN deletions. NPJ Parkinsons Dis 2022; 8:84. [PMID: 35768426 PMCID: PMC9243035 DOI: 10.1038/s41531-022-00346-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
We previously reported on two brothers who carry identical compound heterozygous PRKN mutations yet present with significantly different Parkinson's Disease (PD) clinical phenotypes. Juvenile cases demonstrate that PD is not necessarily an aging-associated disease. Indeed, evidence for a developmental component to PD pathogenesis is accumulating. Thus, we hypothesized that the presence of additional genetic modifiers, including genetic loci relevant to mesencephalic dopamine neuron development, could potentially contribute to the different clinical manifestations of the two brothers. We differentiated human-induced pluripotent stem cells (hiPSCs) derived from the two brothers into mesencephalic neural precursor cells and early postmitotic dopaminergic neurons and performed wholeexome sequencing and transcriptomic and metabolomic analyses. No significant differences in the expression of canonical dopamine neuron differentiation markers were observed. Yet our transcriptomic analysis revealed a significant downregulation of the expression of three neurodevelopmentally relevant cell adhesion molecules, CNTN6, CNTN4 and CHL1, in the cultures of the more severely affected brother. In addition, several HLA genes, known to play a role in neurodevelopment, were differentially regulated. The expression of EN2, a transcription factor crucial for mesencephalic dopamine neuron development, was also differentially regulated. We further identified differences in cellular processes relevant to dopamine metabolism. Lastly, wholeexome sequencing, transcriptomics and metabolomics data all revealed differences in glutathione (GSH) homeostasis, the dysregulation of which has been previously associated with PD. In summary, we identified genetic differences which could potentially, at least partially, contribute to the discordant clinical PD presentation of the two brothers.
Collapse
Affiliation(s)
- Holly N Cukier
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hyunjin Kim
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simona G Codreanu
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Derek M Dykxhoorn
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kevin C Ess
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter Hedera
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, University of Louisville, Louisville, KY, USA
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, IN, USA.
| | - M Diana Neely
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
16
|
Early Signs of Molecular Defects in iPSC-Derived Neural Stems Cells from Patients with Familial Parkinson’s Disease. Biomolecules 2022; 12:biom12070876. [PMID: 35883433 PMCID: PMC9313424 DOI: 10.3390/biom12070876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/14/2022] [Accepted: 06/18/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disorder, classically associated with extensive loss of dopaminergic neurons of the substantia nigra pars compacta. The hallmark of the disease is the accumulation of pathogenic conformations of the presynaptic protein, α-synuclein (αSyn), and the formation of intraneuronal protein aggregate inclusions. Neurodegeneration of dopamine neurons leads to a prominent dopaminergic deficiency in the basal ganglia, responsible for motor disturbances. However, it is now recognized that the disease involves more widespread neuronal dysfunction, leading to early and late non-motor symptoms. The development of in vitro systems based on the differentiation of human-induced pluripotent stem cells provides us the unique opportunity to monitor alterations at the cellular and molecular level throughout the differentiation procedure and identify perturbations that occur early, even at the neuronal precursor stage. Here we aim to identify whether p.A53T-αSyn induced disturbances at the molecular level are already present in neural precursors. Towards this, we present data from transcriptomics analysis of control and p.A53T-αSyn NPCs showing altered expression in transcripts involved in axon guidance, adhesion, synaptogenesis, ion transport, and metabolism. The comparative analysis with the transcriptomics profile of p.A53T-αSyn neurons shows both distinct and overlapping pathways leading to neurodegeneration while meta-analysis with transcriptomics data from both neurodegenerative and neurodevelopmental disorders reveals that p.A53T-pathology has a significant overlap with the latter category. This is the first study showing that molecular dysregulation initiates early at the p.A53T-αSyn NPC level, suggesting that synucleinopathies may have a neurodevelopmental component.
Collapse
|
17
|
Zagare A, Barmpa K, Smajic S, Smits LM, Grzyb K, Grünewald A, Skupin A, Nickels SL, Schwamborn JC. Midbrain organoids mimic early embryonic neurodevelopment and recapitulate LRRK2-p.Gly2019Ser-associated gene expression. Am J Hum Genet 2022; 109:311-327. [PMID: 35077669 PMCID: PMC8874228 DOI: 10.1016/j.ajhg.2021.12.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/13/2021] [Indexed: 12/20/2022] Open
Abstract
Human brain organoid models that recapitulate the physiology and complexity of the human brain have a great potential for in vitro disease modeling, in particular for neurodegenerative diseases, such as Parkinson disease. In the present study, we compare single-cell RNA-sequencing data of human midbrain organoids to the developing human embryonic midbrain. We demonstrate that the in vitro model is comparable to its in vivo equivalents in terms of developmental path and cellular composition. Moreover, we investigate the potential of midbrain organoids for modeling early developmental changes in Parkinson disease. Therefore, we compare the single-cell RNA-sequencing data of healthy-individual-derived midbrain organoids to their isogenic LRRK2-p.Gly2019Ser-mutant counterparts. We show that the LRRK2 p.Gly2019Ser variant alters neurodevelopment, resulting in an untimely and incomplete differentiation with reduced cellular variability. Finally, we present four candidate genes, APP, DNAJC6, GATA3, and PTN, that might contribute to the LRRK2-p.Gly2019Ser-associated transcriptome changes that occur during early neurodevelopment.
Collapse
|
18
|
Prahl J, Pierce SE, Coetzee GA, Tyson T. Alpha-synuclein negatively controls cell proliferation in dopaminergic neurons. Mol Cell Neurosci 2022; 119:103702. [PMID: 35093507 DOI: 10.1016/j.mcn.2022.103702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/20/2022] [Accepted: 01/23/2022] [Indexed: 02/02/2023] Open
Abstract
As researchers grapple with the mechanisms and implications of alpha-synuclein (α-syn) in neuropathology, it is often forgotten that the function(s) of α-syn in healthy cells remain largely elusive. Previous work has relied on observing α-syn localization in the cell or using knockout mouse models. Here, we address the specific role of α-syn in human dopaminergic neurons by disrupting its gene (SNCA) in the human dopaminergic neuron cell line, LUHMES. SNCA-null cells were able to differentiate grossly normally and showed modest effects on gene expression. The effects on gene expression were monodirectional, resulting primarily in the significant decrease of expression for 401 genes, implicating them as direct, or indirect positive targets of α-syn. Gene ontological analysis of these genes showed enrichment in terms associated with proliferation, differentiation, and synapse activity. These results add to the tapestry of α-syn biological functions. SIGNIFICANCE STATEMENT: The normal functions of α-syn have remained controversial, despite its clear importance in Parkinson's Disease pathology, where it accumulates in Lewy bodies and contributes to neurodegeneration. Its name implies synaptic and nuclear functions, but how it participates at these locations has not been resolved. Via knock-out experiments in dopaminergic neurons, we implicate α-syn as a functional participant in synapse activity and in proliferation/differentiation, the latter being novel and provide insight into α-syn's role in neuronal development.
Collapse
Affiliation(s)
- Jordan Prahl
- Department of Neurodegenerative Research, Van Andel Institute, Grand Rapid, MI 49503, USA.
| | - Steven E Pierce
- Department of Neurodegenerative Research, Van Andel Institute, Grand Rapid, MI 49503, USA
| | - Gerhard A Coetzee
- Department of Neurodegenerative Research, Van Andel Institute, Grand Rapid, MI 49503, USA.
| | - Trevor Tyson
- Department of Neurodegenerative Research, Van Andel Institute, Grand Rapid, MI 49503, USA.
| |
Collapse
|
19
|
Pourtoy-Brasselet S, Sciauvaud A, Boza-Moran MG, Cailleret M, Jarrige M, Polvèche H, Polentes J, Chevet E, Martinat C, Peschanski M, Aubry L. Human iPSC-derived neurons reveal early developmental alteration of neurite outgrowth in the late-occurring neurodegenerative Wolfram syndrome. Am J Hum Genet 2021; 108:2171-2185. [PMID: 34699745 DOI: 10.1016/j.ajhg.2021.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/30/2021] [Indexed: 11/18/2022] Open
Abstract
Recent studies indicate that neurodegenerative processes that appear during childhood and adolescence in individuals with Wolfram syndrome (WS) occur in addition to early brain development alteration, which is clinically silent. Underlying pathological mechanisms are still unknown. We have used induced pluripotent stem cell-derived neural cells from individuals affected by WS in order to reveal their phenotypic and molecular correlates. We have observed that a subpopulation of Wolfram neurons displayed aberrant neurite outgrowth associated with altered expression of axon guidance genes. Selective inhibition of the ATF6α arm of the unfolded protein response prevented the altered phenotype, although acute endoplasmic reticulum stress response-which is activated in late Wolfram degenerative processes-was not detected. Among the drugs currently tried in individuals with WS, valproic acid was the one that prevented the pathological phenotypes. These results suggest that early defects in axon guidance may contribute to the loss of neurons in individuals with WS.
Collapse
Affiliation(s)
| | - Axel Sciauvaud
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes 91100, France; Université Paris-Saclay, INSERM, Univ Evry, Institut des Cellules Souches pour le Traitement et l'Étude des Maladies Monogéniques, Corbeil-Essonnes 91100, France
| | - Maria-Gabriela Boza-Moran
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes 91100, France; Université Paris-Saclay, INSERM, Univ Evry, Institut des Cellules Souches pour le Traitement et l'Étude des Maladies Monogéniques, Corbeil-Essonnes 91100, France
| | - Michel Cailleret
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes 91100, France; Université Paris-Saclay, INSERM, Univ Evry, Institut des Cellules Souches pour le Traitement et l'Étude des Maladies Monogéniques, Corbeil-Essonnes 91100, France
| | - Margot Jarrige
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes 91100, France; Université Paris-Saclay, INSERM, Univ Evry, Institut des Cellules Souches pour le Traitement et l'Étude des Maladies Monogéniques, Corbeil-Essonnes 91100, France; CECS/AFM, I-STEM, Corbeil-Essonnes 91100, France
| | | | | | - Eric Chevet
- INSERM U1242, Université Rennes 1, Rennes 35000, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes 35000, France
| | - Cécile Martinat
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes 91100, France; Université Paris-Saclay, INSERM, Univ Evry, Institut des Cellules Souches pour le Traitement et l'Étude des Maladies Monogéniques, Corbeil-Essonnes 91100, France
| | - Marc Peschanski
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes 91100, France; Université Paris-Saclay, INSERM, Univ Evry, Institut des Cellules Souches pour le Traitement et l'Étude des Maladies Monogéniques, Corbeil-Essonnes 91100, France; CECS/AFM, I-STEM, Corbeil-Essonnes 91100, France
| | - Laetitia Aubry
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes 91100, France; Université Paris-Saclay, INSERM, Univ Evry, Institut des Cellules Souches pour le Traitement et l'Étude des Maladies Monogéniques, Corbeil-Essonnes 91100, France.
| |
Collapse
|
20
|
Walter J, Bolognin S, Poovathingal SK, Magni S, Gérard D, Antony PMA, Nickels SL, Salamanca L, Berger E, Smits LM, Grzyb K, Perfeito R, Hoel F, Qing X, Ohnmacht J, Bertacchi M, Jarazo J, Ignac T, Monzel AS, Gonzalez-Cano L, Krüger R, Sauter T, Studer M, de Almeida LP, Tronstad KJ, Sinkkonen L, Skupin A, Schwamborn JC. The Parkinson's-disease-associated mutation LRRK2-G2019S alters dopaminergic differentiation dynamics via NR2F1. Cell Rep 2021; 37:109864. [PMID: 34686322 DOI: 10.1016/j.celrep.2021.109864] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 07/27/2021] [Accepted: 09/29/2021] [Indexed: 11/19/2022] Open
Abstract
Increasing evidence suggests that neurodevelopmental alterations might contribute to increase the susceptibility to develop neurodegenerative diseases. We investigate the occurrence of developmental abnormalities in dopaminergic neurons in a model of Parkinson's disease (PD). We monitor the differentiation of human patient-specific neuroepithelial stem cells (NESCs) into dopaminergic neurons. Using high-throughput image analyses and single-cell RNA sequencing, we observe that the PD-associated LRRK2-G2019S mutation alters the initial phase of neuronal differentiation by accelerating cell-cycle exit with a concomitant increase in cell death. We identify the NESC-specific core regulatory circuit and a molecular mechanism underlying the observed phenotypes. The expression of NR2F1, a key transcription factor involved in neurogenesis, decreases in LRRK2-G2019S NESCs, neurons, and midbrain organoids compared to controls. We also observe accelerated dopaminergic differentiation in vivo in NR2F1-deficient mouse embryos. This suggests a pathogenic mechanism involving the LRRK2-G2019S mutation, where the dynamics of dopaminergic differentiation are modified via NR2F1.
Collapse
Affiliation(s)
- Jonas Walter
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Silvia Bolognin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Suresh K Poovathingal
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Stefano Magni
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Deborah Gérard
- Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Paul M A Antony
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Sarah L Nickels
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Luis Salamanca
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Emanuel Berger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Lisa M Smits
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Rita Perfeito
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal
| | - Fredrik Hoel
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Xiaobing Qing
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Jochen Ohnmacht
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | | | - Javier Jarazo
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Tomasz Ignac
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Anna S Monzel
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Laura Gonzalez-Cano
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Transversal Translational Medicine, Luxembourg Institute of Health, Strassen, Luxembourg; Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg
| | - Thomas Sauter
- Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, 06108 Nice, France
| | - Luis Pereira de Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Karl J Tronstad
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Lasse Sinkkonen
- Department of Life Science and Medicine (DLSM), University of Luxembourg, 4362 Belvaux, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg; Center for Research of Biological Systems, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jens C Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Belvaux, Luxembourg.
| |
Collapse
|
21
|
Zietek T, Boomgaarden WAD, Rath E. Drug Screening, Oral Bioavailability and Regulatory Aspects: A Need for Human Organoids. Pharmaceutics 2021; 13:1280. [PMID: 34452240 PMCID: PMC8399541 DOI: 10.3390/pharmaceutics13081280] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/31/2022] Open
Abstract
The intestinal epithelium critically contributes to oral bioavailability of drugs by constituting an important site for drug absorption and metabolism. In particular, intestinal epithelial cells (IEC) actively serve as gatekeepers of drug and nutrient availability. IECs' transport processes and metabolism are interrelated to the whole-body metabolic state and represent potential points of origin as well as therapeutic targets for a variety of diseases. Human intestinal organoids represent a superior model of the intestinal epithelium, overcoming limitations of currently used in vitro models. Caco-2 cells or rodent explant models face drawbacks such as their cancer and non-human origin, respectively, but are commonly used to study intestinal nutrient absorption, enterocyte metabolism and oral drug bioavailability, despite poorly correlative data. In contrast, intestinal organoids allow investigating distinct aspects of bioavailability including spatial resolution of transport, inter-individual differences and high-throughput screenings. As several countries have already developed strategic roadmaps to phase out animal experiments for regulatory purposes, intestinal organoid culture and organ-on-a-chip technology in combination with in silico approaches are roads to go in the preclinical and regulatory setup and will aid implementing the 3Rs (reduction, refinement and replacement) principle in basic science.
Collapse
Affiliation(s)
- Tamara Zietek
- Doctors against Animal Experiments, 51143 Köln, Germany
| | | | - Eva Rath
- Chair of Nutrition and Immunology, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
22
|
Cerebral Organoids Derived from a Parkinson's Patient Exhibit Unique Pathogenesis from Chikungunya Virus Infection When Compared to a Non-Parkinson's Patient. Pathogens 2021; 10:pathogens10070913. [PMID: 34358063 PMCID: PMC8308834 DOI: 10.3390/pathogens10070913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/05/2021] [Accepted: 07/15/2021] [Indexed: 12/25/2022] Open
Abstract
(1) Background: Arboviruses of medical and veterinary significance have been identified on all seven continents, with every human and animal population at risk for exposure. Like arboviruses, chronic neurodegenerative diseases, like Alzheimer’s and Parkinson’s disease, are found wherever there are humans. Significant differences in baseline gene and protein expression have been determined between human-induced pluripotent stem cell lines derived from non-Parkinson’s disease individuals and from individuals with Parkinson’s disease. It was hypothesized that these inherent differences could impact cerebral organoid responses to viral infection. (2) Methods: In this study, cerebral organoids from a non-Parkinson’s and Parkinson’s patient were infected with Chikungunya virus and observed for two weeks. (3) Results: Parkinson’s organoids lost mass and exhibited a differential antiviral response different from non-Parkinson’s organoids. Neurotransmission data from both infected non-Parkinson’s and Parkinson’s organoids had dysregulation of IL-1, IL-10, and IL-6. These cytokines are associated with mood and could be contributing to persistent depression seen in patients following CHIKV infection. Both organoid types had increased expression of CXCL10, which is linked to demyelination. (4) Conclusions: The differential antiviral response of Parkinson’s organoids compared with non-Parkinson’s organoids highlights the need for more research in neurotropic infections in a neurologically compromised host.
Collapse
|
23
|
Pain B, Baquerre C, Coulpier M. Cerebral organoids and their potential for studies of brain diseases in domestic animals. Vet Res 2021; 52:65. [PMID: 33941270 PMCID: PMC8090903 DOI: 10.1186/s13567-021-00931-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
The brain is a complex organ and any model for studying it in its normal and pathological aspects becomes a tool of choice for neuroscientists. The mastering and dissemination of protocols allowing brain organoids development have paved the way for a whole range of new studies in the field of brain development, modeling of neurodegenerative or neurodevelopmental diseases, understanding tumors as well as infectious diseases that affect the brain. While studies are so far limited to the use of human cerebral organoids, there is a growing interest in having similar models in other species. This review presents what is currently developed in this field, with a particular focus on the potential of cerebral organoids for studying neuro-infectious diseases in human and domestic animals.
Collapse
Affiliation(s)
- Bertrand Pain
- Univ Lyon, Université Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, USC1361, Bron, France.
| | - Camille Baquerre
- Univ Lyon, Université Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, USC1361, Bron, France
| | - Muriel Coulpier
- UMR1161 Virologie, Anses, INRAE, École Nationale Vétérinaire D'Alfort, Université Paris-Est, Maisons-Alfort, France
| |
Collapse
|
24
|
Szlachcic WJ, Ziojla N, Kizewska DK, Kempa M, Borowiak M. Endocrine Pancreas Development and Dysfunction Through the Lens of Single-Cell RNA-Sequencing. Front Cell Dev Biol 2021; 9:629212. [PMID: 33996792 PMCID: PMC8116659 DOI: 10.3389/fcell.2021.629212] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/06/2021] [Indexed: 12/16/2022] Open
Abstract
A chronic inability to maintain blood glucose homeostasis leads to diabetes, which can damage multiple organs. The pancreatic islets regulate blood glucose levels through the coordinated action of islet cell-secreted hormones, with the insulin released by β-cells playing a crucial role in this process. Diabetes is caused by insufficient insulin secretion due to β-cell loss, or a pancreatic dysfunction. The restoration of a functional β-cell mass might, therefore, offer a cure. To this end, major efforts are underway to generate human β-cells de novo, in vitro, or in vivo. The efficient generation of functional β-cells requires a comprehensive knowledge of pancreas development, including the mechanisms driving cell fate decisions or endocrine cell maturation. Rapid progress in single-cell RNA sequencing (scRNA-Seq) technologies has brought a new dimension to pancreas development research. These methods can capture the transcriptomes of thousands of individual cells, including rare cell types, subtypes, and transient states. With such massive datasets, it is possible to infer the developmental trajectories of cell transitions and gene regulatory pathways. Here, we summarize recent advances in our understanding of endocrine pancreas development and function from scRNA-Seq studies on developing and adult pancreas and human endocrine differentiation models. We also discuss recent scRNA-Seq findings for the pathological pancreas in diabetes, and their implications for better treatment.
Collapse
Affiliation(s)
- Wojciech J. Szlachcic
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Natalia Ziojla
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Dorota K. Kizewska
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Marcelina Kempa
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Malgorzata Borowiak
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
25
|
Wulansari N, Darsono WHW, Woo HJ, Chang MY, Kim J, Bae EJ, Sun W, Lee JH, Cho IJ, Shin H, Lee SJ, Lee SH. Neurodevelopmental defects and neurodegenerative phenotypes in human brain organoids carrying Parkinson's disease-linked DNAJC6 mutations. SCIENCE ADVANCES 2021; 7:eabb1540. [PMID: 33597231 PMCID: PMC7888924 DOI: 10.1126/sciadv.abb1540] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 12/28/2020] [Indexed: 05/14/2023]
Abstract
Loss-of-function mutations of DNAJC6, encoding HSP40 auxilin, have recently been identified in patients with early-onset Parkinson's disease (PD). To study the roles of DNAJC6 in PD pathogenesis, we used human embryonic stem cells with CRISPR-Cas9-mediated gene editing. Here, we show that DNAJC6 mutations cause key PD pathologic features, i.e., midbrain-type dopamine (mDA) neuron degeneration, pathologic α-synuclein aggregation, increase of intrinsic neuronal firing frequency, and mitochondrial and lysosomal dysfunctions in human midbrain-like organoids (hMLOs). In addition, neurodevelopmental defects were also manifested in hMLOs carrying the mutations. Transcriptomic analyses followed by experimental validation revealed that defects in DNAJC6-mediated endocytosis impair the WNT-LMX1A signal during the mDA neuron development. Furthermore, reduced LMX1A expression during development caused the generation of vulnerable mDA neurons with the pathologic manifestations. These results suggest that the human model of DNAJC6-PD recapitulates disease phenotypes and reveals mechanisms underlying disease pathology, providing a platform for assessing therapeutic interventions.
Collapse
Affiliation(s)
- Noviana Wulansari
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Wahyu Handoko Wibowo Darsono
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hye-Ji Woo
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Mi-Yoon Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Jinil Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Eun-Jin Bae
- Department of Biomedical Sciences and Medicine, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ju-Hyun Lee
- Department of Anatomy, Brain Korea 21 PLUS Program for Biomedical Science, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Daejeon, Republic of Korea
- School of Electrical and Electronics Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences and Medicine, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Sang-Hun Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
- Hanyang Biomedical Research Institute, Hanyang University, Seoul, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Post-GWAS knowledge gap: the how, where, and when. NPJ PARKINSONS DISEASE 2020; 6:23. [PMID: 32964108 PMCID: PMC7481221 DOI: 10.1038/s41531-020-00125-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/13/2020] [Indexed: 01/05/2023]
Abstract
Genetic risk for complex diseases very rarely reflects only Mendelian-inherited phenotypes where single-gene mutations can be followed in families by linkage analysis. More commonly, a large set of low-penetrance, small effect-size variants combine to confer risk; they are normally revealed in genome-wide association studies (GWAS), which compare large population groups. Whereas Mendelian inheritance points toward disease mechanisms arising from the mutated genes, in the case of GWAS signals, the effector proteins and even general risk mechanism are mostly unknown. Instead, the utility of GWAS currently lies primarily in predictive and diagnostic information. Although an amazing body of GWAS-based knowledge now exists, we advocate for more funding towards the exploration of the fundamental biology in post-GWAS studies; this research will bring us closer to causality and risk gene identification. Using Parkinson's Disease as an example, we ask, how, where, and when do risk loci contribute to disease?
Collapse
|
27
|
Galet B, Cheval H, Ravassard P. Patient-Derived Midbrain Organoids to Explore the Molecular Basis of Parkinson's Disease. Front Neurol 2020; 11:1005. [PMID: 33013664 PMCID: PMC7500100 DOI: 10.3389/fneur.2020.01005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022] Open
Abstract
Induced pluripotent stem cell-derived organoids offer an unprecedented access to complex human tissues that recapitulate features of architecture, composition and function of in vivo organs. In the context of Parkinson's Disease (PD), human midbrain organoids (hMO) are of significant interest, as they generate dopaminergic neurons expressing markers of Substantia Nigra identity, which are the most vulnerable to degeneration. Combined with genome editing approaches, hMO may thus constitute a valuable tool to dissect the genetic makeup of PD by revealing the effects of risk variants on pathological mechanisms in a representative cellular environment. Furthermore, the flexibility of organoid co-culture approaches may also enable the study of neuroinflammatory and neurovascular processes, as well as interactions with other brain regions that are also affected over the course of the disease. We here review existing protocols to generate hMO, how they have been used so far to model PD, address challenges inherent to organoid cultures, and discuss applicable strategies to dissect the molecular pathophysiology of the disease. Taken together, the research suggests that this technology represents a promising alternative to 2D in vitro models, which could significantly improve our understanding of PD and help accelerate therapeutic developments.
Collapse
Affiliation(s)
- Benjamin Galet
- Molecular Pathophysiology of Parkinson's Disease Group, Paris Brain Institute (ICM), INSERM U, CNRS UMR 7225, Sorbonne University, Paris, France
| | | | | |
Collapse
|
28
|
Tang BL. RAB39B's role in membrane traffic, autophagy, and associated neuropathology. J Cell Physiol 2020; 236:1579-1592. [PMID: 32761840 DOI: 10.1002/jcp.29962] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/19/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022]
Abstract
Neuropathological disorders are increasingly associated with dysfunctions in neuronal membrane traffic and autophagy, with defects among members of the Rab family of small GTPases implicated. Mutations in the human Xq28 localized gene RAB39B have been associated with X-linked neurodevelopmental defects including macrocephaly, intellectual disability, autism spectrum disorder (ASD), as well as rare cases of early-onset Parkinson's disease (PD). Despite the finding that RAB39B regulates GluA2 trafficking and could thus influence synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit composition, reasons for the wide-ranging neuropathological consequences associated with RAB39B defects have been unclear. Recent studies have now unraveled possible mechanisms underlying the neuropathological roles of this brain-enriched small GTPase. Studies in RAB39B knockout mice showed that RAB39B interacts with components of Class I phosphatidylinositol-3-kinase (PI3K) signaling. In its absence, the PI3K-AKT-mechanistic target of rapamycin signaling pathway in neural progenitor cells (NPCs) is hyperactivated, which promotes NPC proliferation, leading to macrocephaly and ASD. Pertaining to early-onset PD, a complex of C9orf72, Smith-Magenis syndrome chromosome region candidate 8 and WD repeat domain 41 that functions in autophagy has been identified as a guanine nucleotide exchange factor of RAB39B. Here, recent findings that have shed light on our mechanistic understanding of RAB39B's role in neurodevelopmental and neurodegenerative pathologies are reviewed. Caveats and unanswered questions are also discussed, and future perspectives outlined.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,NUS Graduate School of Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
29
|
Smits LM, Schwamborn JC. Midbrain Organoids: A New Tool to Investigate Parkinson's Disease. Front Cell Dev Biol 2020; 8:359. [PMID: 32509785 PMCID: PMC7248385 DOI: 10.3389/fcell.2020.00359] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
The study of human 3D cell culture models not only bridges the gap between traditional 2D in vitro experiments and in vivo animal models, it also addresses processes that cannot be recapitulated by either of these traditional models. Therefore, it offers an opportunity to better understand complex biology including brain development. The brain organoid technology provides a physiologically relevant context, which holds great potential for its application in modeling neurological diseases. Here, we compare different methods to obtain highly specialized structures that resemble specific features of the human midbrain. Regionally patterned neural stem cells (NSCs) were utilized to derive such human midbrain-specific organoids (hMO). The resulting neural tissue exhibited abundant neurons with midbrain dopaminergic neuron identity, as well as astroglia and oligodendrocyte differentiation. Within the midbrain organoids, neurite myelination, and the formation of synaptic connections were observed. Regular neuronal fire patterning and neural network synchronicity were determined by multielectrode array recordings. In addition to electrophysiologically functional neurons producing and secreting dopamine, responsive neuronal subtypes, such as GABAergic and glutamatergic neurons were also detected. In order to model disorders like Parkinson's disease (PD) in vitro, midbrain organoids carrying a disease specific mutation were derived and compared to healthy control organoids to investigate relevant neurodegenerative pathophysiology. In this way midbrain-specific organoids constitute a powerful tool for human-specific in vitro modeling of neurological disorders with a great potential to be utilized in advanced therapy development.
Collapse
|
30
|
Monzel AS, Hemmer K, Kaoma T, Smits LM, Bolognin S, Lucarelli P, Rosety I, Zagare A, Antony P, Nickels SL, Krueger R, Azuaje F, Schwamborn JC. Machine learning-assisted neurotoxicity prediction in human midbrain organoids. Parkinsonism Relat Disord 2020; 75:105-109. [PMID: 32534431 DOI: 10.1016/j.parkreldis.2020.05.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Brain organoids are highly complex multi-cellular tissue proxies, which have recently risen as novel tools to study neurodegenerative diseases such as Parkinson's disease (PD). However, with increasing complexity of the system, usage of quantitative tools becomes challenging. OBJECTIVES The primary objective of this study was to develop a neurotoxin-induced PD organoid model and to assess the neurotoxic effect on dopaminergic neurons using microscopy-based phenotyping in a high-content fashion. METHODS We describe a pipeline for a machine learning-based analytical method, allowing for detailed image-based cell profiling and toxicity prediction in brain organoids treated with the neurotoxic compound 6-hydroxydopamine (6-OHDA). RESULTS We quantified features such as dopaminergic neuron count and neuronal complexity and built a machine learning classifier with the data to optimize data processing strategies and to discriminate between different treatment conditions. We validated the approach with high content imaging data from PD patient derived midbrain organoids. CONCLUSIONS The here described model is a valuable tool for advanced in vitro PD modeling and to test putative neurotoxic compounds.
Collapse
Affiliation(s)
- Anna S Monzel
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Kathrin Hemmer
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Tony Kaoma
- Computational Biomedicine Research Group, Quantitative Biology Unit, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
| | - Lisa M Smits
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Silvia Bolognin
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Philippe Lucarelli
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Isabel Rosety
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alise Zagare
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Paul Antony
- Clinical and Experimental Neurobiology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sarah L Nickels
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Rejko Krueger
- Clinical and Experimental Neurobiology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Parkinson's Research Clinic, Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg; Luxembourg Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Luxembourg
| | - Francisco Azuaje
- Computational Biomedicine Research Group, Quantitative Biology Unit, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg; Data and Translational Sciences, UCB Celltech, Slough, UK
| | - Jens C Schwamborn
- Developmental and Cellular Biology, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
31
|
McNeill RV, Ziegler GC, Radtke F, Nieberler M, Lesch KP, Kittel-Schneider S. Mental health dished up-the use of iPSC models in neuropsychiatric research. J Neural Transm (Vienna) 2020; 127:1547-1568. [PMID: 32377792 PMCID: PMC7578166 DOI: 10.1007/s00702-020-02197-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Genetic and molecular mechanisms that play a causal role in mental illnesses are challenging to elucidate, particularly as there is a lack of relevant in vitro and in vivo models. However, the advent of induced pluripotent stem cell (iPSC) technology has provided researchers with a novel toolbox. We conducted a systematic review using the PRISMA statement. A PubMed and Web of Science online search was performed (studies published between 2006–2020) using the following search strategy: hiPSC OR iPSC OR iPS OR stem cells AND schizophrenia disorder OR personality disorder OR antisocial personality disorder OR psychopathy OR bipolar disorder OR major depressive disorder OR obsessive compulsive disorder OR anxiety disorder OR substance use disorder OR alcohol use disorder OR nicotine use disorder OR opioid use disorder OR eating disorder OR anorexia nervosa OR attention-deficit/hyperactivity disorder OR gaming disorder. Using the above search criteria, a total of 3515 studies were found. After screening, a final total of 56 studies were deemed eligible for inclusion in our study. Using iPSC technology, psychiatric disease can be studied in the context of a patient’s own unique genetic background. This has allowed great strides to be made into uncovering the etiology of psychiatric disease, as well as providing a unique paradigm for drug testing. However, there is a lack of data for certain psychiatric disorders and several limitations to present iPSC-based studies, leading us to discuss how this field may progress in the next years to increase its utility in the battle to understand psychiatric disease.
Collapse
Affiliation(s)
- Rhiannon V McNeill
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Georg C Ziegler
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Franziska Radtke
- Department of Child and Adolescent Psychiatry, Psychosomatic Medicine and Psychotherapy University Hospital, University of Würzburg, Würzburg, Germany
| | - Matthias Nieberler
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Klaus-Peter Lesch
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
| |
Collapse
|
32
|
Genetic predispositions of Parkinson's disease revealed in patient-derived brain cells. NPJ PARKINSONS DISEASE 2020; 6:8. [PMID: 32352027 PMCID: PMC7181694 DOI: 10.1038/s41531-020-0110-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent neurological disorder and has been the focus of intense investigations to understand its etiology and progression, but it still lacks a cure. Modeling diseases of the central nervous system in vitro with human induced pluripotent stem cells (hiPSC) is still in its infancy but has the potential to expedite the discovery and validation of new treatments. Here, we discuss the interplay between genetic predispositions and midbrain neuronal impairments in people living with PD. We first summarize the prevalence of causal Parkinson's genes and risk factors reported in 74 epidemiological and genomic studies. We then present a meta-analysis of 385 hiPSC-derived neuronal lines from 67 recent independent original research articles, which point towards specific impairments in neurons from Parkinson's patients, within the context of genetic predispositions. Despite the heterogeneous nature of the disease, current iPSC models reveal converging molecular pathways underlying neurodegeneration in a range of familial and sporadic forms of Parkinson's disease. Altogether, consolidating our understanding of robust cellular phenotypes across genetic cohorts of Parkinson's patients may guide future personalized drug screens in preclinical research.
Collapse
|
33
|
Norepinephrine, neurodevelopment and behavior. Neurochem Int 2020; 135:104706. [PMID: 32092327 DOI: 10.1016/j.neuint.2020.104706] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 02/06/2023]
Abstract
Neurotransmitters play critical roles in the developing nervous system. Among the neurotransmitters, norepinephrine (NE) is in particular postulated to be an important regulator of brain development. NE is expressed during early stages of development and is known to regulate both the development of noradrenergic neurons and the development of target areas. NE participates in the shaping and the wiring of the nervous system during the critical periods of development, and perturbations in this process can alter the brain's developmental trajectory, which in turn can cause long-lasting and even permanent changes in the brain function and behavior later in life. Here we will briefly review evidence for the role of noradrenergic system in neurodevelopmental processes and will discuss about the potential disruptors of noradrenergic system during development and their behavioral consequences.
Collapse
|
34
|
Argentati C, Tortorella I, Bazzucchi M, Morena F, Martino S. Harnessing the Potential of Stem Cells for Disease Modeling: Progress and Promises. J Pers Med 2020; 10:E8. [PMID: 32041088 PMCID: PMC7151621 DOI: 10.3390/jpm10010008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/18/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
Ex vivo cell/tissue-based models are an essential step in the workflow of pathophysiology studies, assay development, disease modeling, drug discovery, and development of personalized therapeutic strategies. For these purposes, both scientific and pharmaceutical research have adopted ex vivo stem cell models because of their better predictive power. As matter of a fact, the advancing in isolation and in vitro expansion protocols for culturing autologous human stem cells, and the standardization of methods for generating patient-derived induced pluripotent stem cells has made feasible to generate and investigate human cellular disease models with even greater speed and efficiency. Furthermore, the potential of stem cells on generating more complex systems, such as scaffold-cell models, organoids, or organ-on-a-chip, allowed to overcome the limitations of the two-dimensional culture systems as well as to better mimic tissues structures and functions. Finally, the advent of genome-editing/gene therapy technologies had a great impact on the generation of more proficient stem cell-disease models and on establishing an effective therapeutic treatment. In this review, we discuss important breakthroughs of stem cell-based models highlighting current directions, advantages, and limitations and point out the need to combine experimental biology with computational tools able to describe complex biological systems and deliver results or predictions in the context of personalized medicine.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| |
Collapse
|
35
|
Chang M, Oh B, Choi J, Sulistio YA, Woo H, Jo A, Kim J, Kim E, Kim SW, Hwang J, Park J, Song J, Kwon O, Henry Kim H, Kim Y, Ko JY, Heo JY, Lee MJ, Lee M, Choi M, Chung SJ, Lee H, Lee S. LIN28A loss of function is associated with Parkinson's disease pathogenesis. EMBO J 2019; 38:e101196. [PMID: 31750563 PMCID: PMC6912061 DOI: 10.15252/embj.2018101196] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 10/01/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is neurodegenerative movement disorder characterized by degeneration of midbrain-type dopamine (mDA) neurons in the substantia nigra (SN). The RNA-binding protein Lin28 plays a role in neuronal stem cell development and neuronal differentiation. In this study, we reveal that Lin28 conditional knockout (cKO) mice show degeneration of mDA neurons in the SN, as well as PD-related behavioral deficits. We identify a loss-of-function variant of LIN28A (R192G substitution) in two early-onset PD patients. Using an isogenic human embryonic stem cell (hESC)/human induced pluripotent stem cell (hiPSC)-based disease model, we find that the Lin28 R192G variant leads to developmental defects and PD-related phenotypes in mDA neuronal cells that can be rescued by expression of wild-type Lin28A. Cell transplantation experiments in PD model rats show that correction of the LIN28A variant in the donor patient (pt)-hiPSCs leads to improved behavioral phenotypes. Our data link LIN28A to PD pathogenesis and suggest future personalized medicine targeting this variant in patients.
Collapse
|
36
|
Faravelli I, Costamagna G, Tamanini S, Corti S. Back to the origins: Human brain organoids to investigate neurodegeneration. Brain Res 2019; 1727:146561. [PMID: 31758922 DOI: 10.1016/j.brainres.2019.146561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 10/25/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023]
Abstract
Neurodegenerative disorders represent a high burden in terms of individual, social and economical resources. No ultimate therapy has been established so far; human brain morphology and development can not be entirely reproduced by animal models, and genomic, metabolic and biochemical differences might contribute to a limited predictive power for human translation. Thus, the development of human brain organoid models holds a wide potential to investigate the range of physiological and pathological features that characterise the early onset of the degeneration. Moreover, central nervous system development has gained a crucial role in the study of the pathogenesis of neurodegenerative disorders. Premature alterations during brain maturation have been related to late disease manifestations; genetic mutations responsible for neurodegeneration have been found in genes highly expressed during neural development. Elucidating the mechanisms triggering neuronal susceptibility to degeneration is crucial for pathogenetic studies and therapeutic discoveries. In the present work, we provide an overview on the current applications of human brain organoids towards studies of neurodegenerative diseases, with a survey on the recent discoveries and a closing discussion on the present challenges and future perspectives.
Collapse
Affiliation(s)
- I Faravelli
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - G Costamagna
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - S Tamanini
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - S Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
37
|
González-Casacuberta I, Juárez-Flores DL, Morén C, Garrabou G. Bioenergetics and Autophagic Imbalance in Patients-Derived Cell Models of Parkinson Disease Supports Systemic Dysfunction in Neurodegeneration. Front Neurosci 2019; 13:894. [PMID: 31551675 PMCID: PMC6748355 DOI: 10.3389/fnins.2019.00894] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder worldwide affecting 2-3% of the population over 65 years. This prevalence is expected to rise as life expectancy increases and diagnostic and therapeutic protocols improve. PD encompasses a multitude of clinical, genetic, and molecular forms of the disease. Even though the mechanistic of the events leading to neurodegeneration remain largely unknown, some molecular hallmarks have been repeatedly reported in most patients and models of the disease. Neuroinflammation, protein misfolding, disrupted endoplasmic reticulum-mitochondria crosstalk, mitochondrial dysfunction and consequent bioenergetic failure, oxidative stress and autophagy deregulation, are amongst the most commonly described. Supporting these findings, numerous familial forms of PD are caused by mutations in genes that are crucial for mitochondrial and autophagy proper functioning. For instance, late and early onset PD associated to mutations in Leucine-rich repeat kinase 2 (LRRK2) and Parkin (PRKN) genes, responsible for the most frequent dominant and recessive inherited forms of PD, respectively, have emerged as promising examples of disease due to their established role in commanding bioenergetic and autophagic balance. Concomitantly, the development of animal and cell models to investigate the etiology of the disease, potential biomarkers and therapeutic approaches are being explored. One of the emerging approaches in this context is the use of patient's derived cells models, such as skin-derived fibroblasts that preserve the genetic background and some environmental cues of the patients. An increasing number of reports in these PD cell models postulate that deficient mitochondrial function and impaired autophagic flux may be determinant in PD accelerated nigral cell death in terms of limitation of cell energy supply and accumulation of obsolete and/or unfolded proteins or dysfunctional organelles. The reliance of neurons on mitochondrial oxidative metabolism and their post-mitotic nature, may explain their increased vulnerability to undergo degeneration upon mitochondrial challenges or autophagic insults. In this scenario, proper mitochondrial function and turnover through mitophagy, are gaining in strength as protective targets to prevent neurodegeneration, together with the use of patient-derived fibroblasts to further explore these events. These findings point out the presence of molecular damage beyond the central nervous system (CNS) and proffer patient-derived cell platforms to the clinical and scientific community, which enable the study of disease etiopathogenesis and therapeutic approaches focused on modifying the natural history of PD through, among others, the enhancement of mitochondrial function and autophagy.
Collapse
Affiliation(s)
- Ingrid González-Casacuberta
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Diana Luz Juárez-Flores
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Constanza Morén
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| | - Gloria Garrabou
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences-University of Barcelona, Internal Medicine Service-Hospital Clínic of Barcelona, Barcelona, Spain.,CIBERER-U722, Madrid, Spain
| |
Collapse
|
38
|
Thibaut F. Psychiatric disorders: neurodevelopmental disorders, neurodegenerative disorders, or both? DIALOGUES IN CLINICAL NEUROSCIENCE 2019. [PMID: 30936765 PMCID: PMC6436953 DOI: 10.31887/dcns.2018.20.4/fthibaut] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Central nervous system disorders are traditionally dichotomized between early-onset neurodevelopmental and late-onset neurodegenerative diseases. Yet, there are commonalities in the mechanisms operating in both neurodevelopmental and neurodegenerative diseases.
Collapse
|
39
|
Parker GC. Translational Research on Stem Cells for Parkinson's Disease. Stem Cells Dev 2018; 27:949-950. [PMID: 29995610 DOI: 10.1089/scd.2018.29002.par] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Graham C Parker
- Department of Pediatrics and Children's Hospital of Michigan, Wayne State University School of Medicine , Detroit, Michigan
| |
Collapse
|