1
|
Znaidi R, Massiani-Beaudoin O, Mailly P, Monnet H, Bonnifet T, Joshi RL, Fuchs J. Nuclear translocation of the LINE-1 encoded ORF1 protein alters nuclear envelope integrity in human neurons. Brain Res 2025; 1857:149579. [PMID: 40157412 DOI: 10.1016/j.brainres.2025.149579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
LINE-1 retrotransposons are increasingly implicated in aging and neurodegenerative diseases, yet the precise pathogenic mechanisms remain elusive. While the endonuclease and reverse transcriptase activities of LINE-1-encoded ORF2p can induce DNA damage and inflammation, a role of LINE-1 ORF1p in cellular dysfunctions stays unassigned. Here we demonstrate, using a neuronal cellular model, that ORF1p translocates into the nucleus upon arsenite-induced stress, directly interacting with nuclear import (KPNB1), nuclear pore complex (NUP153), and nuclear lamina (Lamin B1) proteins. Nuclear translocation of ORF1p disrupts nuclear integrity, nucleocytoplasmic transport, and heterochromatin structure, features linked to neurodegeneration and aging. Elevated nuclear ORF1p levels induced either by arsenite-induced stress, ORF1p overexpression, or as observed in Parkinson's disease post-mortem brain tissues correlate with impaired nuclear envelope (NE) morphology. Stress-induced nuclear alterations are mitigated by blocking ORF1p nuclear import or with the anti-aging drug remodelin. This study thus reveals a pathogenic action of nuclear ORF1p in human neurons driving NE alterations and thereby contributing to LINE-1-mediated cell toxicity.
Collapse
Affiliation(s)
- Rania Znaidi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | | | - Philippe Mailly
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Héloïse Monnet
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Tom Bonnifet
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Rajiv L Joshi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| | - Julia Fuchs
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| |
Collapse
|
2
|
Boschen SL, A Mukerjee A, H Faroqi A, E Rabichow B, Fryer J. Research models to study lewy body dementia. Mol Neurodegener 2025; 20:46. [PMID: 40269912 PMCID: PMC12020038 DOI: 10.1186/s13024-025-00837-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: 12/18/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Lewy body dementia (LBD) encompasses neurodegenerative dementias characterized by cognitive fluctuations, visual hallucinations, and parkinsonism. Clinical differentiation of LBD from Alzheimer's disease (AD) remains complex due to symptom overlap, yet approximately 25% of dementia cases are diagnosed as LBD postmortem, primarily identified by the presence of α-synuclein aggregates, tau tangles, and amyloid plaques. These pathological features position LBD as a comorbid condition of both Parkinson's disease (PD) and AD, with over 50% of LBD cases exhibiting co-pathologies. LBD's mixed pathology complicates the development of comprehensive models that reflect the full spectrum of LBD's etiological, clinical, and pathological features. While existing animal and cellular models have facilitated significant discoveries in PD and AD research, they lack specificity in capturing LBD's unique pathogenic mechanisms, limiting the exploration of therapeutic avenues for LBD specifically. This review assesses widely used PD and AD models in terms of their relevance to LBD, particularly focusing on their ability to replicate human disease pathology and assess treatment efficacy. Furthermore, we discuss potential modifications to these models to advance the understanding of LBD mechanisms and propose innovative research directions aimed at developing models with enhanced etiological, face, predictive, and construct validity.
Collapse
Affiliation(s)
- Suelen Lucio Boschen
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
- Department of Neurosurgery, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| | - Aarushi A Mukerjee
- Department of Neuroscience, Mayo Clinic Jacksonville, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Ayman H Faroqi
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Ben E Rabichow
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - John Fryer
- Translational Genomics Research Institute, 445 N 5th St, Phoenix, AZ, 850054, USA
| |
Collapse
|
3
|
Jain G, Trombetta-Lima M, Matlahov I, Ribas HT, Chen T, Parlato R, Portale G, Dolga AM, van der Wel PCA. Inhibitor-based modulation of huntingtin aggregation mechanisms mitigates fibril-induced cellular stress. Nat Commun 2025; 16:3588. [PMID: 40234398 PMCID: PMC12000517 DOI: 10.1038/s41467-025-58691-9] [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: 05/16/2023] [Accepted: 03/31/2025] [Indexed: 04/17/2025] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder in which mutated fragments of the huntingtin protein (Htt) undergo misfolding and aggregation. Since aggregated proteins can cause cellular stress and cytotoxicity, there is an interest in the development of small molecule aggregation inhibitors as potential modulators of HD pathogenesis. Here, we study how a polyphenol modulates the aggregation mechanism of huntingtin exon 1 (HttEx1) even at sub-stoichiometric ratios. Sub-stoichiometric amounts of curcumin impacted the primary and/or secondary nucleation events, extending the pre-aggregation lag phase. Remarkably, the disrupted aggregation process changed both the aggregate structure and its cell metabolic properties. When administered to neuronal cells, the 'break-through' protein aggregates induced significantly reduced cellular stress compared to aggregates formed in absence of inhibitors. Structural analysis by electron microscopy, small angle X-ray scattering (SAXS), and solid-state NMR spectroscopy identified changes in the fibril structures, probing the flanking domains in the fuzzy coat and the fibril core. We propose that changes in the latter relate to the presence or absence of polyglutamine (polyQ) β-hairpin structures. Our findings highlight multifaceted consequences of small molecule inhibitors that modulate the protein misfolding landscape, with potential implications for treatment strategies in HD and other amyloid disorders.
Collapse
Affiliation(s)
- Greeshma Jain
- Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Irina Matlahov
- Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Hennrique Taborda Ribas
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
- Graduate Program in Biochemistry Sciences, Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Tingting Chen
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | - Raffaella Parlato
- Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Giuseppe Portale
- Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands.
| | - Patrick C A van der Wel
- Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
4
|
Witt B, Friese S, Walther V, Ebert F, Bornhorst J, Schwerdtle T. Cellular mechanisms of copper neurotoxicity in human, differentiated neurons. Arch Toxicol 2025; 99:689-699. [PMID: 39680088 PMCID: PMC11774975 DOI: 10.1007/s00204-024-03921-0] [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: 08/15/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024]
Abstract
Copper (Cu) is an essential trace element involved in fundamental physiological processes in the human body. Even slight disturbances in the physiological Cu homeostasis are associated with the manifestation of neurodegenerative diseases. While suggesting a crucial role of Cu in the pathogenesis, the exact mechanisms of Cu neurotoxicity involved in the onset and progression of neurological diseases are far from understood. This study focuses on the molecular and cellular mechanisms of Cu-mediated neurotoxicity in human brain cells. First, the cytotoxic potential of Cu was studied in fully differentiated, human neurons (LUHMES cells). Lysosomal integrity was considerably affected following incubation with 420 µM CuSO4 for 48 h. Further mechanistic studies revealed mitochondria and neuronal network as most susceptible target organelles (already at 100 µM CuSO4, 48 h), while the generation of reactive oxygen species turned out to be a rather later consequence of Cu toxicity. Besides Cu, the homeostasis of other elements might be involved and are likely to contribute to the pathology of Cu-mediated neurological disorders. Besides Cu, also effects on the cellular levels of magnesium, calcium, iron, and manganese were observed in the neurons, presumably aggravating the consequences of Cu neurotoxicity. In conclusion, insights in the underlying mode of action will foster the development of treatment strategies against Cu-mediated neurological diseases. Particularly, the interplay of Cu with other elements might provide a powerful diagnostic tool and might be used as therapeutic approach.
Collapse
Affiliation(s)
- Barbara Witt
- Division of Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, Erwin-Schroedinger-Str. 52, 67663, Kaiserslautern, Germany.
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Potsdam, Germany.
| | - Sharleen Friese
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Potsdam, Germany
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Vanessa Walther
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Potsdam, Germany
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Potsdam, Germany
| | - Julia Bornhorst
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Food Chemistry With Focus On Toxicology, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal, 14558, Potsdam, Germany
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Haid-Und-Neu-Straße 9, 76131, Karlsruhe, Germany
| |
Collapse
|
5
|
Marvian AT, Strauss T, Tang Q, Tuck BJ, Keeling S, Rüdiger D, Mirzazadeh Dizaji N, Mohammad-Beigi H, Nuscher B, Chakraborty P, Sutherland DS, McEwan WA, Köglsperger T, Zahler S, Zweckstetter M, Lichtenthaler SF, Wurst W, Schwarz S, Höglinger G. Distinct regulation of Tau Monomer and aggregate uptake and intracellular accumulation in human neurons. Mol Neurodegener 2024; 19:100. [PMID: 39736627 DOI: 10.1186/s13024-024-00786-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND The prion-like spreading of Tau pathology is the leading cause of disease progression in various tauopathies. A critical step in propagating pathologic Tau in the brain is the transport from the extracellular environment and accumulation inside naïve neurons. Current research indicates that human neurons internalize both the physiological extracellular Tau (eTau) monomers and the pathological eTau aggregates. However, similarities or differences in neuronal transport mechanisms between Tau species remain elusive. METHOD Monomers, oligomers, and fibrils of recombinant 2N4R Tau were produced and characterized by biochemical and biophysical methods. A neuronal eTau uptake and accumulation assay was developed for human induced pluripotent stem cell-derived neurons (iPSCNs) and Lund human mesencephalic cells (LUHMES)-derived neurons. Mechanisms of uptake and cellular accumulation of eTau species were studied by using small molecule inhibitors of endocytic mechanisms and siRNAs targeting Tau uptake mediators. RESULTS Extracellular Tau aggregates accumulated more than monomers in human neurons, mainly due to the higher efficiency of small fibrillar and soluble oligomeric aggregates in intraneuronal accumulation. A competition assay revealed a distinction in the neuronal accumulation between physiological eTau Monomers and pathology-relevant aggregates, suggesting differential transport mechanisms. Blocking heparan sulfate proteoglycans (HSPGs) with heparin only inhibited the accumulation of eTau aggregates, whereas monomers' uptake remained unaltered. At the molecular level, the downregulation of genes involved in HSPG synthesis exclusively blocked neuronal accumulation of eTau aggregates but not monomers, suggesting its role in the transport of pathologic Tau. Moreover, the knockdown of LRP1, as a receptor of Tau, mainly reduced the accumulation of monomeric form, confirming its involvement in Tau's physiological transport. CONCLUSION These data propose that despite the similarity in the cellular mechanism, the uptake and accumulation of eTau Monomers and aggregates in human neurons are regulated by different molecular mediators. Thus, they address the possibility of targeting the pathological spreading of Tau aggregates without disturbing the probable physiological or non-pathogenic transport of Tau Monomers.
Collapse
Affiliation(s)
- Amir T Marvian
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany.
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Tabea Strauss
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
| | - Qilin Tang
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
| | - Benjamin J Tuck
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Sophie Keeling
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel Rüdiger
- Department of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Negar Mirzazadeh Dizaji
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Hossein Mohammad-Beigi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs., Lyngby, Denmark
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000, Aarhus C, Denmark
| | - Brigitte Nuscher
- Division of Metabolic Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Pijush Chakraborty
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Gӧttingen, Germany
| | - Duncan S Sutherland
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000, Aarhus C, Denmark
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Thomas Köglsperger
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany
| | - Stefan Zahler
- Department of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Markus Zweckstetter
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Gӧttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Gӧttingen, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences, Technical University Munich, Freising, Germany
| | - Sigrid Schwarz
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Haag, Geriatric Clinic Haag, Oberbayern, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany.
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Department of Neurology, Hanover Medical School, Hanover, Germany.
- Center for Systems Neuroscience, Hanover, Germany.
| |
Collapse
|
6
|
Zuurbier KR, Fonseca RS, Arneaud SLB, Wall JM, Kim J, Tatge L, Otuzoglu G, Bali S, Metang P, Douglas PM. Yin Yang 1 and guanine quadruplexes protect dopaminergic neurons from cellular stress via transmissive dormancy. Nat Commun 2024; 15:10592. [PMID: 39632864 PMCID: PMC11618784 DOI: 10.1038/s41467-024-54958-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Neurons deploy diverse adaptive strategies to ensure survival and neurotransmission amid cellular stress. When these adaptive pathways are overwhelmed, functional impairment or neurodegeneration follows. Here we show that stressed neurons actively induce a state of transmissive dormancy as a protective measure. Extending observations of neurotrauma in C. elegans and mice, human dopaminergic neurons capable of surviving severe cellular challenges both decrease spontaneous activity and modulate dopamine homeostasis through the transcriptional regulator Yin Yang 1 (YY1). To bolster stress resilience and mitigate dopamine toxicity, YY1 increases expression of the vesicular monoamine transporter 2, vMAT2, while coordinately inhibiting dopamine synthesis through stabilization of a guanine quadruplex in intron 10 of tyrosine hydroxylase, TH. This dopaminergic stress response has the potential to cause circuit inactivation, yet safeguards neurons by minimizing the toxic accumulation of cytosolic dopamine and inducing a state of neuronal dormancy. In essence, neurons appear to actively prioritize viability over functionality.
Collapse
Affiliation(s)
- Kielen R Zuurbier
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Rene Solano Fonseca
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sonja L B Arneaud
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jordan M Wall
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Juhee Kim
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lexus Tatge
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Gupse Otuzoglu
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sofia Bali
- O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA
- Center for Alzheimer's and Neurodegenerative Diseases, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Patrick Metang
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Peter M Douglas
- Department of Molecular Biology; University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Hamon Center for Regenerative Science and Medicine; UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
7
|
Göksu AY. A review article on the development of dopaminergic neurons and establishment of dopaminergic neuron-based in vitro models by using immortal cell lines or stem cells to study and treat Parkinson's disease. Int J Dev Neurosci 2024; 84:817-842. [PMID: 39379284 DOI: 10.1002/jdn.10383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024] Open
Abstract
The primary pathological hallmark of Parkinson's disease (PD) is the degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta, a critical midbrain region. In vitro models based on DA neurons provide a powerful platform for investigating the cellular and molecular mechanisms of PD and testing novel therapeutic strategies. A deep understanding of DA neuron development, including the signalling pathways and transcription factors involved, is essential for advancing PD research. This article first explores the differentiation and maturation processes of DA neurons in the midbrain, detailing the relevant signalling pathways. It then compares various in vitro models, including primary cells, immortalized cell lines, and stem cell-based models, focusing on the advantages and limitations of each. Special attention is given to the role of immortalized and stem cell models in PD research. This review aims to guide researchers in selecting the most appropriate model for their specific research goals. Ethical considerations and clinical implications of using stem cells in PD research are also discussed.
Collapse
Affiliation(s)
- Azize Yasemin Göksu
- Department of Histology and Embryology, Department of Gene and Cell Therapy, Akdeniz University, School of Medicine, Antalya, Turkey
| |
Collapse
|
8
|
Kurita H, Masuda H, Okuda A, Go S, Ohuchi K, Yoshioka H, Fujimura M, Hozumi I, Inden M. Epigenetic alternations in the SYP and DLG4 genes due to low-level methylmercury exposure during neuronal differentiation in vitro. J Appl Toxicol 2024; 44:1986-1996. [PMID: 39187442 DOI: 10.1002/jat.4690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/28/2024]
Abstract
Methylmercury (MeHg) is an environmental toxin known to damage the central nervous system. When pregnant women ingest seafood, which may contain accumulated MeHg, fetal development may be affected. The embryonic period, a time of major epigenetic change, is susceptible to epigenetic disruptions due to chemical exposure. Therefore, understanding the molecular mechanism underlying MeHg's effects on neuronal development requires consideration of epigenetic factors. In this study, we investigated epigenetic modifications in the synaptophysin (SYP) and discs large MAGUK scaffold protein 4 (DLG4) genes. LUHMES cells were exposed to 1 nM MeHg for 6 days during days 2-8 of neural differentiation. MeHg exposure significantly reduced the number of spikes observed on day 16 of differentiation. Both mRNA and protein expression levels of SYP and DLG4 were significantly decreased by MeHg exposure. Additionally, MeHg treatment reduced acetyl histone H3 levels associated with transcriptional activity in the SYP gene while increasing histone H3 lysine 27 tri-methylation (H3K27me3) levels related to transcriptional repression. Conversely, regarding the DLG4 gene, MeHg exposure increased H3K27me3 levels. Differential changes in DNA methylation (high and low methylation states) were observed in the SYP and DLG4 genes due to MeHg exposure depending on CpG site position. In conclusion, this study suggests that epigenetic changes, particularly histone modifications, contribute to decreased MeHg exposure-induced SYP and DLG4 expression during neuronal differentiation.
Collapse
Affiliation(s)
- Hisaka Kurita
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Haruka Masuda
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Ayu Okuda
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Suzuna Go
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Kazuki Ohuchi
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hiroki Yoshioka
- Department of Pharmacy, Faculty of Pharmacy, Gifu University of Medical Science, Kani, Japan
- Department of Hygiene, School of Medicine, Kitasato University, Sagamihara, Japan
| | - Masatake Fujimura
- Basic Medical Sciences, National Institute for Minamata Disease, Minamata, Japan
| | - Isao Hozumi
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Masatoshi Inden
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
9
|
Couch ACM, Brown AM, Raimundo C, Solomon S, Taylor M, Sichlinger L, Matuleviciute R, Srivastava DP, Vernon AC. Transcriptional and cellular response of hiPSC-derived microglia-neural progenitor co-cultures exposed to IL-6. Brain Behav Immun 2024; 122:27-43. [PMID: 39098436 DOI: 10.1016/j.bbi.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
Elevated interleukin (IL-)6 levels during prenatal development have been linked to increased risk for neurodevelopmental disorders (NDD) in the offspring, but the mechanism remains unclear. Human-induced pluripotent stem cell (hiPSC) models offer a valuable tool to study the effects of IL-6 on features relevant for human neurodevelopment in vitro. We previously reported that hiPSC-derived microglia-like cells (MGLs) respond to IL-6, but neural progenitor cells (NPCs) in monoculture do not. Therefore, we investigated whether co-culturing hiPSC-derived MGLs with NPCs would trigger a cellular response to IL-6 stimulation via secreted factors from the MGLs. Using N=4 donor lines without psychiatric diagnosis, we first confirmed that NPCs can respond to IL-6 through trans-signalling when recombinant IL-6Ra is present, and that this response is dose-dependent. MGLs secreted soluble IL-6R, but at lower levels than found in vivo and below that needed to activate trans-signalling in NPCs. Whilst transcriptomic and secretome analysis confirmed that MGLs undergo substantial transcriptomic changes after IL-6 exposure and subsequently secrete a cytokine milieu, NPCs in co-culture with MGLs exhibited a minimal transcriptional response. Furthermore, there were no significant cell fate-acquisition changes when differentiated into post-mitotic cultures, nor alterations in synaptic densities in mature neurons. These findings highlight the need to investigate if trans-IL-6 signalling to NPCs is a relevant disease mechanism linking prenatal IL-6 exposure to increased risk for psychiatric disorders. Moreover, our findings underscore the importance of establishing more complex in vitro human models with diverse cell types, which may show cell-specific responses to microglia-released cytokines to fully understand how IL-6 exposure may influence human neurodevelopment.
Collapse
Affiliation(s)
- Amalie C M Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Amelia M Brown
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Catarina Raimundo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Shiden Solomon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Morgan Taylor
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Laura Sichlinger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Rugile Matuleviciute
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
10
|
Nagai M, Porter RS, Miyasato M, Wang A, Gavilan CM, Hughes ED, Wu MC, Saunders TL, Iwase S. Neuronal splicing of the unmethylated histone H3K4 reader, PHF21A, prevents excessive synaptogenesis. J Biol Chem 2024; 300:107881. [PMID: 39395799 PMCID: PMC11605454 DOI: 10.1016/j.jbc.2024.107881] [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: 01/17/2024] [Revised: 08/25/2024] [Accepted: 09/16/2024] [Indexed: 10/14/2024] Open
Abstract
PHF21A is a histone-binding protein that recognizes unmethylated histone H3K4, the reaction product of LSD1 histone demethylase. PHF21A and LSD1 form a complex, and both undergo neuron-specific microexon splicing. The PHF21A neuronal microexon interferes with nucleosome binding, whereas the LSD1 neuronal microexon weakens H3K4 demethylation activity and can alter the substrate specificity to H3K9 or H4K20. However, the temporal expression patterns of PHF21A and LSD1 splicing isoforms during brain development and their biological roles remain unknown. In this work, we report that neuronal PHF21A isoform expression precedes neuronal LSD1 expression during human neuron differentiation and mouse brain development. The asynchronous splicing events resulted in stepwise deactivation of the LSD1-PHF21A complex in reversing H3K4 methylation. An unbiased proteomics survey revealed that the enzymatically inactive LSD1-PHF21A complex interacts with neuron-specific binding partners, including MYT1-family transcription factors and post-transcriptional mRNA processing proteins such as VIRMA. The interaction with the neuron-specific components, however, did not require the PHF21A microexon, indicating that the neuronal proteomic milieu, rather than the microexon-encoded PHF21A segment, is responsible for neuron-specific complex formation. Finally, by using two Phf21a mutant mouse models, we found that Phf21a neuronal splicing prevents excess synapse formation that otherwise would occur when canonical PHF21A is expressed in neurons. These results suggest that the role of the PHF21A microexon is to dampen LSD1-mediated H3K4 demethylation, thereby containing aberrant synaptogenesis.
Collapse
Affiliation(s)
- Masayoshi Nagai
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert S Porter
- Genetics & Genomics Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Maxwell Miyasato
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Aijia Wang
- University of Michigan College of Literature, Science, and the Arts, Ann Arbor, Michigan, USA
| | - Cecilia M Gavilan
- Genetics & Genomics Graduate Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Elizabeth D Hughes
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, Michigan, USA; Division of Genetic Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA; Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA; Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
11
|
Nelke A, García-López S, Caso JR, Pereira MP. The therapeutic use of clonal neural stem cells in experimental Parkinson´s disease. Stem Cell Res Ther 2024; 15:356. [PMID: 39385216 PMCID: PMC11465761 DOI: 10.1186/s13287-024-03965-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Parkinson´s disease (PD), the second most common neurodegenerative disease in the world, is characterized by the death or impairment of dopaminergic neurons (DAn) in the substantia nigra pars compacta and dopamine depletion in the striatum. Currently, there is no cure for PD, and treatments only help to reduce the symptoms of the disease, and do not repair or replace the DAn damaged or lost in PD. Cell replacement therapy (CRT) seeks to relieve both pathological and symptomatic PD manifestations and has been shown to have beneficial effects in experimental PD models as well as in PD patients, but an apt cell line to be used in the treatment of PD has yet to be established. The purpose of this study was to examine the effects of the transplantation of hVM1 clone 32 cells, a bankable line of human neural stem cells (hNSCs), in a PD mouse model at four months post-transplant. METHODS Adult (five month-old) C57BL/6JRccHsd male mice were injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and subsequently transplanted with hVM1 clone 32 cells, or buffer, in the left striatum. Four months post-transplant, behavioral effects were explored using the open field and paw print tests, and histological analyses were performed. RESULTS Transplantation of hVM1 clone 32 cells rescued dopaminergic nigrostriatal populations in adult Parkinsonian mice. Motor and neurological deterioration were observed in buffer-treated mice, the latter of which had a tendency to improve in hNSC-transplanted mice. Detection of mast cell migration to the superficial cervical lymph nodes in cell-transplanted mice denoted a peripheral effect. Transplantation of hNSCs also rescued neuroblast neurogenesis in the subgranular zone, which was correlated with dopaminergic recovery and is indicative of local recovery mechanisms. CONCLUSIONS In this proof-of-concept study, the transplantation of hVM1 clone 32 cells provided neuroprotection in adult Parkinsonian mice by restoring the dopaminergic nigrostriatal pathway and hippocampal neurogenesis, demonstrating the efficacy of cell replacement therapy as a treatment for PD.
Collapse
Affiliation(s)
- Anna Nelke
- Unit of Molecular Neuropathology, Physiological and pathological processes Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Calle Nicolás Cabrera, 1, Madrid, 28049, Spain.
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, 28049, Spain.
- Institute for Molecular Biology - IUBM (Universidad Autónoma de Madrid), Madrid, Spain.
| | - Silvia García-López
- Unit of Molecular Neuropathology, Physiological and pathological processes Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Calle Nicolás Cabrera, 1, Madrid, 28049, Spain
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, 28049, Spain
- Institute for Molecular Biology - IUBM (Universidad Autónoma de Madrid), Madrid, Spain
| | - Javier R Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Avda. Complutense s/n, Madrid, 28040, Spain
| | - Marta P Pereira
- Unit of Molecular Neuropathology, Physiological and pathological processes Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Calle Nicolás Cabrera, 1, Madrid, 28049, Spain.
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, 28049, Spain.
- Institute for Molecular Biology - IUBM (Universidad Autónoma de Madrid), Madrid, Spain.
| |
Collapse
|
12
|
Rashidi SK, Khodagholi F, Rafie S, Kashipazha D, Safarian H, Khoshnam SE, Dezfouli MA. Methamphetamine and the brain: Emerging molecular targets and signaling pathways involved in neurotoxicity. TOXIN REV 2024; 43:553-571. [DOI: 10.1080/15569543.2024.2360425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/19/2024] [Accepted: 05/21/2024] [Indexed: 01/03/2025]
Affiliation(s)
- Seyed Khalil Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Rafie
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Neuroscience Lab, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Davood Kashipazha
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Neuroscience Lab, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Haleh Safarian
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Neuroscience Lab, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mitra Ansari Dezfouli
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Neuroscience Lab, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
13
|
Ghiasvand K, Amirfazli M, Moghimi P, Safari F, Takhshid MA. The role of neuron-like cell lines and primary neuron cell models in unraveling the complexity of neurodegenerative diseases: a comprehensive review. Mol Biol Rep 2024; 51:1024. [PMID: 39340590 DOI: 10.1007/s11033-024-09964-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Neurodegenerative diseases (NDs) are characterized by the progressive loss of neurons. As to developing effective therapeutic interventions, it is crucial to understand the underlying mechanisms of NDs. Cellular models have become invaluable tools for studying the complex pathogenesis of NDs, offering insights into disease mechanisms, determining potential therapeutic targets, and aiding in drug discovery. This review provides a comprehensive overview of various cellular models used in ND research, focusing on Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Cell lines, such as SH-SY5Y and PC12 cells, have emerged as valuable tools due to their ease of use, reproducibility, and scalability. Additionally, co-culture models, involving the growth of distinct cell types like neurons and astrocytes together, are highlighted for simulating brain interactions and microenvironment. While cell lines cannot fully replicate the complexity of the human brain, they provide a scalable method for examining important aspects of neurodegenerative diseases. Advancements in cell line technologies, including the incorporation of patient-specific genetic variants and improved co-culture models, hold promise for enhancing our understanding and expediting the development of effective treatments. Integrating multiple cellular models and advanced technologies offers the potential for significant progress in unraveling the intricacies of these debilitating diseases and improving patient outcomes.
Collapse
Affiliation(s)
- Kianoush Ghiasvand
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Amirfazli
- School of biological sciences, Illinois State University, Normal, United States of America
| | - Parvaneh Moghimi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
14
|
Nishimura K, Osaki H, Tezuka K, Nakashima D, Numata S, Masamizu Y. Recent advances and applications of human brain models. Front Neural Circuits 2024; 18:1453958. [PMID: 39161368 PMCID: PMC11330844 DOI: 10.3389/fncir.2024.1453958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Recent advances in human pluripotent stem cell (hPSC) technologies have prompted the emergence of new research fields and applications for human neurons and brain organoids. Brain organoids have gained attention as an in vitro model system that recapitulates the higher structure, cellular diversity and function of the brain to explore brain development, disease modeling, drug screening, and regenerative medicine. This progress has been accelerated by abundant interactions of brain organoid technology with various research fields. A cross-disciplinary approach with human brain organoid technology offers a higher-ordered advance for more accurately understanding the human brain. In this review, we summarize the status of neural induction in two- and three-dimensional culture systems from hPSCs and the modeling of neurodegenerative diseases using brain organoids. We also highlight the latest bioengineered technologies for the assembly of spatially higher-ordered neural tissues and prospects of brain organoid technology toward the understanding of the potential and abilities of the human brain.
Collapse
Affiliation(s)
- Kaneyasu Nishimura
- Laboratory of Functional Brain Circuit Construction, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | | | | | | | | | | |
Collapse
|
15
|
Bracha S, Johnson HJ, Pranckevicius NA, Catto F, Economides AE, Litvinov S, Hassi K, Rigoli MT, Cheroni C, Bonfanti M, Valenti A, Stucchi S, Attreya S, Ross PD, Walsh D, Malachi N, Livne H, Eshel R, Krupalnik V, Levin D, Cobb S, Koumoutsakos P, Caporale N, Testa G, Aguzzi A, Koshy AA, Sheiner L, Rechavi O. Engineering Toxoplasma gondii secretion systems for intracellular delivery of multiple large therapeutic proteins to neurons. Nat Microbiol 2024; 9:2051-2072. [PMID: 39075233 PMCID: PMC11306108 DOI: 10.1038/s41564-024-01750-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 06/05/2024] [Indexed: 07/31/2024]
Abstract
Delivering macromolecules across biological barriers such as the blood-brain barrier limits their application in vivo. Previous work has demonstrated that Toxoplasma gondii, a parasite that naturally travels from the human gut to the central nervous system (CNS), can deliver proteins to host cells. Here we engineered T. gondii's endogenous secretion systems, the rhoptries and dense granules, to deliver multiple large (>100 kDa) therapeutic proteins into neurons via translational fusions to toxofilin and GRA16. We demonstrate delivery in cultured cells, brain organoids and in vivo, and probe protein activity using imaging, pull-down assays, scRNA-seq and fluorescent reporters. We demonstrate robust delivery after intraperitoneal administration in mice and characterize 3D distribution throughout the brain. As proof of concept, we demonstrate GRA16-mediated brain delivery of the MeCP2 protein, a putative therapeutic target for Rett syndrome. By characterizing the potential and current limitations of the system, we aim to guide future improvements that will be required for broader application.
Collapse
Affiliation(s)
- Shahar Bracha
- Department of Neurobiology, Biochemistry and Biophysics, Wise Faculty of Life Sciences and Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA.
| | - Hannah J Johnson
- Neuroscience Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, USA
- Departments of Neurology and Immunobiology, College of Medicine, and BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Nicole A Pranckevicius
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Francesca Catto
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athena E Economides
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sergey Litvinov
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Karoliina Hassi
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Marco Tullio Rigoli
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Cristina Cheroni
- Human Technopole, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | | | - Alessia Valenti
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Sarah Stucchi
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Shruti Attreya
- Undergraduate Biology Research Program, University of Arizona, Tucson, AZ, USA
| | - Paul D Ross
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Daniel Walsh
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | | | | | | - Stuart Cobb
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Nicolò Caporale
- Human Technopole, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Giuseppe Testa
- Human Technopole, Milan, Italy.
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Anita A Koshy
- Departments of Neurology and Immunobiology, College of Medicine, and BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| | - Lilach Sheiner
- Centre for Parasitology, School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Oded Rechavi
- Department of Neurobiology, Biochemistry and Biophysics, Wise Faculty of Life Sciences and Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
16
|
Wang Q, Gu X, Yang L, Jiang Y, Zhang J, He J. Emerging perspectives on precision therapy for Parkinson's disease: multidimensional evidence leading to a new breakthrough in personalized medicine. Front Aging Neurosci 2024; 16:1417515. [PMID: 39026991 PMCID: PMC11254646 DOI: 10.3389/fnagi.2024.1417515] [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: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
PD is a prevalent and progressive neurodegenerative disorder characterized by both motor and non-motor symptoms. Genes play a significant role in the onset and progression of the disease. While the complexity and pleiotropy of gene expression networks have posed challenges for gene-targeted therapies, numerous pathways of gene variant expression show promise as therapeutic targets in preclinical studies, with some already in clinical trials. With the recognition of the numerous genes and complex pathways that can influence PD, it may be possible to take a novel approach to choose a treatment for the condition. This approach would be based on the symptoms, genomics, and underlying mechanisms of the disease. We discuss the utilization of emerging genetic and pathological knowledge of PD patients to categorize the disease into subgroups. Our long-term objective is to generate new insights for the therapeutic approach to the disease, aiming to delay and treat it more effectively, and ultimately reduce the burden on individuals and society.
Collapse
Affiliation(s)
- Qiaoli Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuan Gu
- Department of Trauma center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Le Yang
- Department of Endocrinology, The People’s Hospital of Jilin Province, Changchun, China
| | - Yan Jiang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiao Zhang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinting He
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Pantier R, Brown M, Han S, Paton K, Meek S, Montavon T, Shukeir N, McHugh T, Kelly DA, Hochepied T, Libert C, Jenuwein T, Burdon T, Bird A. MeCP2 binds to methylated DNA independently of phase separation and heterochromatin organisation. Nat Commun 2024; 15:3880. [PMID: 38719804 PMCID: PMC11079052 DOI: 10.1038/s41467-024-47395-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 03/29/2024] [Indexed: 05/12/2024] Open
Abstract
Correlative evidence has suggested that the methyl-CpG-binding protein MeCP2 contributes to the formation of heterochromatin condensates via liquid-liquid phase separation. This interpretation has been reinforced by the observation that heterochromatin, DNA methylation and MeCP2 co-localise within prominent foci in mouse cells. The findings presented here revise this view. MeCP2 localisation is independent of heterochromatin as MeCP2 foci persist even when heterochromatin organisation is disrupted. Additionally, MeCP2 foci fail to show hallmarks of phase separation in live cells. Importantly, we find that mouse cellular models are highly atypical as MeCP2 distribution is diffuse in most mammalian species, including humans. Notably, MeCP2 foci are absent in Mus spretus which is a mouse subspecies lacking methylated satellite DNA repeats. We conclude that MeCP2 has no intrinsic tendency to form condensates and its localisation is independent of heterochromatin. Instead, the distribution of MeCP2 in the nucleus is primarily determined by global DNA methylation patterns.
Collapse
Affiliation(s)
- Raphaël Pantier
- The Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, The King's Buildings, Edinburgh, EH9 3BF, UK
| | - Megan Brown
- The Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, The King's Buildings, Edinburgh, EH9 3BF, UK
| | - Sicheng Han
- The Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, The King's Buildings, Edinburgh, EH9 3BF, UK
| | - Katie Paton
- The Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, The King's Buildings, Edinburgh, EH9 3BF, UK
| | - Stephen Meek
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Thomas Montavon
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108, Freiburg, Germany
| | - Nicholas Shukeir
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108, Freiburg, Germany
| | - Toni McHugh
- The Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, The King's Buildings, Edinburgh, EH9 3BF, UK
| | - David A Kelly
- The Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, The King's Buildings, Edinburgh, EH9 3BF, UK
| | - Tino Hochepied
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Thomas Jenuwein
- Max Planck Institute of Immunobiology and Epigenetics, Stübeweg 51, 79108, Freiburg, Germany
| | - Tom Burdon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - Adrian Bird
- The Wellcome Centre for Cell Biology, University of Edinburgh, Michael Swann Building, Max Born Crescent, The King's Buildings, Edinburgh, EH9 3BF, UK.
| |
Collapse
|
18
|
Sharma K, Chib S, Gupta A, Singh R, Chalotra R. Interplay between α-synuclein and parkin genes: Insights of Parkinson's disease. Mol Biol Rep 2024; 51:586. [PMID: 38683365 DOI: 10.1007/s11033-024-09520-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Parkinson's disease (PD) is a complex and debilitating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra. The pathogenesis of PD is intimately linked to the roles of two key molecular players, α-synuclein (α-syn) and Parkin. Understanding the intricate interplay between α-syn and Parkin is essential for unravelling the molecular underpinnings of PD. Their roles in synaptic function and protein quality control underscore their significance in neuronal health. Dysregulation of these processes, as seen in PD, highlights the potential for targeted therapeutic strategies aimed at restoring normal protein homeostasis and mitigating neurodegeneration. Investigating the connections between α-syn, Parkin, and various pathological mechanisms provides insights into the complex web of factors contributing to PD pathogenesis and offers hope for the development of more effective treatments for this devastating neurological disorder. The present compilation provides an overview of their structures, regional and cellular locations, associations, physiological functions, and pathological roles in the context of PD.
Collapse
Affiliation(s)
- Kajal Sharma
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Shivani Chib
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Aniket Gupta
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India.
| | - Rishabh Chalotra
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| |
Collapse
|
19
|
Tong ZB, Huang R, Braisted J, Chu PH, Simeonov A, Gerhold DL. 3D-Suspension culture platform for high throughput screening of neurotoxic chemicals using LUHMES dopaminergic neurons. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100143. [PMID: 38280460 PMCID: PMC11056300 DOI: 10.1016/j.slasd.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/21/2023] [Accepted: 01/22/2024] [Indexed: 01/29/2024]
Abstract
Three-dimensional (3D) cell culture in vitro promises to improve representation of neuron physiology in vivo. This inspired development of a 3D culture platform for LUHMES (Lund Human Mesencephalic) dopaminergic neurons for high-throughput screening (HTS) of chemicals for neurotoxicity. Three culture platforms, adhesion (2D-monolayer), 3D-suspension, and 3D-shaken, were compared to monitor mRNA expression of seven neuronal marker genes, DCX, DRD2, ENO2, NEUROD4, SYN1, TH, and TUBB3. These seven marker genes reached similar maxima in all three formats, with the two 3D platforms showing similar kinetics, whereas several markers peaked earlier in 2D adhesion compared to both 3D culture platforms. The differentiated LUHMES (dLUHMES) neurons treated with ziram, methylmercury or thiram dynamically increased expression of metallothionein biomarker genes MT1G, MT1E and MT2A at 6 h. These gene expression increases were generally more dynamic in 2D adhesion cultures than in 3D cultures, but were generally comparable between 3D-suspension and 3D-u plate (low binding) platforms. Finally, we adapted 3D-suspension culture of dLUHMES and neural stem cells to 1536 well plates with a HTS cytotoxicity assay. This HTS assay revealed that cytotoxicity IC50 values were not significantly different between adhesion and 3D-suspension platforms for 31 of 34 (91%) neurotoxicants tested, whereas IC50 values were significantly different for at least two toxicants. In summary, the 3D-suspension culture platform for LUHMES dopaminergic neurons supported full differentiation and reproducible assay results, enabling quantitative HTS (qHTS) for cytotoxicity in 1536 well format with a Robust Z' score of 0.68.
Collapse
Affiliation(s)
- Zhi-Bin Tong
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - John Braisted
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - Pei-Hsuan Chu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States
| | - David L Gerhold
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States.
| |
Collapse
|
20
|
Nagai M, Porter RS, Hughes E, Saunders TL, Iwase S. Asynchronous microexon splicing of LSD1 and PHF21A during neurodevelopment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586181. [PMID: 38562691 PMCID: PMC10983945 DOI: 10.1101/2024.03.21.586181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
LSD1 histone H3K4 demethylase and its binding partner PHF21A, a reader protein for unmethylated H3K4, both undergo neuron-specific microexon splicing. The LSD1 neuronal microexon weakens H3K4 demethylation activity and can alter the substrate specificity to H3K9 or H4K20. Meanwhile, the PHF21A neuronal microexon interferes with nucleosome binding. However, the temporal expression patterns of LSD1 and PHF21A splicing isoforms during brain development remain unknown. In this work, we report that neuronal PHF21A isoform expression precedes neuronal LSD1 isoform expression during human neuron differentiation and mouse brain development. The asynchronous splicing events resulted in stepwise deactivation of the LSD1-PHF21A complex in reversing H3K4 methylation. We further show that the enzymatically inactive LSD1-PHF21A complex interacts with neuron-specific binding partners, including MYT1-family transcription factors and post-transcriptional mRNA processing proteins such as VIRMA. The interaction with the neuron-specific components, however, did not require the PHF21A microexon, indicating that the neuronal proteomic milieu, rather than the microexon-encoded PHF21A segment, is responsible for neuron-specific complex formation. These results indicate that the PHF21A microexon is dispensable for neuron-specific protein-protein interactions, yet the enzymatically inactive LSD1-PHF21A complex might have unique gene-regulatory roles in neurons.
Collapse
Affiliation(s)
- Masayoshi Nagai
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Robert S. Porter
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Elizabeth Hughes
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas L. Saunders
- Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
21
|
Bou Ghanem A, Hussayni Y, Kadbey R, Ratel Y, Yehya S, Khouzami L, Ghadieh HE, Kanaan A, Azar S, Harb F. Exploring the complexities of 1C metabolism: implications in aging and neurodegenerative diseases. Front Aging Neurosci 2024; 15:1322419. [PMID: 38239489 PMCID: PMC10794399 DOI: 10.3389/fnagi.2023.1322419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
The intricate interplay of one-carbon metabolism (OCM) with various cellular processes has garnered substantial attention due to its fundamental implications in several biological processes. OCM serves as a pivotal hub for methyl group donation in vital biochemical reactions, influencing DNA methylation, protein synthesis, and redox balance. In the context of aging, OCM dysregulation can contribute to epigenetic modifications and aberrant redox states, accentuating cellular senescence and age-associated pathologies. Furthermore, OCM's intricate involvement in cancer progression is evident through its capacity to provide essential one-carbon units crucial for nucleotide synthesis and DNA methylation, thereby fueling uncontrolled cell proliferation and tumor development. In neurodegenerative disorders like Alzheimer's and Parkinson's, perturbations in OCM pathways are implicated in the dysregulation of neurotransmitter synthesis and mitochondrial dysfunction, contributing to disease pathophysiology. This review underscores the profound impact of OCM in diverse disease contexts, reinforcing the need for a comprehensive understanding of its molecular complexities to pave the way for targeted therapeutic interventions across inflammation, aging and neurodegenerative disorders.
Collapse
Affiliation(s)
- Ayman Bou Ghanem
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Yaman Hussayni
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Raghid Kadbey
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Yara Ratel
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Shereen Yehya
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Lara Khouzami
- College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Hilda E. Ghadieh
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
- AUB Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| | - Amjad Kanaan
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Sami Azar
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
| | - Frederic Harb
- Faculty of Medicine and Medical Sciences, University of Balamand, Tripoli, Lebanon
- AUB Diabetes, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
22
|
de Wet S, Theart R, Loos B. Cogs in the autophagic machine-equipped to combat dementia-prone neurodegenerative diseases. Front Mol Neurosci 2023; 16:1225227. [PMID: 37720551 PMCID: PMC10500130 DOI: 10.3389/fnmol.2023.1225227] [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: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
Neurodegenerative diseases are often characterized by hydrophobic inclusion bodies, and it may be the case that the aggregate-prone proteins that comprise these inclusion bodies are in fact the cause of neurotoxicity. Indeed, the appearance of protein aggregates leads to a proteostatic imbalance that causes various interruptions in physiological cellular processes, including lysosomal and mitochondrial dysfunction, as well as break down in calcium homeostasis. Oftentimes the approach to counteract proteotoxicity is taken to merely upregulate autophagy, measured by an increase in autophagosomes, without a deeper assessment of contributors toward effective turnover through autophagy. There are various ways in which autophagy is regulated ranging from the mammalian target of rapamycin (mTOR) to acetylation status of proteins. Healthy mitochondria and the intracellular energetic charge they preserve are key for the acidification status of lysosomes and thus ensuring effective clearance of components through the autophagy pathway. Both mitochondria and lysosomes have been shown to bear functional protein complexes that aid in the regulation of autophagy. Indeed, it may be the case that minimizing the proteins associated with the respective neurodegenerative pathology may be of greater importance than addressing molecularly their resulting inclusion bodies. It is in this context that this review will dissect the autophagy signaling pathway, its control and the manner in which it is molecularly and functionally connected with the mitochondrial and lysosomal system, as well as provide a summary of the role of autophagy dysfunction in driving neurodegenerative disease as a means to better position the potential of rapamycin-mediated bioactivities to control autophagy favorably.
Collapse
Affiliation(s)
- Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Rensu Theart
- Department of Electric and Electronic Engineering, Stellenbosch University, Stellenbosch, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
23
|
Review on the interactions between dopamine metabolites and α-Synuclein in causing Parkinson's disease. Neurochem Int 2023; 162:105461. [PMID: 36460239 DOI: 10.1016/j.neuint.2022.105461] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Parkinson's disease (PD) is characterized by an abnormal post-translational modifications (PTM) in amino acid sequence and aggregation of alpha-synuclein (α-Syn) protein. It is generally believed that dopamine (DA) metabolite in dopaminergic (DAergic) neurons promotes the aggregation of toxic α-Syn oligomers and protofibrils, whereas DA inhibits the formation of toxic fibers and even degrades the toxic fibers. Therefore, the study on interaction between DA metabolites and α-Syn oligomers is one of the current hot topics in neuroscience, because this effect may have direct relevance to the selective DAergic neuron loss in PD. Several mechanisms have been reported for DA metabolites induced α-Syn oligomers viz. i) The reactive oxygen species (ROS) released during the auto-oxidation or enzymatic oxidation of DA changes the structure of α-Syn by the oxidation of amino acid residue leading to misfolding, ii) The oxidized DA metabolites directly interact with α-Syn through covalent or non-covalent bonding leading to the formation of oligomers, iii) DA interacts with lipid or autophagy related proteins to decreases the degradation efficiency of α-Syn aggregates. However, there is no clear-cut mechanism proposed for the interaction between DA and α-Syn. However, it is believed that the lysine (Lys) side chain of α-Syn sequence is the initial trigger site for the oligomer formation. Herein, we review different chemical mechanism involved during the interaction of Lys side chain of α-Syn with DA metabolites such as dopamine-o-quinone (DAQ), dopamine-chrome (DAC), dopamine-aldehyde (DOPAL) and neuromelanin. This review also provides the promotive effect of divalent Cu2+ ions on DA metabolites induced α-Syn oligomers and its inhibition effect by antioxidant glutathione (GSH).
Collapse
|
24
|
Gospodinova KO, Olsen D, Kaas M, Anderson SM, Phillips J, Walker RM, Bermingham ML, Payne AL, Giannopoulos P, Pandya D, Spires-Jones TL, Abbott CM, Porteous DJ, Glerup S, Evans KL. Loss of SORCS2 is Associated with Neuronal DNA Double-Strand Breaks. Cell Mol Neurobiol 2023; 43:237-249. [PMID: 34741697 PMCID: PMC9813074 DOI: 10.1007/s10571-021-01163-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/29/2021] [Indexed: 01/09/2023]
Abstract
SORCS2 is one of five proteins that constitute the Vps10p-domain receptor family. Members of this family play important roles in cellular processes linked to neuronal survival, differentiation and function. Genetic and functional studies implicate SORCS2 in cognitive function, as well as in neurodegenerative and psychiatric disorders. DNA damage and DNA repair deficits are linked to ageing and neurodegeneration, and transient neuronal DNA double-strand breaks (DSBs) also occur as a result of neuronal activity. Here, we report a novel role for SORCS2 in DSB formation. We show that SorCS2 loss is associated with elevated DSB levels in the mouse dentate gyrus and that knocking out SORCS2 in a human neuronal cell line increased Topoisomerase IIβ-dependent DSB formation and reduced neuronal viability. Neuronal stimulation had no impact on levels of DNA breaks in vitro, suggesting that the observed differences may not be the result of aberrant neuronal activity in these cells. Our findings are consistent with studies linking the VPS10 receptors and DNA damage to neurodegenerative conditions.
Collapse
Affiliation(s)
- Katerina O. Gospodinova
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Ditte Olsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Susan M. Anderson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Jonathan Phillips
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Rosie M. Walker
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK ,Present Address: University of Edinburgh, Chancellor’s Building, 49, Edinburgh, EH16 4SB UK
| | - Mairead L. Bermingham
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Abigail L. Payne
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Panagiotis Giannopoulos
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Divya Pandya
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Tara L. Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Catherine M. Abbott
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - David J. Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Kathryn L. Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| |
Collapse
|
25
|
LUHMES Cells: Phenotype Refinement and Development of an MPP +-Based Test System for Screening Antiparkinsonian Drugs. Int J Mol Sci 2023; 24:ijms24010733. [PMID: 36614176 PMCID: PMC9821222 DOI: 10.3390/ijms24010733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
The low effectiveness of symptomatic pharmacotherapy for Parkinson's disease (PD), which compensates for dopamine (DA) deficiency under degeneration of nigrostriatal dopaminergic (DAergic) neurons, could apparently be improved with neuroprotective therapy, which slows down neurodegeneration and PD progression. For this, it is necessary to have a DAergic cell line for the development of a PD model to screen neuroprotectors. We used immortalized human embryonic mesencephalon LUHMES cells (LCs) differentiated into DAergic neurons. The aim of this study was to characterize the phenotype of differentiated LCs and develop an 1-methyl-4-phenylpyridinium iodide (MPP+)-based test system for screening neuroprotectors. Using polymerase chain reaction (PCR) and immunocytochemistry, it has been shown that all differentiated LCs express genes and synthesize proteins characteristic of all neurons (microtubule-associated protein 2, bIII-tubulin, synaptotagmin 1) and specifically of DAergic neurons (tyrosine hydroxylase, aromatic L-amino acid decarboxylase, DA transporter, vesicular monoamine transporter 2). Furthermore, LCs are able to produce a small amount of DA, but under special conditions. To assess the mechanisms of neurodegeneration and neuroplasticity under the influence of toxins and antiparkinsonian drugs, including neuroprotectors, we have developed an LCs-based MPP+ PD model and proposed an original panel of markers for testing functional and structural cell disorders.
Collapse
|
26
|
Goloborshcheva VV, Kucheryanu VG, Voronina NA, Teterina EV, Ustyugov AA, Morozov SG. Synuclein Proteins in MPTP-Induced Death of Substantia Nigra Pars Compacta Dopaminergic Neurons. Biomedicines 2022; 10:biomedicines10092278. [PMID: 36140378 PMCID: PMC9496024 DOI: 10.3390/biomedicines10092278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/24/2022] Open
Abstract
Parkinson’s disease (PD) is one of the key neurodegenerative disorders caused by a dopamine deficiency in the striatum due to the death of dopaminergic (DA) neurons of the substantia nigra pars compacta. The initially discovered A53T mutation in the alpha-synuclein gene was linked to the formation of cytotoxic aggregates: Lewy bodies in the DA neurons of PD patients. Further research has contributed to the discovery of beta- and gamma-synucleins, which presumably compensate for the functional loss of either member of the synuclein family. Here, we review research from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity models and various synuclein-knockout animals. We conclude that the differences in the sensitivity of the synuclein-knockout animals compared with the MPTP neurotoxin are due to the ontogenetic selection of early neurons followed by a compensatory effect of beta-synuclein, which optimizes dopamine capture in the synapses. Triple-knockout synuclein studies have confirmed the higher sensitivity of DA neurons to the toxic effects of MPTP. Nonetheless, beta-synuclein could modulate the alpha-synuclein function, preventing its aggregation and loss of function. Overall, the use of knockout animals has helped to solve the riddle of synuclein functions, and these proteins could be promising molecular targets for the development of therapies that are aimed at optimizing the synaptic function of dopaminergic neurons.
Collapse
Affiliation(s)
- Valeria V. Goloborshcheva
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Correspondence: ; Tel.: +7-(909)-644-92-31
| | | | | | - Ekaterina V. Teterina
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia
| | - Aleksey A. Ustyugov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia
| | - Sergei G. Morozov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| |
Collapse
|
27
|
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: 1.3] [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
|
28
|
Allen CNS, Arjona SP, Santerre M, De Lucia C, Koch WJ, Sawaya BE. Metabolic Reprogramming in HIV-Associated Neurocognitive Disorders. Front Cell Neurosci 2022; 16:812887. [PMID: 35418836 PMCID: PMC8997587 DOI: 10.3389/fncel.2022.812887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/21/2022] [Indexed: 12/20/2022] Open
Abstract
A significant number of patients infected with HIV-1 suffer from HIV-associated neurocognitive disorders (HAND) such as spatial memory impairments and learning disabilities (SMI-LD). SMI-LD is also observed in patients using combination antiretroviral therapy (cART). Our lab has demonstrated that the HIV-1 protein, gp120, promotes SMI-LD by altering mitochondrial functions and energy production. We have investigated cellular processes upstream of the mitochondrial functions and discovered that gp120 causes metabolic reprogramming. Effectively, the addition of gp120 protein to neuronal cells disrupted the glycolysis pathway at the pyruvate level. Looking for the players involved, we found that gp120 promotes increased expression of polypyrimidine tract binding protein 1 (PTBP1), causing the splicing of pyruvate kinase M (PKM) into PKM1 and PKM2. We have also shown that these events lead to the accumulation of advanced glycation end products (AGEs) and prevent the cleavage of pro-brain-derived neurotrophic factor (pro-BDNF) protein into mature brain-derived neurotrophic factor (BDNF). The accumulation of proBDNF results in signaling that increases the expression of the inducible cAMP early repressor (ICER) protein which then occupies the cAMP response element (CRE)-binding sites within the BDNF promoters II and IV, thus altering normal synaptic plasticity. We reversed these events by adding Tepp-46, which stabilizes the tetrameric form of PKM2. Therefore, we concluded that gp120 reprograms cellular metabolism, causing changes linked to disrupted memory in HIV-infected patients and that preventing the disruption of the metabolism presents a potential cure against HAND progression.
Collapse
Affiliation(s)
- Charles N. S. Allen
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Sterling P. Arjona
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Claudio De Lucia
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Walter J. Koch
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Bassel E. Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- *Correspondence: Bassel E. Sawaya,
| |
Collapse
|
29
|
Bourseguin J, Cheng W, Talbot E, Hardy L, Lai J, Jeffries A, Lodato MA, Lee EA, Khoronenkova S. Persistent DNA damage associated with ATM kinase deficiency promotes microglial dysfunction. Nucleic Acids Res 2022; 50:2700-2718. [PMID: 35212385 PMCID: PMC8934660 DOI: 10.1093/nar/gkac104] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 01/21/2023] Open
Abstract
The autosomal recessive genome instability disorder Ataxia-telangiectasia, caused by mutations in ATM kinase, is characterized by the progressive loss of cerebellar neurons. We find that DNA damage associated with ATM loss results in dysfunctional behaviour of human microglia, immune cells of the central nervous system. Microglial dysfunction is mediated by the pro-inflammatory RELB/p52 non-canonical NF-κB transcriptional pathway and leads to excessive phagocytic clearance of neuronal material. Activation of the RELB/p52 pathway in ATM-deficient microglia is driven by persistent DNA damage and is dependent on the NIK kinase. Activation of non-canonical NF-κB signalling is also observed in cerebellar microglia of individuals with Ataxia-telangiectasia. These results provide insights into the underlying mechanisms of aberrant microglial behaviour in ATM deficiency, potentially contributing to neurodegeneration in Ataxia-telangiectasia.
Collapse
Affiliation(s)
- Julie Bourseguin
- Department of Biochemistry, University of Cambridge, 80 Tennis Court road, CambridgeCB2 1GA, UK
| | - Wen Cheng
- Department of Biochemistry, University of Cambridge, 80 Tennis Court road, CambridgeCB2 1GA, UK
| | - Emily Talbot
- Department of Biochemistry, University of Cambridge, 80 Tennis Court road, CambridgeCB2 1GA, UK
| | - Liana Hardy
- Department of Biochemistry, University of Cambridge, 80 Tennis Court road, CambridgeCB2 1GA, UK
| | - Jenny Lai
- Division of Genetics and Genomics, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Neuroscience, Harvard University, Boston, MA 02115, USA
| | - Ailsa M Jeffries
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Michael A Lodato
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Boston Children's Hospital; Department of Pediatrics, Harvard Medical School, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Svetlana V Khoronenkova
- Department of Biochemistry, University of Cambridge, 80 Tennis Court road, CambridgeCB2 1GA, UK
| |
Collapse
|
30
|
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: 0.3] [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
|
31
|
Spathopoulou A, Edenhofer F, Fellner L. Targeting α-Synuclein in Parkinson's Disease by Induced Pluripotent Stem Cell Models. Front Neurol 2022; 12:786835. [PMID: 35145469 PMCID: PMC8821105 DOI: 10.3389/fneur.2021.786835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/24/2021] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disorder characterized by motor and non-motor symptoms. To date, no specific treatment to halt disease progression is available, only medication to alleviate symptoms can be prescribed. The main pathological hallmark of PD is the development of neuronal inclusions, positive for α-synuclein (α-syn), which are termed Lewy bodies (LBs) or Lewy neurites. However, the cause of the inclusion formation and the loss of neurons remain largely elusive. Various genetic determinants were reported to be involved in PD etiology, including SNCA, DJ-1, PRKN, PINK1, LRRK2, and GBA. Comprehensive insights into pathophysiology of PD critically depend on appropriate models. However, conventional model organisms fall short to faithfully recapitulate some features of this complex disease and as a matter-of-fact access to physiological tissue is limiting. The development of disease models replicating PD that are close to human physiology and dynamic enough to analyze the underlying molecular mechanisms of disease initiation and progression, as well as the generation of new treatment options, is an important and overdue step. Recently, the establishment of induced pluripotent stem cell (iPSC)-derived neural models, particularly from genetic PD-variants, developed into a promising strategy to investigate the molecular mechanisms regarding formation of inclusions and neurodegeneration. As these iPSC-derived neurons can be generated from accessible biopsied samples of PD patients, they carry pathological alterations and enable the possibility to analyze the differences compared to healthy neurons. This review focuses on iPSC models carrying genetic PD-variants of α-syn that will be especially helpful in elucidating the pathophysiological mechanisms of PD. Furthermore, we discuss how iPSC models can be instrumental in identifying cellular targets, potentially leading to the development of new therapeutic treatments. We will outline the enormous potential, but also discuss the limitations of iPSC-based α-syn models.
Collapse
|
32
|
Nelke A, García-López S, Martínez-Serrano A, Pereira MP. Multifactoriality of Parkinson's Disease as Explored Through Human Neural Stem Cells and Their Transplantation in Middle-Aged Parkinsonian Mice. Front Pharmacol 2022; 12:773925. [PMID: 35126116 PMCID: PMC8807563 DOI: 10.3389/fphar.2021.773925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease (PD) is an age-associated neurodegenerative disorder for which there is currently no cure. Cell replacement therapy is a potential treatment for PD; however, this therapy has more clinically beneficial outcomes in younger patients with less advanced PD. In this study, hVM1 clone 32 cells, a line of human neural stem cells, were characterized and subsequently transplanted in middle-aged Parkinsonian mice in order to examine cell replacement therapy as a treatment for PD. In vitro analyses revealed that these cells express standard dopamine-centered markers as well as others associated with mitochondrial and peroxisome function, as well as glucose and lipid metabolism. Four months after the transplantation of the hVM1 clone 32 cells, striatal expression of tyrosine hydroxylase was minimally reduced in all Parkinsonian mice but that of dopamine transporter was decreased to a greater extent in buffer compared to cell-treated mice. Behavioral tests showed marked differences between experimental groups, and cell transplant improved hyperactivity and gait alterations, while in the striatum, astroglial populations were increased in all groups due to age and a higher amount of microglia were found in Parkinsonian mice. In the motor cortex, nonphosphorylated neurofilament heavy was increased in all Parkinsonian mice. Overall, these findings demonstrate that hVM1 clone 32 cell transplant prevented motor and non-motor impairments and that PD is a complex disorder with many influencing factors, thus reinforcing the idea of novel targets for PD treatment that tend to be focused on dopamine and nigrostriatal damage.
Collapse
Affiliation(s)
- Anna Nelke
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Madrid, Spain
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Madrid, Spain
| | - Silvia García-López
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Madrid, Spain
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Martínez-Serrano
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Madrid, Spain
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta P. Pereira
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Madrid, Spain
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
33
|
Marino G, Calabresi P, Ghiglieri V. Alpha-synuclein and cortico-striatal plasticity in animal models of Parkinson disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:153-166. [PMID: 35034731 DOI: 10.1016/b978-0-12-819410-2.00008-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Alpha-synuclein (α-synuclein) is a small, acidic protein containing 140 amino acids, highly expressed in the brain and primarily localized in the presynaptic terminals. It is found in high concentrations in Lewy Bodies, proteinaceous aggregates that constitute a typical histopathologic hallmark of Parkinson's disease. Altered environmental conditions, genetic mutations and post-translational changes can trigger abnormal aggregation processes with the increased frequency of oligomers, protofibrils, and fibrils formation that perturbs the neuronal homeostasis leading to cell death. Relevant to neuronal activity, a function of α-synuclein that has been extensively detailed is its regulatory actions in the trafficking of synaptic vesicles, including the processes of exocytosis, endocytosis and neurotransmitter release. Most recently, increasing attention has been paid to the possible role that α-synuclein plays at a postsynaptic level by interacting with selective subunits of the glutamate N-methyl-d-aspartate receptor, altering the corticostriatal plasticity of distinct neuronal populations.
Collapse
Affiliation(s)
- Gioia Marino
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy; Dipartimento di Medicina, Università degli Studi di Perugia, Perugia, Italy
| | - Paolo Calabresi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy; UOC Neurologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | |
Collapse
|
34
|
Bougea A, Stefanis L, Chrousos G. Stress system and related biomarkers in Parkinson's disease. Adv Clin Chem 2022; 111:177-215. [DOI: 10.1016/bs.acc.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
35
|
Shan FY, Fung KM, Zieneldien T, Kim J, Cao C, Huang JH. Examining the Toxicity of α-Synuclein in Neurodegenerative Disorders. Life (Basel) 2021; 11:life11111126. [PMID: 34833002 PMCID: PMC8621244 DOI: 10.3390/life11111126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Neurodegenerative disorders are complex disorders that display a variety of clinical manifestations. The second-most common neurodegenerative disorder is Parkinson’s disease, and the leading pathological protein of the disorder is considered to be α-synuclein. Nonetheless, α-synuclein accumulation also seems to result in multiple system atrophy and dementia with Lewy bodies. In order to obtain a more proficient understanding in the pathological progression of these synucleinopathies, it is crucial to observe the post-translational modifications of α-synuclein and the conformations of α-synuclein, as well as its role in the dysfunction of cellular pathways. Abstract α-synuclein is considered the main pathological protein in a variety of neurodegenerative disorders, such as Parkinson’s disease, multiple system atrophy, and dementia with Lewy bodies. As of now, numerous studies have been aimed at examining the post-translational modifications of α-synuclein to determine their effects on α-synuclein aggregation, propagation, and oligomerization, as well as the potential cellular pathway dysfunctions caused by α-synuclein, to determine the role of the protein in disease progression. Furthermore, α-synuclein also appears to contribute to the fibrilization of tau and amyloid beta, which are crucial proteins in Alzheimer’s disease, advocating for α-synuclein’s preeminent role in neurodegeneration. Due to this, investigating the mechanisms of toxicity of α-synuclein in neurodegeneration may lead to a more proficient understanding of the timeline progression in neurodegenerative synucleinopathies and could thereby lead to the development of potent targeted therapies.
Collapse
Affiliation(s)
- Frank Y. Shan
- Department of Anatomic Pathology, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA
- Correspondence: (F.Y.S.); (T.Z.)
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Medical Center, University of Oklahoma, Norman, OK 73019, USA;
| | - Tarek Zieneldien
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
- Correspondence: (F.Y.S.); (T.Z.)
| | - Janice Kim
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Jason H. Huang
- Department of Neurosurgery, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA;
| |
Collapse
|
36
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
37
|
Vazquez-Villasenor I, Garwood CJ, Simpson JE, Heath PR, Mortiboys H, Wharton SB. Persistent DNA damage alters the neuronal transcriptome suggesting cell cycle dysregulation and altered mitochondrial function. Eur J Neurosci 2021; 54:6987-7005. [PMID: 34536321 DOI: 10.1111/ejn.15466] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 08/14/2021] [Accepted: 09/06/2021] [Indexed: 12/01/2022]
Abstract
Oxidative DNA damage induces changes in the neuronal cell cycle and activates a DNA damage response (DDR) to promote repair, but these processes may be altered under a chronic oxidative environment, leading to the accumulation of unrepaired DNA damage and continued activation of a DDR. Failure to repair DNA damage can lead to apoptosis or senescence, which is characterized by a permanent cell cycle arrest. Increased oxidative stress and accumulation of oxidative DNA damage are features of brain ageing and neurodegeneration, but the effects of persistent DNA damage in neurons are not well characterized. We developed a model of persistent oxidative DNA damage in immortalized post-mitotic neurons in vitro by exposing them to a sublethal concentration of hydrogen peroxide following a 'double stress' protocol and performed a detailed characterization of the neuronal transcriptome using microarray analysis. Persistent DNA damage significantly altered the expression of genes involved in cell cycle regulation, DDR and repair mechanisms, and mitochondrial function, suggesting an active DDR response to replication stress and alterations in mitochondrial electron transport chain. Quantitative polymerase chain reaction (qPCR) and functional validation experiments confirmed hyperactivation of mitochondrial Complex I in response to persistent DNA damage. These changes in response to persistent oxidative DNA damage may lead to further oxidative stress, contributing to neuronal dysfunction and ultimately neurodegeneration.
Collapse
Affiliation(s)
| | - Claire J Garwood
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| |
Collapse
|
38
|
Shin EJ, Jeong JH, Hwang Y, Sharma N, Dang DK, Nguyen BT, Nah SY, Jang CG, Bing G, Nabeshima T, Kim HC. Methamphetamine-induced dopaminergic neurotoxicity as a model of Parkinson's disease. Arch Pharm Res 2021; 44:668-688. [PMID: 34286473 DOI: 10.1007/s12272-021-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, 900000, Can Tho City, Vietnam
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, 05029, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Guoying Bing
- Department of Neuroscience, College of Medicine, University of Kentucky, KY, 40536, Lexington, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Science, Fujita Health University, 470-1192, Toyoake, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea. .,Neuropsychopharmacology & Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.
| |
Collapse
|
39
|
Calamini B, Geyer N, Huss-Braun N, Bernhardt A, Harsany V, Rival P, Cindhuchao M, Hoffmann D, Gratzer S. Development of a physiologically relevant and easily scalable LUHMES cell-based model of G2019S LRRK2-driven Parkinson's disease. Dis Model Mech 2021; 14:dmm048017. [PMID: 34114604 PMCID: PMC8214734 DOI: 10.1242/dmm.048017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/27/2021] [Indexed: 11/20/2022] Open
Abstract
Parkinson's disease (PD) is a fatal neurodegenerative disorder that is primarily caused by the degeneration and loss of dopaminergic neurons of the substantia nigra in the ventral midbrain. Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of late-onset PD identified to date, with G2019S being the most frequent LRRK2 mutation, which is responsible for up to 1-2% of sporadic PD and up to 6% of familial PD cases. As no treatment is available for this devastating disease, developing new therapeutic strategies is of foremost importance. Cellular models are commonly used for testing novel potential neuroprotective compounds. However, current cellular PD models either lack physiological relevance to dopaminergic neurons or are too complex and costly for scaling up the production process and for screening purposes. In order to combine biological relevance and throughput, we have developed a PD model in Lund human mesencephalic (LUHMES) cell-derived dopaminergic neurons by overexpressing wild-type (WT) and G2019S LRRK2 proteins. We show that these cells can differentiate into dopaminergic-like neurons and that expression of mutant LRRK2 causes a range of different phenotypes, including reduced nuclear eccentricity, altered mitochondrial and lysosomal morphologies, and increased dopaminergic cell death. This model could be used to elucidate G2019S LRRK2-mediated dopaminergic neural dysfunction and to identify novel molecular targets for disease intervention. In addition, our model could be applied to high-throughput and phenotypic screenings for the identification of novel PD therapeutics.
Collapse
Affiliation(s)
- Barbara Calamini
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Nathalie Geyer
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Nathalie Huss-Braun
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Annie Bernhardt
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Véronique Harsany
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| | - Pierrick Rival
- BioTherapeutics/e-Biology - Bioinformatics, Sanofi Biologics Research, 13 quai Jules Guesde, 94400 Vitry-sur-Seine, France
| | - May Cindhuchao
- Molecular Screening Technology, Sanofi Biologics Research, 270 Albany Street, Cambridge, MA 02139, USA
| | - Dietmar Hoffmann
- Molecular Screening Technology, Sanofi Biologics Research, 270 Albany Street, Cambridge, MA 02139, USA
| | - Sabine Gratzer
- Molecular Discovery, Immuno-Oncology Therapeutic Research Area, Sanofi Strasbourg R&D Center, 16 rue d'Ankara, 67000 Strasbourg, France
| |
Collapse
|
40
|
Aromatic-Turmerone Analogs Protect Dopaminergic Neurons in Midbrain Slice Cultures through Their Neuroprotective Activities. Cells 2021; 10:cells10051090. [PMID: 34063571 PMCID: PMC8147616 DOI: 10.3390/cells10051090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 12/19/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra. The inflammatory activation of microglia participates in dopaminergic neurodegeneration in PD. Therefore, chemicals that inhibit microglial activation are considered to have therapeutic potential for PD. Aromatic (ar)-turmerone is a main component of turmeric oil extracted from Curcuma longa and has anti-inflammatory activity in cultured microglia. The aims of the present study are (1) to investigate whether naturally occurring S-enantiomer of ar-turmerone (S-Tur) protects dopaminergic neurons in midbrain slice cultures and (2) to examine ar-turmerone analogs that have higher activities than S-Tur in inhibiting microglial activation and protecting dopaminergic neurons. R-enantiomer (R-Tur) and two analogs showed slightly higher anti-inflammatory effects in microglial BV2 cells. S- and R-Tur and these two analogs reversed dopaminergic neurodegeneration triggered by microglial activation in midbrain slice cultures. Unexpectedly, this neuroprotection was independent of the inhibition of microglial activation. Additionally, two analogs more potently inhibited dopaminergic neurodegeneration triggered by a neurotoxin, 1-methyl-4-phenylpyridinium, than S-Tur. Taken together, we identified two ar-turmerone analogs that directly and potently protected dopaminergic neurons. An investigation using dopaminergic neuronal precursor cells suggested the possible involvement of nuclear factor erythroid 2-related factor 2 in this neuroprotection.
Collapse
|
41
|
Neuhof A, Tian Y, Reska A, Falkenburger BH, Gründer S. Large Acid-Evoked Currents, Mediated by ASIC1a, Accompany Differentiation in Human Dopaminergic Neurons. Front Cell Neurosci 2021; 15:668008. [PMID: 33986647 PMCID: PMC8110905 DOI: 10.3389/fncel.2021.668008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
Acid-sensing ion channels (ASICs) are proton-gated Na+ channels. They contribute to synaptic transmission, neuronal differentiation and neurodegeneration. ASICs have been mainly characterized in neurons from mice or rats and our knowledge of their properties in human neurons is scarce. Here, we functionally characterized ASICs in differentiating LUHMES cells, a human mesencephalic cell line with characteristics of dopaminergic neurons. We find that LUHMES cells express functional ASICs, predominantly homomeric ASIC1a. Expression starts early during differentiation with a striking surge in current amplitude at days 4-6 of differentiation, a time point where-based on published data-LUHMES cells start expressing synaptic markers. Peak ASIC expression therefore coincides with a critical period of LUHMES cell differentiation. It was associated with increased excitability, but not paralleled by an increase in ASIC1 mRNA or protein. In differentiating as well as in terminally differentiated LUHMES cells, ASIC activation by slight acidification elicited large currents, action potentials and a rise in cytosolic Ca2+. Applying the ASIC pore blocker diminazene during differentiation reduced the length of neurites, consistent with the hypothesis that ASICs play a critical role in LUHMES cell differentiation. In summary, our study establishes LUHMES cells as a valuable model to study the role of ASICs for neuronal differentiation and potentially also cell death in a human cell line.
Collapse
Affiliation(s)
- Andreas Neuhof
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany.,Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Yuemin Tian
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Anna Reska
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | | | - Stefan Gründer
- Department of Neurology, Institute of Physiology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
42
|
Pavia-Collado R, Cóppola-Segovia V, Miquel-Rio L, Alarcón-Aris D, Rodríguez-Aller R, Torres-López M, Paz V, Ruiz-Bronchal E, Campa L, Artigas F, Montefeltro A, Revilla R, Bortolozzi A. Intracerebral Administration of a Ligand-ASO Conjugate Selectively Reduces α-Synuclein Accumulation in Monoamine Neurons of Double Mutant Human A30P*A53T*α-Synuclein Transgenic Mice. Int J Mol Sci 2021; 22:ijms22062939. [PMID: 33805843 PMCID: PMC8001805 DOI: 10.3390/ijms22062939] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022] Open
Abstract
α-Synuclein (α-Syn) protein is involved in the pathogenesis of Parkinson's disease (PD). Point mutations and multiplications of the α-Syn, which encodes the SNCA gene, are correlated with early-onset PD, therefore the reduction in a-Syn synthesis could be a potential therapy for PD if delivered to the key affected neurons. Several experimental strategies for PD have been developed in recent years using oligonucleotide therapeutics. However, some of them have failed or even caused neuronal toxicity. One limiting step in the success of oligonucleotide-based therapeutics is their delivery to the brain compartment, and once there, to selected neuronal populations. Previously, we developed an indatraline-conjugated antisense oligonucleotide (IND-1233-ASO), that selectively reduces α-Syn synthesis in midbrain monoamine neurons of mice, and nonhuman primates. Here, we extended these observations using a transgenic male mouse strain carrying both A30P and A53T mutant human α-Syn (A30P*A53T*α-Syn). We found that A30P*A53T*α-Syn mice at 4-5 months of age showed 3.5-fold increases in human α-Syn expression in dopamine (DA) and norepinephrine (NE) neurons of the substantia nigra pars compacta (SNc) and locus coeruleus (LC), respectively, compared with mouse α-Syn levels. In parallel, transgenic mice exhibited altered nigrostriatal DA neurotransmission, motor alterations, and an anxiety-like phenotype. Intracerebroventricular IND-1233-ASO administration (100 µg/day, 28 days) prevented the α-Syn synthesis and accumulation in the SNc and LC, and recovered DA neurotransmission, although it did not reverse the behavioral phenotype. Therefore, the present therapeutic strategy based on a conjugated ASO could be used for the selective inhibition of α-Syn expression in PD-vulnerable monoamine neurons, showing the benefit of the optimization of ASO molecules as a disease modifying therapy for PD and related α-synucleinopathies.
Collapse
Affiliation(s)
- Rubén Pavia-Collado
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Valentín Cóppola-Segovia
- Laboratory of Neurobiology and Redox Pathology, Department of Basic Pathology, Federal University of Paraná (UFPR), Curitiba 81531-980, Brazil;
| | - Lluís Miquel-Rio
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Diana Alarcón-Aris
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Raquel Rodríguez-Aller
- CHU de Quebec Research Center, Axe Neurosciences. Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 4G2, Canada;
- CERVO Brain Research Centre, Quebec City, QC G1J 2G3, Canada; (A.M.); (R.R.)
| | - María Torres-López
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Verónica Paz
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Esther Ruiz-Bronchal
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Leticia Campa
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Francesc Artigas
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
| | - Andrés Montefeltro
- CERVO Brain Research Centre, Quebec City, QC G1J 2G3, Canada; (A.M.); (R.R.)
- n-Life Therapeutics, S.L., 18100 Granada, Spain
| | - Raquel Revilla
- CERVO Brain Research Centre, Quebec City, QC G1J 2G3, Canada; (A.M.); (R.R.)
- n-Life Therapeutics, S.L., 18100 Granada, Spain
| | - Analia Bortolozzi
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (R.P.-C.); (L.M.-R.); (D.A.-A.); (M.T.-L.); (V.P.); (E.R.-B.); (L.C.); (F.A.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
43
|
Ke M, Chong CM, Zhu Q, Zhang K, Cai CZ, Lu JH, Qin D, Su H. Comprehensive Perspectives on Experimental Models for Parkinson's Disease. Aging Dis 2021; 12:223-246. [PMID: 33532138 PMCID: PMC7801282 DOI: 10.14336/ad.2020.0331] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/31/2020] [Indexed: 11/19/2022] Open
Abstract
Parkinson’s disease (PD) ranks second among the most common neurodegenerative diseases, characterized by progressive and selective loss of dopaminergic neurons. Various cross-species preclinical models, including cellular models and animal models, have been established through the decades to study the etiology and mechanism of the disease from cell lines to nonhuman primates. These models are aimed at developing effective therapeutic strategies for the disease. None of the current models can replicate all major pathological and clinical phenotypes of PD. Selection of the model for PD largely relies on our interest of study. In this review, we systemically summarized experimental PD models, including cellular and animal models used in preclinical studies, to understand the pathogenesis of PD. This review is intended to provide current knowledge about the application of these different PD models, with focus on their strengths and limitations with respect to their contributions to the assessment of the molecular pathobiology of PD and identification of the therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Minjing Ke
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cheong-Meng Chong
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qi Zhu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ke Zhang
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cui-Zan Cai
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jia-Hong Lu
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dajiang Qin
- 2Guangzhou Regenerative Medicine and Health Guangdong Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,3South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Huanxing Su
- 1State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
44
|
Valadez-Barba V, Cota-Coronado A, Hernández-Pérez O, Lugo-Fabres PH, Padilla-Camberos E, Díaz NF, Díaz-Martínez NE. iPSC for modeling neurodegenerative disorders. Regen Ther 2021; 15:332-339. [PMID: 33426236 PMCID: PMC7770414 DOI: 10.1016/j.reth.2020.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative disorders such as Parkinson's and Alzheimer's disease, are fundamental health concerns all around the world. The development of novel treatments and new techniques to address these disorders, are being actively studied by researchers and medical personnel. In the present review we will discuss the application of induced Pluripotent Stem Cells (iPSCs) for cell-therapy replacement and disease modelling. The aim of iPSCs is to restore the functionality of the damaged tissue by replacing the impaired cells with competitive ones. To achieve this objective, iPSCs can be properly differentiated into virtually any cell fate and can be strongly translated into human health via in vitro and in vivo disease modeling for the development of new therapies, the discovery of biomarkers for several disorders, the elaboration and testing of new drugs as novel treatments, and as a tool for personalized medicine. Novel treatments to address neurodegenerative disorders. Induced pluripotent stem cell therapy and disease modelling. Parkinson's & Alzheimer's disease research.
Collapse
Key Words
- AD, Alzheimer's disease
- AFP, Alpha-Fetoprotein
- Alzheimer
- Aβ, β-Amyloid
- B-III-TUB, β–III–Tubulin
- BBB, Blood Brain Barrier
- CRISPR, Clustered Regularly Interspaced Short Palindromic Repeats
- DOPAL, 3,4-Dihydroxyphenylacetaldehyde
- EBs, Embryoid Bodies
- FLASH, Fast Length Adjustment of Short Reads
- LUHMES, Lund Human Mesencephalic Cell Line
- MHC, Mayor Histocompatibility Complex
- Neurodegenerative diseasaes
- PCR, Polymerase Chain Reaction
- PD, Parkinson's Disease
- Parkinson
- ROS, Reactive Oxygen Species
- SCs, Stem Cells
- SMA, Smooth-Muscle Antibody
- SNPc, Substantia Nigra Pars Compacta
- TH, Tyrosine Hydroxylase
- WGS, Whole Genome Sequencing
- gRNA, guide RNA
- hESC, Human Embryonic Stem Cells
- iPSCs
- iPSCs, Induced Pluripotent Stem Cells
- nsSNVs, nonsynonymous single nucleotide variants
- pTau, Phosphorylated Tau
Collapse
Affiliation(s)
- Valeria Valadez-Barba
- Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. Av. Normalistas 800, Colinas de las Normal, Jalisco, Mexico, P.C.44270
| | - A. Cota-Coronado
- Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. Av. Normalistas 800, Colinas de las Normal, Jalisco, Mexico, P.C.44270
- The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - O.R. Hernández-Pérez
- Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. Av. Normalistas 800, Colinas de las Normal, Jalisco, Mexico, P.C.44270
| | - Pavel H. Lugo-Fabres
- Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. Av. Normalistas 800, Colinas de las Normal, Jalisco, Mexico, P.C.44270
| | - Eduardo Padilla-Camberos
- Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. Av. Normalistas 800, Colinas de las Normal, Jalisco, Mexico, P.C.44270
| | - Néstor Fabián Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - N. Emmanuel Díaz-Martínez
- Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. Av. Normalistas 800, Colinas de las Normal, Jalisco, Mexico, P.C.44270
- Corresponding author. Department of Medical and Pharmaceutical Biotechnology, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. Jalisco, Mexico.
| |
Collapse
|
45
|
Leah T, Vazquez-Villaseñor I, Ferraiuolo L, Wharton SB, Mortiboys H. A Parkinson's Disease-relevant Mitochondrial and Neuronal Morphology High-throughput Screening Assay in LUHMES Cells. Bio Protoc 2021; 11:e3881. [PMID: 33732769 DOI: 10.21769/bioprotoc.3881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 11/02/2022] Open
Abstract
Parkinson's disease is a devastating neurodegenerative disorder affecting 2-3% of the population over 65 years of age. There is currently no disease-modifying treatment. One of the predominant pathological features of Parkinson's disease is mitochondrial dysfunction, and much work has aimed to identify therapeutic compounds which can restore the disrupted mitochondrial physiology. However, modelling mitochondrial dysfunction in a disease-relevant model, suitable for screening large compound libraries for ameliorative effects, represents a considerable challenge. Primary patient derived cells, SHSY-5Y cells and in vivo models of Parkinson's disease have been utilized extensively to study the contribution of mitochondrial dysfunction in Parkinson's. Indeed many studies have utilized LUHMES cells to study Parkinson's disease, however LUHMES cells have not been used as a compound screening model for PD-associated mitochondrial dysfunction previously, despite possessing several advantages compared to other frequently used models, such as rapid differentiation and high uniformity (e.g., in contrast to iPSC-derived neurons), and relevant physiology as human mesencephalic tissue capable of differentiating into dopaminergic-like neurons that highly express characteristic markers. After previously generating GFP+-LUHMES cells to model metabolic dysfunction, we report this protocol using GFP+-LUHMES cells for high-throughput compound screening in a restoration model of PD-associated mitochondrial dysfunction. This protocol describes the use of a robust and reproducible toxin-induced GFP+-LUHMES cell model for high throughput compound screening by assessing a range of mitochondrial and neuronal morphological parameters. We also provide detailed instructions for data and statistical analysis, including example calculations of Z'-score to assess statistical effect size across independent experiments.
Collapse
Affiliation(s)
- Tom Leah
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
46
|
Shah FJ, Caviglia C, Zór K, Carminati M, Ferrari G, Sampietro M, Martínez-Serrano A, Emnéus JK, Heiskanen AR. Impedance-based Real-time Monitoring of Neural Stem Cell Differentiation. JOURNAL OF ELECTRICAL BIOIMPEDANCE 2021; 12:34-49. [PMID: 34966467 PMCID: PMC8667812 DOI: 10.2478/joeb-2021-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Indexed: 06/14/2023]
Abstract
We present here the first impedance-based characterization of the differentiation process of two human mesencephalic fetal neural stem lines. The two dopaminergic neural stem cell lines used in this study, Lund human mesencephalic (LUHMES) and human ventral mesencephalic (hVM1 Bcl-XL), have been developed for the study of Parkinsonian pathogenesis and its treatment using cell replacement therapy. We show that if only relying on impedance magnitude analysis, which is by far the most usual approach in, e.g., cytotoxicity evaluation and drug screening applications, one may not be able to distinguish whether the neural stem cells in a population are proliferating or differentiating. However, the presented results highlight that equivalent circuit analysis can provide detailed information on cellular behavior, e.g. simultaneous changes in cell morphology, cell-cell contacts, and cell adhesion during formation of neural projections, which are the fundamental behavioral differences between proliferating and differentiating neural stem cells. Moreover, our work also demonstrates the sensitivity of impedance-based monitoring with capability to provide information on changes in cellular behavior in relation to proliferation and differentiation. For both of the studied cell lines, in already two days (one day after induction of differentiation) equivalent circuit analysis was able to show distinction between proliferation and differentiation conditions, which is significantly earlier than by microscopic imaging. This study demonstrates the potential of impedance-based monitoring as a technique of choice in the study of stem cell behavior, laying the foundation for screening assays to characterize stem cell lines and testing the efficacy epigenetic control.
Collapse
Affiliation(s)
- F. J. Shah
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Particle Analytical ApS, Agern Allé 3, 2970 Hørsholm, Denmark
| | - C. Caviglia
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Radiometer Medical ApS, Åkandevej 21, 2700 Brønshøj, Denmark
| | - K. Zór
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics, Department of Health Technology, Technical University of Denmark, Ørsteds Plads, Building 345C, 2800Kongens Lyngby, Denmark
| | - M. Carminati
- Dipartimento di Elettronica, Informazione e Bioingegneria - DEIB, Politecnico di Milano, P.za L. da Vinci 32, 20133Milano, Italy
| | - G. Ferrari
- Dipartimento di Elettronica, Informazione e Bioingegneria - DEIB, Politecnico di Milano, P.za L. da Vinci 32, 20133Milano, Italy
| | - M. Sampietro
- Dipartimento di Elettronica, Informazione e Bioingegneria - DEIB, Politecnico di Milano, P.za L. da Vinci 32, 20133Milano, Italy
| | - A. Martínez-Serrano
- Department of Molecular Neuropathology, Center of Molecular Biology Severo Ochoa, Universidad Autónoma de Madrid, Calle Nicolás Cabrera 1, Cantoblanco, 28049Madrid, Spain
| | - J. K. Emnéus
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Present affiliation: Department of Biotechnology and Biomedicine, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
| | - A. R. Heiskanen
- Department of Micro- and Nanotechnology, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
- Present affiliation: Department of Biotechnology and Biomedicine, Technical University of Denmark, Produktionstorvet, Building 423, 2800Kongens Lyngby, Denmark
| |
Collapse
|
47
|
Wen S, Aki T, Unuma K, Uemura K. Chemically Induced Models of Parkinson's Disease: History and Perspectives for the Involvement of Ferroptosis. Front Cell Neurosci 2020; 14:581191. [PMID: 33424553 PMCID: PMC7786020 DOI: 10.3389/fncel.2020.581191] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Ferroptosis is a newly discovered form of necrotic cell death characterized by its dependency on iron and lipid peroxidation. Ferroptosis has attracted much attention recently in the area of neurodegeneration since the involvement of ferroptosis in Parkinson’s disease (PD), a major neurodegenerative disease, has been indicated using animal models. Although PD is associated with both genetic and environmental factors, sporadic forms of PD account for more than 90% of total PD. Following the importance of environmental factors, various neurotoxins are used as chemical inducers of PD both in vivo and in vitro. In contrast to other neurodegenerative diseases such as Alzheimer’s and Huntington’s diseases (AD and HD), many of the characteristics of PD can be reproduced in vivo by the use of specific neurotoxins. Given the indication of ferroptosis in PD pathology, several studies have been conducted to examine whether ferroptosis plays role in the loss of dopaminergic neurons in PD. However, there are still few reports showing an authentic form of ferroptosis in neuronal cells during exposure to the neurotoxins used as PD inducers. In this review article, we summarize the history of the uses of chemicals to create PD models in vivo and in vitro. Besides, we also survey recent reports examining the possible involvement of ferroptosis in chemical models of PD.
Collapse
Affiliation(s)
- Shuheng Wen
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
48
|
Lauter G, Coschiera A, Yoshihara M, Sugiaman-Trapman D, Ezer S, Sethurathinam S, Katayama S, Kere J, Swoboda P. Differentiation of ciliated human midbrain-derived LUHMES neurons. J Cell Sci 2020; 133:jcs249789. [PMID: 33115758 DOI: 10.1242/jcs.249789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Many human cell types are ciliated, including neural progenitors and differentiated neurons. Ciliopathies are characterized by defective cilia and comprise various disease states, including brain phenotypes, where the underlying biological pathways are largely unknown. Our understanding of neuronal cilia is rudimentary, and an easy-to-maintain, ciliated human neuronal cell model is absent. The Lund human mesencephalic (LUHMES) cell line is a ciliated neuronal cell line derived from human fetal mesencephalon. LUHMES cells can easily be maintained and differentiated into mature, functional neurons within one week. They have a single primary cilium as proliferating progenitor cells and as postmitotic, differentiating neurons. These developmental stages are completely separable within one day of culture condition change. The sonic hedgehog (SHH) signaling pathway is active in differentiating LUHMES neurons. RNA-sequencing timecourse analyses reveal molecular pathways and gene-regulatory networks critical for ciliogenesis and axon outgrowth at the interface between progenitor cell proliferation, polarization and neuronal differentiation. Gene expression dynamics of cultured LUHMES neurons faithfully mimic the corresponding in vivo dynamics of human fetal midbrain. In LUHMES cells, neuronal cilia biology can be investigated from proliferation through differentiation to mature neurons.
Collapse
Affiliation(s)
- Gilbert Lauter
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | - Andrea Coschiera
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | - Masahito Yoshihara
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | | | - Sini Ezer
- University of Helsinki, Research Program of Molecular Neurology and Folkhälsan Institute of Genetics, FI-00290 Helsinki, Finland
| | - Shalini Sethurathinam
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| | - Shintaro Katayama
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
- University of Helsinki, Stem Cells and Metabolism Research Program and Folkhälsan Research Center, FI-00290 Helsinki, Finland
| | - Juha Kere
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
- University of Helsinki, Research Program of Molecular Neurology and Folkhälsan Institute of Genetics, FI-00290 Helsinki, Finland
| | - Peter Swoboda
- Karolinska Institute, Department of Biosciences and Nutrition, SE-141 83 Huddinge, Sweden
| |
Collapse
|
49
|
Amitriptyline interferes with autophagy-mediated clearance of protein aggregates via inhibiting autophagosome maturation in neuronal cells. Cell Death Dis 2020; 11:874. [PMID: 33070168 PMCID: PMC7568721 DOI: 10.1038/s41419-020-03085-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/13/2022]
Abstract
Amitriptyline is a tricyclic antidepressant commonly prescribed for major depressive disorders, as well as depressive symptoms associated with various neurological disorders. A possible correlation between the use of tricyclic antidepressants and the occurrence of Parkinson's disease has been reported, but its underlying mechanism remains unknown. The accumulation of misfolded protein aggregates has been suggested to cause cellular toxicity and has been implicated in the common pathogenesis of neurodegenerative diseases. Here, we examined the effect of amitriptyline on protein clearance and its relevant mechanisms in neuronal cells. Amitriptyline exacerbated the accumulation of abnormal aggregates in both in vitro neuronal cells and in vivo mice brain by interfering with the (1) formation of aggresome-like aggregates and (2) autophagy-mediated clearance of aggregates. Amitriptyline upregulated LC3B-II, but LC3B-II levels did not increase further in the presence of NH4Cl, which suggests that amitriptyline inhibited autophagic flux rather than autophagy induction. Amitriptyline interfered with the fusion of autophagosome and lysosome through the activation of PI3K/Akt/mTOR pathway and Beclin 1 acetylation, and regulated lysosome positioning by increasing the interaction between proteins Arl8, SKIP, and kinesin. To the best of our knowledge, we are the first to demonstrate that amitriptyline interferes with autophagic flux by regulating the autophagosome maturation during autophagy in neuronal cells. The present study could provide neurobiological clue for the possible correlation between the amitriptyline use and the risk of developing neurodegenerative diseases.
Collapse
|
50
|
Oxidative Stress in Parkinson's Disease: Potential Benefits of Antioxidant Supplementation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2360872. [PMID: 33101584 PMCID: PMC7576349 DOI: 10.1155/2020/2360872] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) occurs in approximately 1% of the population over 65 years of age and has become increasingly more common with advances in age. The number of individuals older than 60 years has been increasing in modern societies, as well as life expectancy in developing countries; therefore, PD may pose an impact on the economic, social, and health structures of these countries. Oxidative stress is highlighted as an important factor in the genesis of PD, involving several enzymes and signaling molecules in the underlying mechanisms of the disease. This review presents updated data on the involvement of oxidative stress in the disease, as well as the use of antioxidant supplements in its therapy.
Collapse
|