1
|
Kishore A, Sturm M, Soman Pillai K, Hakkaart C, Kalikavil Puthanveedu D, Urulangodi M, Krishnan S, Ashok Kumar Sreelatha A, Rajan R, Pal PK, Yadav R, Sarma G, Casadei N, Gasser T, Bauer P, Riess O, Sharma M. Resequencing the complete SNCA locus in Indian patients with Parkinson's disease. NPJ Parkinsons Dis 2024; 10:85. [PMID: 38622158 PMCID: PMC11018851 DOI: 10.1038/s41531-024-00676-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024] Open
Abstract
The genetic loci implicated in familial Parkinson's disease (PD) have limited generalizability to the Indian PD population. We tested mutations and the frequency of known mutations in the SNCA gene in a PD cohort from India. We selected 298 PD cases and 301 age-matched controls for targeted resequencing (before QC), along with 363 PD genomes of Indian ancestry and 1029 publicly available whole genomes from India as healthy controls (IndiGenomes), to determine the frequency of monogenic SNCA mutations. The raw sequence reads were analyzed using an in-house analysis pipeline, allowing the detection of small variants and structural variants using Manta. The in-depth analysis of the SNCA locus did not identify missense or structural variants, including previously identified SNCA mutations, in the Indian population. The familial forms of SNCA gene variants do not play a major role in the Indian PD population and this warrants further research in the under-represented population.
Collapse
Affiliation(s)
- Asha Kishore
- Comprehensive Care Centre for Movement Disorders, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kochi, Kerala, India
- Parkinson and Movement Disorder Centre, Centre for Excellence in Neurosciences, Aster Medcity, Kochi, Kerala, India
| | - Marc Sturm
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Kanchana Soman Pillai
- Parkinson and Movement Disorder Centre, Centre for Excellence in Neurosciences, Aster Medcity, Kochi, Kerala, India
| | - Christopher Hakkaart
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Divya Kalikavil Puthanveedu
- Comprehensive Care Centre for Movement Disorders, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kochi, Kerala, India
| | - Madhusoodanan Urulangodi
- Comprehensive Care Centre for Movement Disorders, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kochi, Kerala, India
| | - Syam Krishnan
- Comprehensive Care Centre for Movement Disorders, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kochi, Kerala, India
| | - Ashwin Ashok Kumar Sreelatha
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany
| | - Roopa Rajan
- Department of Neurology, All India Institute for Medical Sciences, New Delhi, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Gangadhara Sarma
- Comprehensive Care Centre for Movement Disorders, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kochi, Kerala, India
| | - Nicolas Casadei
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Thomas Gasser
- Department for Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Peter Bauer
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centogene GmbH, Rostock, Germany
- University Medicine Rostock, Internal Medicine III, Hematology, Rostock, Germany
| | - Olaf Riess
- Institute for Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Manu Sharma
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
2
|
Sastre D, Zafar F, Torres CAM, Piper D, Kirik D, Sanders LH, Qi LS, Schüle B. Inactive S. aureus Cas9 downregulates alpha-synuclein and reduces mtDNA damage and oxidative stress levels in human stem cell model of Parkinson's disease. Sci Rep 2023; 13:17796. [PMID: 37853101 PMCID: PMC10584834 DOI: 10.1038/s41598-023-45078-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases, but no disease modifying therapies have been successful in clinical translation presenting a major unmet medical need. A promising target is alpha-synuclein or its aggregated form, which accumulates in the brain of PD patients as Lewy bodies. While it is not entirely clear which alpha-synuclein protein species is disease relevant, mere overexpression of alpha-synuclein in hereditary forms leads to neurodegeneration. To specifically address gene regulation of alpha-synuclein, we developed a CRISPR interference (CRISPRi) system based on the nuclease dead S. aureus Cas9 (SadCas9) fused with the transcriptional repressor domain Krueppel-associated box to controllably repress alpha-synuclein expression at the transcriptional level. We screened single guide (sg)RNAs across the SNCA promoter and identified several sgRNAs that mediate downregulation of alpha-synuclein at varying levels. CRISPRi downregulation of alpha-synuclein in iPSC-derived neuronal cultures from a patient with an SNCA genomic triplication showed functional recovery by reduction of oxidative stress and mitochondrial DNA damage. Our results are proof-of-concept in vitro for precision medicine by targeting the SNCA gene promoter. The SNCA CRISPRi approach presents a new model to understand safe levels of alpha-synuclein downregulation and a novel therapeutic strategy for PD and related alpha-synucleinopathies.
Collapse
Affiliation(s)
- Danuta Sastre
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr., R271/217, Stanford, CA, 94305, USA
| | - Faria Zafar
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr., R271/217, Stanford, CA, 94305, USA
| | - C Alejandra Morato Torres
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr., R271/217, Stanford, CA, 94305, USA
| | - Desiree Piper
- Department of Biological Sciences, San Jose State University, San Jose, CA, 95192, USA
| | - Deniz Kirik
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Laurie H Sanders
- Departments of Neurology and Pathology, Duke Center for Neurodegeneration and Neurotherapeutics, Duke University Medical Center, Durham, NC, 27710, USA
| | - L Stanley Qi
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Birgitt Schüle
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Dr., R271/217, Stanford, CA, 94305, USA.
- Biosciences Division, SRI International, Menlo Park, CA, 94025, USA.
| |
Collapse
|
3
|
Sastre D, Zafar F, Torres CAM, Piper D, Kirik D, Sanders LH, Qi S, Schüle B. Nuclease-dead S. aureus Cas9 downregulates alpha-synuclein and reduces mtDNA damage and oxidative stress levels in patient-derived stem cell model of Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.525105. [PMID: 36747875 PMCID: PMC9900844 DOI: 10.1101/2023.01.24.525105] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases, but no disease modifying therapies have been successful in clinical translation presenting a major unmet medical need. A promising target is alpha-synuclein or its aggregated form, which accumulates in the brain of PD patients as Lewy bodies. While it is not entirely clear which alpha-synuclein protein species is disease relevant, mere overexpression of alpha-synuclein in hereditary forms leads to neurodegeneration. To specifically address gene regulation of alpha-synuclein, we developed a CRISPR interference (CRISPRi) system based on the nuclease dead S. aureus Cas9 (SadCas9) fused with the transcriptional repressor domain Krueppel-associated box to controllably repress alpha-synuclein expression at the transcriptional level. We screened single guide (sg)RNAs across the SNCA promoter and identified several sgRNAs that mediate downregulation of alpha-synuclein at varying levels. CRISPRi downregulation of alpha-synuclein in iPSC-derived neuronal cultures from a patient with an SNCA genomic triplication showed functional recovery by reduction of oxidative stress and mitochondrial DNA damage. Our results are proof-of-concept in vitro for precision medicine by targeting the SNCA gene promoter. The SNCA CRISPRi approach presents a new model to understand safe levels of alpha-synuclein downregulation and a novel therapeutic strategy for PD and related alpha-synucleinopathies.
Collapse
Affiliation(s)
- Danuta Sastre
- Stanford University School of Medicine, Department of Pathology, Stanford, CA 94305, U.S.A
| | - Faria Zafar
- Stanford University School of Medicine, Department of Pathology, Stanford, CA 94305, U.S.A
| | | | - Desiree Piper
- San Jose State University, Department of Biological Sciences, San Jose, 95192 CA, U.S.A
| | - Deniz Kirik
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Laurie H. Sanders
- Departments of Neurology and Pathology, Duke Center for Neurodegeneration and Neurotherapeutics, Duke University Medical Center, Durham, NC 27710, U.S.A
| | - Stanley Qi
- Stanford University, Department of Bioengineering, Stanford, CA 94305, U.S.A
| | - Birgitt Schüle
- Stanford University School of Medicine, Department of Pathology, Stanford, CA 94305, U.S.A
| |
Collapse
|
4
|
Yang X, Wang S. Down-Regulation of p38 Mitogen-Activated Protein Kinases/Nuclear Factor Kappa Light Chain Enhancer of Activated B Cells (p38 MAPK/NF- κB) Signaling Pathway Promotes Bone Marrow Mesenchymal Stem Cells Differentiation into Neural Stem Cells in Healing Neurodegeneration. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study intends to promote bone marrow mesenchymal stem cells (BMSCs) differentiation into neural stem cells by down-regulating p38 MAPK/NF-κB to heal neurodegeneration. 26 patients with neurodegenerative diseases were enrolled from the Department of Neurology along
with recruitment of 26 other healthy controls followed by analysis of p38 MAPK/NF-κB signaling pathway expression by ELISA. BMSCs were cultured and characterized by flow cytometry. Western blot and qRTPCR measured the p38 MAPK/NF-κB expression in the absence or presence
of p38 MAPK/NF-κB inhibitors. p38 MAPK/NF-κB expression in 26 neurodegenerative patients was significantly higher than that of 26 healthy controls. The qRT-PCR and western blot results showed that the neural stem cell-specific proteins expression was increased as
days went; after addition of p38 MAPK/NF-κB inhibitor, the expression of related specific genes were significantly decreased. In conclusion, inhibition of the expression of p38 MAPK/NF-κB signaling pathway can heal neurodegeneration by promoting the differentiation
of BMSCs into neural stem cells.
Collapse
Affiliation(s)
- Xin Yang
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China
| | - Shandan Wang
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China
| |
Collapse
|
5
|
Guo Y, Sun Y, Song Z, Zheng W, Xiong W, Yang Y, Yuan L, Deng H. Genetic Analysis and Literature Review of SNCA Variants in Parkinson's Disease. Front Aging Neurosci 2021; 13:648151. [PMID: 34456707 PMCID: PMC8397385 DOI: 10.3389/fnagi.2021.648151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/29/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is the fastest-growing neurodegenerative disorder. Aging, environmental factors, and genetics are considered as risk factors. The alpha-synuclein gene (SNCA), the first pathogenic gene identified in a familial form of PD, was indisputably involved as a heritable component for familial and sporadic PD. In this study, whole-exome sequencing and Sanger sequencing were performed to evaluate the association between the SNCA gene variants and PD. The genetic data of 438 clinically diagnosed patients with PD and 543 matched control populations of the Han Chinese were analyzed. The literature review of SNCA variants for 231 cases reported in 89 articles was extracted from the PubMed and the Movement Disorder Society Genetic mutation database. No potentially causative variant(s) in the SNCA gene, excepting two single-nucleotide nonsynonymous variants c.158C>T (p.A53V, rs542171324) and c.349C>T (p.P117S, rs145138372), were detected. There was no statistically significant difference in the genotypic or allelic frequencies for either variant between the PD group and the control group (all P > 0.05). No copy number variants of the SNCA gene were detected. The results of this study suggest that the variants in the exons of the SNCA gene may have less or no role in the development of PD in the Han Chinese populations. The literature review suggests that psychiatric signs and cognitive decline/dementia were more common among patients with SNCA duplication or triplication (psychiatric signs: χ2 = 7.892, P = 0.005; cognitive decline/dementia: χ2 = 8.991, P = 0.003).
Collapse
Affiliation(s)
- Yi Guo
- Department of Medical Information, School of Life Sciences, Central South University, Changsha, China
| | - Yan Sun
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Song
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wen Zheng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiong
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yan Yang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lamei Yuan
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| | - Hao Deng
- Center for Experimental Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China.,Disease Genome Research Center, Central South University, Changsha, China
| |
Collapse
|
6
|
Genetics of synucleins in neurodegenerative diseases. Acta Neuropathol 2021; 141:471-490. [PMID: 32740728 DOI: 10.1007/s00401-020-02202-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022]
Abstract
The SNCA locus currently has an indisputable role in Parkinson's disease and other synucleinopathies. The role of genetic variability in the other members of the synuclein family (SNCB and SNCG) in disease is far less clear. In this review, we critically assess the pathogenicity, main characteristics, and roles of genetic variants in these genes reported to be causative of synucleinopathies. We also summarize the different association signals identified in the SNCA locus that have been associated with risk for disease. We take a bird's eye view of the variability currently reported in the general population for the three genes and use these data to infer on the potential relationship between each of the genes and human disease.
Collapse
|
7
|
Abdelwahab SA, Elsebay SAG, Ibrahim MFG, Abdel Hafez SMN. Cerebral and cerebellar histological changes in the rat animal model of rotenone induced parkinsonism can be ameliorated by bone marrow derived stem cell conditioned media. J Chem Neuroanat 2020; 111:101892. [PMID: 33220428 DOI: 10.1016/j.jchemneu.2020.101892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 11/29/2022]
Abstract
Parkinson disease is the second most common neurodegenerative disease affecting elderly patients. It occurs due to the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc). We continue our work in this model focusing on other brain areas affected with this disorder; cerebral cortex and cerebellum (areas other than substantia nigra) for better understanding the motor and behavior effect of the Parkinson disease as a forward steep for its treatment and medical control. This work aims to evaluate the therapeutic effect of stem cell-conditioned medium in the Parkinsonism model. In this study, Parkinsonism model was induced in rats by daily subcutaneous injection of 0.5 mg/Kg of rotenone for 28 days. Thirty rats were divided randomly into 3 groups; control, Parkinson, and conditioned medium (CM) treated groups. Cerebral Cortex and Cerebellum were obtained for histological, immunohistochemical and biochemical studies. In the Parkinsonism group, marked histological changes were observed. These findings were nearly ameliorated in CM treated group as confirmed by the biochemical, histological, and immunohistochemical (anti-alpha synculein, anti GFAP and anti nestin) studies. It could be concluded that CM had a good therapeutic effect on Parkinsonism induced damage in both the cerebral cortex and cerebellum.
Collapse
|
8
|
The Role of Alpha-Synuclein and Other Parkinson's Genes in Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2020; 21:ijms21165724. [PMID: 32785033 PMCID: PMC7460874 DOI: 10.3390/ijms21165724] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 07/29/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodevelopmental and late-onset neurodegenerative disorders present as separate entities that are clinically and neuropathologically quite distinct. However, recent evidence has highlighted surprising commonalities and converging features at the clinical, genomic, and molecular level between these two disease spectra. This is particularly striking in the context of autism spectrum disorder (ASD) and Parkinson's disease (PD). Genetic causes and risk factors play a central role in disease pathophysiology and enable the identification of overlapping mechanisms and pathways. Here, we focus on clinico-genetic studies of causal variants and overlapping clinical and cellular features of ASD and PD. Several genes and genomic regions were selected for our review, including SNCA (alpha-synuclein), PARK2 (parkin RBR E3 ubiquitin protein ligase), chromosome 22q11 deletion/DiGeorge region, and FMR1 (fragile X mental retardation 1) repeat expansion, which influence the development of both ASD and PD, with converging features related to synaptic function and neurogenesis. Both PD and ASD display alterations and impairments at the synaptic level, representing early and key disease phenotypes, which support the hypothesis of converging mechanisms between the two types of diseases. Therefore, understanding the underlying molecular mechanisms might inform on common targets and therapeutic approaches. We propose to re-conceptualize how we understand these disorders and provide a new angle into disease targets and mechanisms linking neurodevelopmental disorders and neurodegeneration.
Collapse
|
9
|
Abdelwahab S, Elsebay SAG, Fouli Gaber M, Abdel-Hafez SMN. Comparative study between bone marrow mesenchymal stem cell and their conditioned medium in the treatment of rat model of Parkinsonism. J Cell Physiol 2020; 236:440-457. [PMID: 32557610 DOI: 10.1002/jcp.29872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/30/2020] [Indexed: 12/17/2022]
Abstract
Parkinsonism is one of the most common aging neurodegenerative disorders. This study aims to compare the therapeutic effect of stem cell versus its conditioned medium in the Parkinsonism model. Parkinsonism was induced by daily subcutaneous injection of 0.5 mg/kg of rotenone dissolved in dimethyl sulfoxide for 28 days. Fifty rats were divided randomly into five groups: control, dimethyl sulfoxide, Parkinsonism, stem cell-treated, and conditioned medium-treated groups. Midbrain specimens were obtained for histological, immunohistochemical, and biochemical studies. Lewy bodies were observed in the Parkinsonism group in the dopaminergic neuron and neuropil as well. Almost all of the pathological changes were clearly ameliorated in both stem cell- and conditioned medium-treated groups as confirmed by biochemical, histological, and immunohistochemical (anti-nestin, anti-glial fibrillary acidic protein, and anti-α synuclein) studies. However, the conditioned medium showed more superior therapeutic effect establishing nearly the normal histological architecture of substantia nigra. These results may pave the future for using stem cell-conditioned medium as a more convenient and effective adjuvant therapy in Parkinsonism and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Soha Abdelwahab
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Sara Abdel Gawad Elsebay
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Manar Fouli Gaber
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Minia, Egypt
| | | |
Collapse
|
10
|
Piras IS, Bleul C, Schrauwen I, Talboom J, Llaci L, De Both MD, Naymik MA, Halliday G, Bettencourt C, Holton JL, Serrano GE, Sue LI, Beach TG, Stefanova N, Huentelman MJ. Transcriptional profiling of multiple system atrophy cerebellar tissue highlights differences between the parkinsonian and cerebellar sub-types of the disease. Acta Neuropathol Commun 2020; 8:76. [PMID: 32493431 PMCID: PMC7268362 DOI: 10.1186/s40478-020-00950-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/19/2020] [Indexed: 01/04/2023] Open
Abstract
Multiple system atrophy (MSA) is a rare adult-onset neurodegenerative disease of unknown cause, with no effective therapeutic options, and no cure. Limited work to date has attempted to characterize the transcriptional changes associated with the disease, which presents as either predominating parkinsonian (MSA-P) or cerebellar (MSC-C) symptoms. We report here the results of RNA expression profiling of cerebellar white matter (CWM) tissue from two independent cohorts of MSA patients (n = 66) and healthy controls (HC; n = 66). RNA samples from bulk brain tissue and from oligodendrocytes obtained by laser capture microdissection (LCM) were sequenced. Differentially expressed genes (DEGs) were obtained and were examined before and after stratifying by MSA clinical sub-type.We detected the highest number of DEGs in the MSA-C group (n = 747) while only one gene was noted in MSA-P, highlighting the larger dysregulation of the transcriptome in the MSA-C CWM. Results from both bulk tissue and LCM analysis showed a downregulation of oligodendrocyte genes and an enrichment for myelination processes with a key role noted for the QKI gene. Additionally, we observed a significant upregulation of neuron-specific gene expression in MSA-C and enrichment for synaptic processes. A third cluster of genes was associated with the upregulation of astrocyte and endothelial genes, two cell types with a key role in inflammation processes. Finally, network analysis in MSA-C showed enrichment for β-amyloid related functional classes, including the known Alzheimer's disease (AD) genes, APP and PSEN1.This is the largest RNA profiling study ever conducted on post-mortem brain tissue from MSA patients. We were able to define specific gene expression signatures for MSA-C highlighting the different stages of the complex neurodegenerative cascade of the disease that included alterations in several cell-specific transcriptional programs. Finally, several results suggest a common transcriptional dysregulation between MSA and AD-related genes despite the clinical and neuropathological distinctions between the two diseases.
Collapse
Affiliation(s)
- Ignazio S Piras
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Christiane Bleul
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Isabelle Schrauwen
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
- Present Address: Department of Neurology, Center for Statistical Genetics, Gertrude H. Sergievsky Center, Columbia University Medical Center, 630 W 168th St, New York, NY, 10032, USA
| | - Joshua Talboom
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Lorida Llaci
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
- Present address: Division of Biology and Biomedical Sciences, Molecular Genetics and Genomics Program, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Matthew D De Both
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Marcus A Naymik
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Glenda Halliday
- The University of Sydney Brain and Mind Centre and Faculty of Medicine and Health, School of Medical Science, and Neuroscience Research Australia, Sydney, Australia
| | - Conceicao Bettencourt
- Queen Square Brain Bank for Neurological Disorders and Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Janice L Holton
- Queen Square Brain Bank for Neurological Disorders and Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Geidy E Serrano
- Civin Laboratory of Neuropathology at Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Lucia I Sue
- Civin Laboratory of Neuropathology at Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Thomas G Beach
- Civin Laboratory of Neuropathology at Banner Sun Health Research Institute, Sun City, AZ, 85351, USA
| | - Nadia Stefanova
- Department of Neurology, Division of Neurobiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Matthew J Huentelman
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ, USA.
| |
Collapse
|
11
|
Safari F, Hatam G, Behbahani AB, Rezaei V, Barekati-Mowahed M, Petramfar P, Khademi F. CRISPR System: A High-throughput Toolbox for Research and Treatment of Parkinson's Disease. Cell Mol Neurobiol 2020; 40:477-493. [PMID: 31773362 DOI: 10.1007/s10571-019-00761-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/14/2019] [Indexed: 12/13/2022]
Abstract
In recent years, the innovation of gene-editing tools such as the CRISPR/Cas9 system improves the translational gap of treatments mediated by gene therapy. The privileges of CRISPR/Cas9 such as working in living cells and organs candidate this technology for using in research and treatment of the central nervous system (CNS) disorders. Parkinson's disease (PD) is a common, debilitating, neurodegenerative disorder which occurs due to loss of dopaminergic neurons and is associated with progressive motor dysfunction. Knowledge about the pathophysiological basis of PD has altered the classification system of PD, which manifests in familial and sporadic forms. The first genetic linkage studies in PD demonstrated the involvement of Synuclein alpha (SNCA) mutations and SNCA genomic duplications in the pathogenesis of PD familial forms. Subsequent studies have also insinuated mutations in leucine repeat kinase-2 (LRRK2), Parkin, PTEN-induced putative kinase 1 (PINK1), as well as DJ-1 causing familial forms of PD. This review will attempt to discuss the structure, function, and development in genome editing mediated by CRISP/Cas9 system. Further, it describes the genes involved in the pathogenesis of PD and the pertinent alterations to them. We will pursue this line by delineating the PD linkage studies in which CRISPR system was employed. Finally, we will discuss the pros and cons of CRISPR employment vis-à-vis the process of genome editing in PD patients' iPSCs.
Collapse
Affiliation(s)
- Fatemeh Safari
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, 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
| | - Gholamreza Hatam
- Basic Sciences in Infectious Diseases Research Center, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Behzad Behbahani
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Rezaei
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mazyar Barekati-Mowahed
- Department of Physiology & Biophysics, School of Medicine, Case Western Reserve University, Ohio, USA
| | - Peyman Petramfar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Khademi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
12
|
Delenclos M, Burgess JD, Lamprokostopoulou A, Outeiro TF, Vekrellis K, McLean PJ. Cellular models of alpha-synuclein toxicity and aggregation. J Neurochem 2019; 150:566-576. [PMID: 31265132 DOI: 10.1111/jnc.14806] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/19/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
Misfolding and aggregation of alpha-synuclein (α-synuclein) with concomitant cytotoxicity is a hallmark of Lewy body related disorders such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Although it plays a pivotal role in pathogenesis and disease progression, the function of α-synuclein and the molecular mechanisms underlying α-synuclein-induced neurotoxicity in these diseases are still elusive. Many in vitro and in vivo experimental models mimicking α-synuclein pathology such as oligomerization, toxicity and more recently neuronal propagation have been generated over the years. In particular, cellular models have been crucial for our comprehension of the pathogenic process of the disease and are beneficial for screening of molecules capable of modulating α-synuclein toxicity. Here, we review α-synuclein based cell culture models that reproduce some features of the neuronal populations affected in patients, from basic unicellular organisms to mammalian cell lines and primary neurons, to the cutting edge models of patient-specific cell lines. These reprogrammed cells known as induced pluripotent stem cells (iPSCs) have garnered attention because they closely reproduce the characteristics of neurons found in patients and provide a valuable tool for mechanistic studies. We also discuss how different cell models may constitute powerful tools for high-throughput screening of molecules capable of modulating α-synuclein toxicity and prevention of its propagation. This article is part of the Special Issue "Synuclein".
Collapse
Affiliation(s)
- Marion Delenclos
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jeremy D Burgess
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA.,Neuroscience PhD Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, Florida, USA
| | - Agaristi Lamprokostopoulou
- Department of Neuroscience, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Kostas Vekrellis
- Department of Neuroscience, Center for Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA.,Neuroscience PhD Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, Florida, USA
| |
Collapse
|
13
|
Jellinger KA. Animal models of synucleinopathies and how they could impact future drug discovery and delivery efforts. Expert Opin Drug Discov 2019; 14:969-982. [DOI: 10.1080/17460441.2019.1638908] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Riederer P, Berg D, Casadei N, Cheng F, Classen J, Dresel C, Jost W, Krüger R, Müller T, Reichmann H, Rieß O, Storch A, Strobel S, van Eimeren T, Völker HU, Winkler J, Winklhofer KF, Wüllner U, Zunke F, Monoranu CM. α-Synuclein in Parkinson's disease: causal or bystander? J Neural Transm (Vienna) 2019; 126:815-840. [PMID: 31240402 DOI: 10.1007/s00702-019-02025-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) comprises a spectrum of disorders with differing subtypes, the vast majority of which share Lewy bodies (LB) as a characteristic pathological hallmark. The process(es) underlying LB generation and its causal trigger molecules are not yet fully understood. α-Synuclein (α-syn) is a major component of LB and SNCA gene missense mutations or duplications/triplications are causal for rare hereditary forms of PD. As typical sporadic PD is associated with LB pathology, a factor of major importance is the study of the α-syn protein and its pathology. α-Syn pathology is, however, also evident in multiple system atrophy (MSA) and Lewy body disease (LBD), making it non-specific for PD. In addition, there is an overlap of these α-synucleinopathies with other protein-misfolding diseases. It has been proven that α-syn, phosphorylated tau protein (pτ), amyloid beta (Aβ) and other proteins show synergistic effects in the underlying pathogenic mechanisms. Multiple cell death mechanisms can induce pathological protein-cascades, but this can also be a reverse process. This holds true for the early phases of the disease process and especially for the progression of PD. In conclusion, while rare SNCA gene mutations are causal for a minority of familial PD patients, in sporadic PD (where common SNCA polymorphisms are the most consistent genetic risk factor across populations worldwide, accounting for 95% of PD patients) α-syn pathology is an important feature. Conversely, with regard to the etiopathogenesis of α-synucleinopathies PD, MSA and LBD, α-syn is rather a bystander contributing to multiple neurodegenerative processes, which overlap in their composition and individual strength. Therapeutic developments aiming to impact on α-syn pathology should take this fact into consideration.
Collapse
Affiliation(s)
- Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, University of Würzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany. .,Department of Psychiatry, University of South Denmark, Odense, Denmark.
| | - Daniela Berg
- Department of Neurology, UKHS, Christian-Albrechts-Universität, Campus Kiel, Kiel, Germany
| | - Nicolas Casadei
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Fubo Cheng
- NGS Competence Center Tübingen, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Joseph Classen
- Department of Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Christian Dresel
- Department of Neurology, Center for Movement Disorders, Neuroimaging Center Mainz, Clinical Neurophysiology, Forschungszentrum Translationale Neurowissenschaften (FTN), Rhein-Main-Neuronetz, Mainz, Germany
| | | | - Rejko Krüger
- Clinical and Experimental Neuroscience, LCSB (Luxembourg Centre for Systems, Biomedicine), University of Luxembourg, Esch-sur-Alzette and Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg.,National Center for Excellence in Research, Parkinson's disease (NCER-PD), Parkinson Research Clinic, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Thomas Müller
- Department of Neurology, Alexianer St. Joseph Berlin-Weißensee, Berlin, Germany
| | - Heinz Reichmann
- Department of Neurology, University of Dresden, Dresden, Germany
| | - Olaf Rieß
- Institute of Medical Genetics and Applied Genomics, Tübingen, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany.,German Centre for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Sabrina Strobel
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Thilo van Eimeren
- Department of Neurology, University Hospital of Cologne, Cologne, Germany
| | | | - Jürgen Winkler
- Department Kopfkliniken, Molekulare Neurologie, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Konstanze F Winklhofer
- Institute of Biochemistry and Pathobiochemistry, Ruhr-Universität Bochum, Bochum, Germany
| | - Ullrich Wüllner
- Department of Neurology, University of Bonn, German Center for Neurodegenerative Diseases (DZNE Bonn), Bonn, Germany
| | - Friederike Zunke
- Department of Biochemistry, Medical Faculty, University of Kiel, Kiel, Germany
| | - Camelia-Maria Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
15
|
CCAAT/enhancer binding protein δ is a transcriptional repressor of α-synuclein. Cell Death Differ 2019; 27:509-524. [PMID: 31209363 DOI: 10.1038/s41418-019-0368-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/30/2019] [Accepted: 05/02/2019] [Indexed: 12/16/2022] Open
Abstract
α-Synuclein is the main component of Lewy bodies, the intracellular protein aggregates representing the histological hallmark of Parkinson's disease. Elevated α-synuclein levels and mutations in SNCA gene are associated with increased risk for Parkinson's disease. Despite this, little is known about the molecular mechanisms regulating SNCA transcription. CCAAT/enhancer binding protein (C/EBP) β and δ are b-zip transcription factors that play distinct roles in neurons and glial cells. C/EBPβ overexpression increases SNCA expression in neuroblastoma cells and putative C/EBPβ and δ binding sites are present in the SNCA genomic region suggesting that these proteins could regulate SNCA transcription. Based on these premises, the goal of this study was to determine if C/EBPβ and δ regulate the expression of SNCA. We first observed that α-synuclein CNS expression was not affected by C/EBPβ deficiency but it was markedly increased in C/EBPδ-deficient mice. This prompted us to characterize further the role of C/EBPδ in SNCA transcription. C/EBPδ absence led to the in vivo increase of α-synuclein in all brain regions analyzed, both at mRNA and protein level, and in primary neuronal cultures. In agreement with this, CEBPD overexpression in neuroblastoma cells and in primary neuronal cultures markedly reduced SNCA expression. ChIP experiments demonstrated C/EBPδ binding to the SNCA genomic region of mice and humans and luciferase experiments showed decreased expression of a reporter gene attributable to C/EBPδ binding to the SNCA promoter. Finally, decreased CEBPD expression was observed in the substantia nigra and in iPSC-derived dopaminergic neurons from Parkinson patients resulting in a significant negative correlation between SNCA and CEBPD levels. This study points to C/EBPδ as an important repressor of SNCA transcription and suggests that reduced C/EBPδ neuronal levels could be a pathogenic factor in Parkinson's disease and other synucleinopathies and C/EBPδ activity a potential pharmacological target for these neurological disorders.
Collapse
|
16
|
Genetic fine-mapping of the Iowan SNCA gene triplication in a patient with Parkinson's disease. NPJ PARKINSONS DISEASE 2018; 4:18. [PMID: 29928688 PMCID: PMC6003950 DOI: 10.1038/s41531-018-0054-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 04/26/2018] [Accepted: 05/10/2018] [Indexed: 02/07/2023]
Abstract
The “Iowa kindred,” a large Iowan family with autosomal-dominant Parkinson’s disease, has been followed clinically since the 1920s at the Mayo Clinic. In 2003, the genetic cause was determined to be a 1.7 Mb triplication of the alpha-synuclein genomic locus. Affected individuals present with an early-onset, severe parkinsonism-dementia syndrome. Here, we present a descendant of the Iowa kindred with novel, disease-associated non-motor findings of reduced heart rate variability, complete anosmia, and a rare skin condition called colloid milium. At autopsy, key neuropathological findings were compatible with diffuse Lewy body disease. Using high-resolution comparative genomic hybridization (CGH) array analysis to fine-map the genomic breakpoints, we observed two independent recombination events of the SNCA locus that resulted in a genomic triplication of twelve genes, including SNCA, and the disruption of two genes, HERC6 and CCSER1, at the genomic breakpoints. In conclusion, we provide further evidence that the mere two-fold overexpression of alpha-synuclein leads to a fulminant alpha-synucleinopathy with rapid progression and severe clinical and neuropathological features.
Collapse
|