1
|
Brzozowski CF, Challa H, Gcwensa NZ, Hall D, Nabert D, Chambers N, Gallardo I, Millet M, Volpicelli-Daley L, Moehle MS. Early α-synuclein aggregation decreases corticostriatal glutamate drive and synapse density. Neurobiol Dis 2025; 210:106918. [PMID: 40250719 DOI: 10.1016/j.nbd.2025.106918] [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: 01/29/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025] Open
Abstract
Neuronal inclusions of α-synuclein (α-syn) are pathological hallmarks of Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB). α-Syn pathology accumulates in cortical neurons which project to the striatum. To understand how α-syn pathology affects cortico-striatal synapses at early time points before significant dopamine neuron loss, pre-formed α-syn fibrils (PFF) were injected into the striatum to induce endogenous α-syn aggregation in corticostriatal-projecting neurons. Electrophysiological recordings of striatal spiny projection neurons (SPNs) from acute slices found a significant decrease in evoked corticostriatal glutamate release and corticostriatal synaptic release sites in mice with PFF-induced aggregates compared to monomer injected mice. Expansion microscopy, confocal microscopy and Imaris reconstructions were used to identify VGLUT1 positive presynaptic terminals juxtaposed to Homer1 positive postsynaptic densities, termed synaptic loci. Quantitation of synaptic loci density revealed an early loss of corticostriatal synapses. Immunoblots of the striatum showed reductions in expression of pre-synaptic proteins VGLUT1, VAMP2 and Snap25, in mice with α-syn aggregates compared to controls. Paradoxically, a small percentage of remaining VGLUT1+ synaptic loci positive for pS129-α-syn aggregates showed enlarged volumes compared to nearby synapses without α-syn aggregates. Our combined physiology and high-resolution imaging data point to an early loss of corticostriatal synapses in mice harboring α-synuclein inclusions, which may contribute to impaired basal ganglia circuitry in PD and DLB.
Collapse
Affiliation(s)
- Charlotte F Brzozowski
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Harshita Challa
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nolwazi Z Gcwensa
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dominic Hall
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Douglas Nabert
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Nicole Chambers
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Ignacio Gallardo
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Michael Millet
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA
| | - Laura Volpicelli-Daley
- Department of Neurology, Killon Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mark S Moehle
- Department of Pharmacology and Therapeutics, Center for Translational Research in Neurodegeneration, and Fixel Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
2
|
Mingo YB, Escobar Galvis ML, Henderson MX. α-Synuclein pathology and mitochondrial dysfunction: Toxic partners in Parkinson's disease. Neurobiol Dis 2025; 209:106889. [PMID: 40157617 DOI: 10.1016/j.nbd.2025.106889] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025] Open
Abstract
Two major neuropathological features of Parkinson's disease (PD) are α-synuclein Lewy pathology and mitochondrial dysfunction. Although both α-synuclein pathology and mitochondrial dysfunction may independently contribute to PD pathogenesis, the interaction between these two factors is not yet fully understood. In this review, we discuss the physiological functions of α-synuclein and mitochondrial homeostasis in neurons as well as the pathological defects that ensue when these functions are disturbed in PD. Recent studies have highlighted that dysfunctional mitochondria can become sequestered within Lewy bodies, and cell biology studies have suggested that α-synuclein can directly impair mitochondrial function. There are also PD cases caused by genetic or environmental perturbation of mitochondrial homeostasis. Together, these studies suggest that mitochondrial dysfunction may be a common pathway to neurodegeneration in PD, triggered by multiple insults. We review the literature surrounding the interaction between α-synuclein and mitochondria and highlight open questions in the field that may be explored to advance our understanding of PD and develop novel, disease-modifying therapies.
Collapse
Affiliation(s)
- Yakum B Mingo
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America
| | | | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America.
| |
Collapse
|
3
|
Taddei RN, Duff KE. Synapse vulnerability and resilience across the clinical spectrum of dementias. Nat Rev Neurol 2025:10.1038/s41582-025-01094-7. [PMID: 40404832 DOI: 10.1038/s41582-025-01094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2025] [Indexed: 05/24/2025]
Abstract
Preservation of synapses is crucial for healthy cognitive ageing, and synapse loss is one of the closest anatomical correlates of cognitive decline in Alzheimer disease, dementia with Lewy bodies and frontotemporal dementia. In these conditions, some synapses seem particularly vulnerable to degeneration whereas others are resilient and remain preserved. Evidence has highlighted that vulnerability and resilience are intrinsically distinct phenomena linked to specific brain structural and/or functional signatures, yet the key features of vulnerable and resilient synapses in the dementias remain incompletely understood. Defining the characteristics of vulnerable and resilient synapses in each form of dementia could offer novel insight into the mechanisms of synapse preservation and of synapse loss that underlies cognitive decline, thereby facilitating the discovery of targeted biomarkers and disease-modifying therapies. In this Review, we consider the concepts of synapse vulnerability and resilience, and provide an overview of our current understanding of the associations between synaptic protein changes, neuropathology and cognitive decline. We also consider how understanding of the underlying mechanisms could identify novel strategies to mitigate the cognitive dysfunction associated with dementias.
Collapse
Affiliation(s)
- Raquel N Taddei
- Neurology Department, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- UK Dementia Research Institute at UCL, Institute of Neurology, University College London, London, UK.
| | - Karen E Duff
- UK Dementia Research Institute at UCL, Institute of Neurology, University College London, London, UK
| |
Collapse
|
4
|
Fan X, Li H. Integration of Single-Cell and Spatial Transcriptomic Data Reveals Spatial Architecture and Potential Biomarkers in Alzheimer's Disease. Mol Neurobiol 2025; 62:5395-5412. [PMID: 39543008 DOI: 10.1007/s12035-024-04617-3] [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: 05/21/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by the gradual loss of neurons and the accumulation of amyloid plaques and neurofibrillary tangles. Despite advancements in the understanding of AD's pathophysiology, the cellular organization and interactions in the prefrontal cortex (PFC) remain elusive. Eight single-cell RNA sequencing (scRNA-seq) datasets from both normal controls and individuals with AD were harmonized. Stringent preprocessing protocols were implemented to uphold dataset integrity. Unsupervised clustering and annotation revealed 22 distinct cell clusters corresponding to 19 unique cell types. The spatial architecture of the PFC region was constructed using the CARD tool. Further analyses encompassed trajectory examination of Oligodendrocyte subtypes, evaluation of regulon activity scores, and spot clustering within white matter regions (WM). Differential expression analysis and functional enrichment assays unveiled molecular signatures linked to AD progression and were validated using microarray data sourced from neurodegenerative disorder patients. Our investigation employs scRNA-seq and spatial transcriptomics to uncover the cellular atlas and spatial architecture of the human PFC in AD. Moreover, our results indicate that Oligodendrocytes are more prevalent in AD patients, showcasing diverse subtypes and spatial organization within WM regions. Each subtype appears to be associated with distinct biological processes and transcriptional regulators, shedding light on their involvement in AD pathology. Notably, the Oligodendrocyte_C6 subtype is linked to neurological damage in AD patients, characterized by heightened expression of genes involved in cell-cell connections, cell membrane stability, and myelination. Additionally, 12 target genes regulated by NFIA were identified, which are upregulated in AD patients and associated with disease progression. Elevated PLXDC2 expression in peripheral blood was also identified, suggesting its potential as a non-invasive biomarker for early AD detection. Our study provides novel insights into the role of Oligodendrocytes in AD and highlights the potential of PLXDC2 as a blood biomarker for non-invasive diagnosis and monitoring of AD patients.
Collapse
Affiliation(s)
- Xing Fan
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong, 226001, PR, China
| | - Huamei Li
- Department of Rheumatology and Immunology, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, 210008, PR, China.
| |
Collapse
|
5
|
Wang Q, Zhu H, Deng L, Xu S, Xie W, Li M, Wang R, Tie L, Zhan L, Yu G. Spatial Transcriptomics: Biotechnologies, Computational Tools, and Neuroscience Applications. SMALL METHODS 2025; 9:e2401107. [PMID: 39760243 DOI: 10.1002/smtd.202401107] [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: 07/19/2024] [Revised: 12/22/2024] [Indexed: 01/07/2025]
Abstract
Spatial transcriptomics (ST) represents a revolutionary approach in molecular biology, providing unprecedented insights into the spatial organization of gene expression within tissues. This review aims to elucidate advancements in ST technologies, their computational tools, and their pivotal applications in neuroscience. It is begun with a historical overview, tracing the evolution from early image-based techniques to contemporary sequence-based methods. Subsequently, the computational methods essential for ST data analysis, including preprocessing, cell type annotation, spatial clustering, detection of spatially variable genes, cell-cell interaction analysis, and 3D multi-slices integration are discussed. The central focus of this review is the application of ST in neuroscience, where it has significantly contributed to understanding the brain's complexity. Through ST, researchers advance brain atlas projects, gain insights into brain development, and explore neuroimmune dysfunctions, particularly in brain tumors. Additionally, ST enhances understanding of neuronal vulnerability in neurodegenerative diseases like Alzheimer's and neuropsychiatric disorders such as schizophrenia. In conclusion, while ST has already profoundly impacted neuroscience, challenges remain issues such as enhancing sequencing technologies and developing robust computational tools. This review underscores the transformative potential of ST in neuroscience, paving the way for new therapeutic insights and advancements in brain research.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hongyuan Zhu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lin Deng
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuangbin Xu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenqin Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ming Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rui Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Liang Tie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li Zhan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
6
|
Lee Y, Lee M, Shin Y, Kim K, Kim T. Spatial Omics in Clinical Research: A Comprehensive Review of Technologies and Guidelines for Applications. Int J Mol Sci 2025; 26:3949. [PMID: 40362187 PMCID: PMC12071594 DOI: 10.3390/ijms26093949] [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: 03/02/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Spatial omics integrates molecular profiling with spatial tissue context, enabling high-resolution analysis of gene expression, protein interactions, and epigenetic modifications. This approach provides critical insights into disease mechanisms and therapeutic responses, with applications in cancer, neurology, and immunology. Spatial omics technologies, including spatial transcriptomics, proteomics, and epigenomics, facilitate the study of cellular heterogeneity, tissue organization, and cell-cell interactions within their native environments. Despite challenges in data complexity and integration, advancements in multi-omics pipelines and computational tools are enhancing data accuracy and biological interpretation. This review provides a comprehensive overview of key spatial omics technologies, their analytical methods, validation strategies, and clinical applications. By integrating spatially resolved molecular data with traditional omics, spatial omics is transforming precision medicine, biomarker discovery, and personalized therapy. Future research should focus on improving standardization, reproducibility, and multimodal data integration to fully realize the potential of spatial omics in clinical and translational research.
Collapse
Affiliation(s)
- Yoonji Lee
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (Y.L.); (M.L.); (Y.S.)
| | - Mingyu Lee
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (Y.L.); (M.L.); (Y.S.)
| | - Yoojin Shin
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (Y.L.); (M.L.); (Y.S.)
| | - Kyuri Kim
- College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul 07804, Republic of Korea;
| | - Taejung Kim
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Republic of Korea
| |
Collapse
|
7
|
Li Y, Liu X, Guo L, Han K, Fang S, Wan X, Wang D, Xu X, Jiang L, Fan G, Xu M. SpaGRN: Investigating spatially informed regulatory paths for spatially resolved transcriptomics data. Cell Syst 2025; 16:101243. [PMID: 40179878 DOI: 10.1016/j.cels.2025.101243] [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: 11/27/2023] [Revised: 08/30/2024] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Cells spatially organize into distinct cell types or functional domains through localized gene regulatory networks. However, current spatially resolved transcriptomics analyses fail to integrate spatial constraints and proximal cell influences, limiting the mechanistic understanding of tissue organization. Here, we introduce SpaGRN, a statistical framework that reconstructs cell-type- or functional-domain-specific, dynamic, and spatial regulons by coupling intracellular spatial regulatory causality with extracellular signaling path information. Benchmarking across synthetic and real datasets demonstrates SpaGRN's superior precision over state-of-the-art tools in identifying context-dependent regulons. Applied to diverse spatially resolved transcriptomics platforms (Stereo-seq, STARmap, MERFISH, CosMx, Slide-seq, and 10x Visium), complex cancerous samples, and 3D datasets of developing Drosophila embryos and larvae, SpaGRN not only provides a versatile toolkit for decoding receptor-mediated spatial regulons but also reveals spatiotemporal regulatory mechanisms underlying organogenesis and inflammation.
Collapse
Affiliation(s)
- Yao Li
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China
| | | | - Lidong Guo
- BGI Research, Qingdao 266555, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Han
- BGI Research, Qingdao 266555, China
| | - Shuangsang Fang
- BGI Research, Beijing 102601, China; BGI Research, Shenzhen 518083, China
| | - Xinjiang Wan
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, and Center of Deep Sea Research Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | | | - Xun Xu
- BGI Research, Wuhan 430074, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Ling Jiang
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Guangyi Fan
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| | - Mengyang Xu
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| |
Collapse
|
8
|
Antico O, Thompson PW, Hertz NT, Muqit MMK, Parton LE. Targeting mitophagy in neurodegenerative diseases. Nat Rev Drug Discov 2025; 24:276-299. [PMID: 39809929 DOI: 10.1038/s41573-024-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Mitochondrial dysfunction is a hallmark of idiopathic neurodegenerative diseases, including Parkinson disease, amyotrophic lateral sclerosis, Alzheimer disease and Huntington disease. Familial forms of Parkinson disease and amyotrophic lateral sclerosis are often characterized by mutations in genes associated with mitophagy deficits. Therefore, enhancing the mitophagy pathway may represent a novel therapeutic approach to targeting an underlying pathogenic cause of neurodegenerative diseases, with the potential to deliver neuroprotection and disease modification, which is an important unmet need. Accumulating genetic, molecular and preclinical model-based evidence now supports targeting mitophagy in neurodegenerative diseases. Despite clinical development challenges, small-molecule-based approaches for selective mitophagy enhancement - namely, USP30 inhibitors and PINK1 activators - are entering phase I clinical trials for the first time.
Collapse
Affiliation(s)
- Odetta Antico
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Paul W Thompson
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK
| | | | - Miratul M K Muqit
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura E Parton
- Mission Therapeutics Ltd, Babraham Research Campus, Cambridge, UK.
| |
Collapse
|
9
|
Pesti B, Langa X, Kumpesa N, Valdeolivas A, Sultan M, Rottenberg S, Hahn K. Mini Review: Spatial Transcriptomics to Decode the Central Nervous System. Toxicol Pathol 2025:1926233251325204. [PMID: 40119776 DOI: 10.1177/01926233251325204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Spatial transcriptomics (ST) is revolutionizing our understanding of the central nervous system (CNS) by providing spatially resolved gene expression data. This mini review explores the impact of ST on CNS research, particularly in neurodegenerative diseases like Alzheimer's, Parkinson's, multiple sclerosis, and amyotrophic lateral sclerosis. We describe two foundational ST methods: sequencing-based and imaging-based. Key studies are reviewed highlighting the power of ST data sets to map transcriptomes to disease-specific histomorphology, elucidate molecular mechanisms of regional and cellular vulnerability, integrate single-cell data with tissue mapping, and reveal receptor-ligand interactions. Despite current challenges like data interpretation and resolution limits, ST holds promise for identifying novel drug targets, evaluating their therapeutic potential, and bridging gaps between animal models and human studies to advance development of CNS-targeting compounds.
Collapse
Affiliation(s)
- Benedek Pesti
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Xavi Langa
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Nadine Kumpesa
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Alberto Valdeolivas
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Marc Sultan
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Kerstin Hahn
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
10
|
Liang M, Chu L, Yue Z. New Multiomic Studies Shed Light on Cellular Diversity and Neuronal Susceptibility in Parkinson's Disease. Mov Disord 2025; 40:431-437. [PMID: 39812497 DOI: 10.1002/mds.30097] [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: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Parkinson's disease is a complex neurodegenerative disorder characterized by degeneration of dopaminergic neurons, with patients manifesting varying motor and nonmotor symptoms. Previous studies using single-cell RNA sequencing in rodent models and humans have identified distinct heterogeneity of neurons and glial cells with differential vulnerability. Recent studies have increasingly leveraged multiomics approaches, including spatial transcriptomics, epigenomics, and proteomics, in the study of Parkinson's disease, providing new insights into pathogenic mechanisms. Continued advancements in experimental technologies and sophisticated computational tools will be essential in uncovering a network of neuronal vulnerability and prioritizing disease modifiers for novel therapeutics development. © 2025 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marianna Liang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Parkinson's Disease Neurobiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Linh Chu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Parkinson's Disease Neurobiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics & Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zhenyu Yue
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Parkinson's Disease Neurobiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
11
|
Dutta S, Pang M, Coughlin GM, Gudavalli S, Roukes ML, Chou TF, Gradinaru V. Molecularly-guided spatial proteomics captures single-cell identity and heterogeneity of the nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637505. [PMID: 39990460 PMCID: PMC11844393 DOI: 10.1101/2025.02.10.637505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Single-cell proteomics is an emerging field with significant potential to characterize heterogeneity within biological tissues. It offers complementary insights to single-cell transcriptomics by revealing unbiased proteomic changes downstream of the transcriptome. Recent advancements have focused on enhancing proteome coverage and depth, mostly in cultured cell lines, and a few recent studies have explored the potential of analyzing tissue micro-samples but were limited to homogenous peripheral tissues. In this current work, we utilize the power of spatial single cell-proteomics through immunostaining-guided laser capture microdissection (LCM) coupled with LC-MS to investigate the heterogenous central nervous system. We used this method to compare neuronal populations from cortex and substantia nigra, two brain regions associated with motor and cognitive function and various neurological disorders. Moreover, we used the technique to understand the neuroimmune changes associated with stab wound injury. Finally, we focus our application on the peripheral nervous system, where we compare the proteome of the myenteric plexus cell ganglion to the nerve bundle. This study demonstrates the utility of spatial single-cell proteomics in neuroscience research toward understanding fundamental biology and the molecular drivers of neurological conditions.
Collapse
Affiliation(s)
- Sayan Dutta
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Marion Pang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Gerard M. Coughlin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Sirisha Gudavalli
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Michael L. Roukes
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Division of Physics, Mathematics, and Astronomy, California Institute of Technology, Pasadena, CA 91125
- Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125
| | - Tsui-Fen Chou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| |
Collapse
|
12
|
Dutta S, Hensel J, Scott A, Mohallem R, Rossitto LAM, Khan HF, Johnson T, Ferreira CR, Marmolejo JF, Chen X, Jayant K, Aryal UK, Volpicelli-Daley L, Rochet JC. Synaptic phosphoproteome modifications and cortical circuit dysfunction are linked to the early-stage progression of alpha-synuclein aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634820. [PMID: 39896549 PMCID: PMC11785254 DOI: 10.1101/2025.01.24.634820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cortical dysfunction is increasingly recognized as a major contributor to the non-motor symptoms associated with Parkinson's disease (PD) and other synucleinopathies. Although functional alterations in cortical circuits have been observed in preclinical PD models, the underlying molecular mechanisms are unclear. To bridge this knowledge gap, we investigated tissue-level changes in the cortices of rats and mice treated with alpha-synuclein (aSyn) seeds using a multi-omics approach. Our study revealed significant phosphoproteomic changes, but not global proteomic or lipid profiling changes, in the rat sensorimotor cortex 3 months after intrastriatal injection with aSyn preformed fibrils (PFFs). Gene ontology analysis of the phosphoproteomic data indicated that PFF administration impacted pathways related to synaptic transmission and cytoskeletal organization. Similar phosphoproteomic perturbations were observed in the sensorimotor cortex of mice injected intrastriatally or intracortically with aSyn PFFs. Functional analyses demonstrated increased neuronal firing rates and enhanced spike-spike coherence in the sensorimotor cortices of PFF-treated mice, indicating seed-dependent cortical circuit dysfunction. Bioinformatic analysis of the altered phosphosites suggested the involvement of several kinases, including casein kinase-2 (CK2), which has been previously implicated in PD pathology. Collectively, these findings highlight the importance of phosphorylation-mediated signaling pathways in the cortical response to aSyn pathology spread in PD and related synucleinopathies, setting the stage for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Sayan Dutta
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Jennifer Hensel
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Alicia Scott
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Rodrigo Mohallem
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, 47907, USA
| | - Leigh-Ana M Rossitto
- Department of Neurosciences, School of Medicine, University of California, San Diego, 92161, USA
| | - Hammad Furqan Khan
- Weldon School of Biomedical Engineering, West Lafayette, Indiana, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Teshawn Johnson
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
| | - Christina R Ferreira
- Metabolite Profiling Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907
| | - Jackeline F. Marmolejo
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, 47907, USA
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, 47906, USA
| | - Xu Chen
- Department of Neurosciences, School of Medicine, University of California, San Diego, 92161, USA
| | - Krishna Jayant
- Weldon School of Biomedical Engineering, West Lafayette, Indiana, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Uma K. Aryal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, 47907, USA
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, 47906, USA
| | - Laura Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jean-Christophe Rochet
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
13
|
Chen L, Chehade HD, Chu HY. Motor cortical neuronal hyperexcitability associated with α-synuclein aggregation. NPJ Parkinsons Dis 2025; 11:18. [PMID: 39809792 PMCID: PMC11733020 DOI: 10.1038/s41531-024-00867-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
ΑBSTRACT: In Parkinson's disease (PD), Lewy pathology deposits in the cerebral cortex, but how the pathology disrupts cortical circuit integrity and function remains poorly understood. To begin to address this question, we injected α-synuclein (αSyn) preformed fibrils (PFFs) into the dorsolateral striatum of mice to seed αSyn pathology in the cortical cortex and induce degeneration of midbrain dopaminergic neurons. We reported that αSyn aggregates accumulate in the motor cortex in a layer- and cell-subtype-specific pattern. Specifically, αSyn aggregates-bearing intratelencephalic neurons (ITNs) showed hyperexcitability, increased input resistance, and decreased cell capacitance, which were associated with impaired HCN channel function. Morphologically, the αSyn aggregates-bearing ITNs showed shrinkage of cell bodies and loss of dendritic spines. Last, we showed that partial dopamine depletion is not sufficient to alter thalamocortical transmission to cortical pyramidal neurons. Our results provide a novel mechanistic understanding of cortical circuit dysfunction in PD.
Collapse
Affiliation(s)
- Liqiang Chen
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, USA
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington, DC, 20007, USA
| | - Hiba Douja Chehade
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, USA
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington, DC, 20007, USA
| | - Hong-Yuan Chu
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, USA.
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, 49503, USA.
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington, DC, 20007, USA.
| |
Collapse
|
14
|
Vidyadhara DJ, Bäckström D, Chakraborty R, Ruan J, Park JM, Mistry PK, Chandra SS. Synaptic vesicle endocytosis deficits underlie GBA-linked cognitive dysfunction in Parkinson's disease and Dementia with Lewy bodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.23.619548. [PMID: 39484386 PMCID: PMC11527026 DOI: 10.1101/2024.10.23.619548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
GBA is the major risk gene for Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB), two common α-synucleinopathies with cognitive deficits. We investigated the role of mutant GBA in cognitive decline by utilizing Gba (L444P) mutant, SNCA transgenic (tg), and Gba-SNCA double mutant mice. Notably, Gba mutant mice showed early cognitive deficits but lacked PD-like motor deficits or α-synuclein pathology. Conversely, SNCA tg mice displayed age-related motor deficits, without cognitive abnormalities. Gba-SNCA mice exhibited both cognitive decline and exacerbated motor deficits, accompanied by greater cortical phospho-α-synuclein pathology, especially in layer 5 neurons. Single-nucleus RNA sequencing of the cortex uncovered synaptic vesicle (SV) endocytosis defects in excitatory neurons of Gba mutant and Gba-SNCA mice, via robust downregulation of genes regulating SV cycle and synapse assembly. Immunohistochemistry and electron microscopy validated these findings. Our results indicate that Gba mutations, while exacerbating pre-existing α-synuclein aggregation and PD-like motor deficits, contribute to cognitive deficits through α-synuclein-independent mechanisms, involving dysfunction in SV endocytosis.
Collapse
Affiliation(s)
- D J Vidyadhara
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
- Discipline of Neuroscience and Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago IL, USA
- Center for Neurodegenerative Disease and Therapeutics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - David Bäckström
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
- Department of Clinical Science, Neurosciences, Umeå University, Sweden
| | - Risha Chakraborty
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
| | - Jiapeng Ruan
- Department of Internal Medicine, Yale University, CT, USA
| | - Jae-Min Park
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
- Van Andel Institute, MI, USA
| | | | - Sreeganga. S. Chandra
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University, CT, USA
| |
Collapse
|
15
|
Vidyadhara DJ, Bäckström D, Chakraborty R, Ruan J, Park JM, Mistry PK, Chandra SS. Synaptic vesicle endocytosis deficits underlie GBA-linked cognitive dysfunction in Parkinson's disease and Dementia with Lewy bodies. RESEARCH SQUARE 2024:rs.3.rs-5649173. [PMID: 39764119 PMCID: PMC11703330 DOI: 10.21203/rs.3.rs-5649173/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
GBA is the major risk gene for Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB), two common α-synucleinopathies with cognitive deficits. We investigated the role of mutant GBA in cognitive decline by utilizing Gba (L444P) mutant, SNCA transgenic (tg), and Gba-SNCA double mutant mice. Notably, Gba mutant mice showed early cognitive deficits but lacked PD-like motor deficits or α-synuclein pathology. Conversely, SNCA tg mice displayed age-related motor deficits, without cognitive abnormalities. Gba-SNCA mice exhibited both cognitive decline and exacerbated motor deficits, accompanied by greater cortical phospho-α-synuclein pathology, especially in layer 5 neurons. Single-nucleus RNA sequencing of the cortex uncovered synaptic vesicle (SV) endocytosis defects in excitatory neurons of Gba mutant and Gba-SNCA mice, via robust downregulation of genes regulating SV cycle and synapse assembly. Immunohistochemistry and electron microscopy validated these findings. Our results indicate that Gba mutations, while exacerbating pre-existing α-synuclein aggregation and PD-like motor deficits, contribute to cognitive deficits through α-synuclein-independent mechanisms, involving dysfunction in SV endocytosis.
Collapse
Affiliation(s)
- D J Vidyadhara
- Departments of Neurology, Yale University, CT, USA
- Neuroscience, Yale University, CT, USA
- Discipline of Neuroscience, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
- Center for Neurodegenerative Disease and Therapeutics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - David Bäckström
- Departments of Neurology, Yale University, CT, USA
- Neuroscience, Yale University, CT, USA
- Department of Clinical Science, Neurosciences, Umeå University, Sweden
| | - Risha Chakraborty
- Departments of Neurology, Yale University, CT, USA
- Neuroscience, Yale University, CT, USA
| | - Jiapeng Ruan
- Department of Internal Medicine, Yale University, CT, USA
| | - Jae-Min Park
- Departments of Neurology, Yale University, CT, USA
- Neuroscience, Yale University, CT, USA
- Van Andel Institute, MI, USA
| | | | - Sreeganga. S. Chandra
- Departments of Neurology, Yale University, CT, USA
- Neuroscience, Yale University, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University, CT, USA
| |
Collapse
|
16
|
Onay M, Harter PN, Weber K, Piiper A, Czabanka M, Plate KH, Freiman TM, Gessler F, Puhahn-Schmeiser B. Mossy Fiber Sprouting in Temporal Lobe Epilepsy: The Impact of Netrin-1, DCC, and Gene Expression Changes. Biomedicines 2024; 12:2869. [PMID: 39767775 PMCID: PMC11672915 DOI: 10.3390/biomedicines12122869] [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: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Temporal lobe epilepsy (TLE) is the most common form of drug-resistant epilepsy, often associated with hippocampal sclerosis (HS), which involves selective neuronal loss in the Cornu Ammonis subregion 1 CA1 and CA4 regions of the hippocampus. Granule cells show migration and mossy fiber sprouting, though the mechanisms remain unclear. Microglia play a role in neurogenesis and synaptic modulation, suggesting they may contribute to epilepsy. This study examines the role of microglia and axonal guidance molecules in neuronal reorganization in TLE. METHODS Nineteen hippocampal samples from patients with TLE undergoing epilepsy surgery were analyzed. Microglial activity (M1/M2-like microglia) and neuronal guidance molecules were assessed using microscopy and semi-automated techniques. Gene expression was evaluated using the nCounter Expression Profiling method. RESULTS Neuronal cell loss was correlated with decreased activity of the M1 microglial phenotype. In the CA2 region, neuronal preservation was linked to increased mossy fiber sprouting and microglial presence. Neuronal markers such as Deleted in Colorectal Cancer (DCC) and Synaptopodin were reduced in areas of cell death, while Netrin-1 was elevated in the granule cell layer, potentially influencing mossy fiber sprouting. The nCounter analysis revealed downregulation of genes involved in neuronal activity (e.g., NPAS4, BCL-2, GRIA1) and upregulation of IκB, indicating reduced neuroinflammation. CONCLUSIONS This study suggests reduced neuroinflammation in areas of neuronal loss, while regions with preserved neurons showed mossy fiber sprouting associated with microglia, Netrin-1, and DCC.
Collapse
Affiliation(s)
- Melis Onay
- Department of Internal Medicine I, Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
- Department of Neurosurgery, Rostock University Medical Center, Schilingallee 35, 18057 Rostock, Germany
| | - Patrick N. Harter
- Department of Neuropathology, Ludwig-Maximillians-University Munich, Feodor-Lynen-Str. 23, 81377 Munich, Germany
| | - Katherina Weber
- Department of Neuropathology (Edinger Institute), Goethe University, Heinrich-Hoffmann-Straße 7, 60528 Frankfurt am Main, Germany
| | - Albrecht Piiper
- Department of Internal Medicine I, Goethe University, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, Goethe University, Schleusenweg 16, 60528 Frankfurt am Main, Germany
| | - Karl H. Plate
- Department of Neuropathology (Edinger Institute), Goethe University, Heinrich-Hoffmann-Straße 7, 60528 Frankfurt am Main, Germany
| | - Thomas M. Freiman
- Department of Neurosurgery, Rostock University Medical Center, Schilingallee 35, 18057 Rostock, Germany
| | - Florian Gessler
- Department of Neurosurgery, Rostock University Medical Center, Schilingallee 35, 18057 Rostock, Germany
| | - Barbara Puhahn-Schmeiser
- Department of Neurosurgery, Freiburg University Medical Center, Breisacher Str. 64, 79106 Freiburg, Germany
| |
Collapse
|
17
|
Horan-Portelance L, Iba M, Acri DJ, Gibbs JR, Cookson MR. Imaging spatial transcriptomics reveals molecular patterns of vulnerability to pathology in a transgenic α-synucleinopathy model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606032. [PMID: 39372781 PMCID: PMC11451628 DOI: 10.1101/2024.07.31.606032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
In Parkinson's disease and dementia with Lewy bodies, aggregated and phosphorylated α-synuclein pathology appears in select neurons throughout cortical and subcortical regions, but little is currently known about why certain populations are selectively vulnerable. Here, using imaging spatial transcriptomics (IST) coupled with downstream immunofluorescence for α-synuclein phosphorylated at Ser129 (pSyn) in the same tissue sections, we identified neuronal subtypes in the cortex and hippocampus of transgenic human α-synuclein-overexpressing mice that preferentially developed pSyn pathology. Additionally, we investigated the transcriptional underpinnings of this vulnerability, pointing to expression of Plk2, which phosphorylates α-synuclein at Ser129, and human SNCA (hSNCA), as key to pSyn pathology development. Finally, we performed differential expression analysis, revealing gene expression changes broadly downstream of hSNCA overexpression, as well as pSyn-dependent alterations in mitochondrial and endolysosomal genes. Overall, this study yields new insights into the formation of α-synuclein pathology and its downstream effects in a synucleinopathy mouse model.
Collapse
Affiliation(s)
- Liam Horan-Portelance
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michiyo Iba
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dominic J. Acri
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - J. Raphael Gibbs
- Computational Biology Group, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Palushaj B, Lewis SJG, Abdelnour C. What is the future for dementia with Lewy bodies? J Neurol 2024; 272:43. [PMID: 39666092 DOI: 10.1007/s00415-024-12734-1] [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: 10/07/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
Dementia with Lewy bodies (DLB) is the second most common neurodegenerative dementia after Alzheimer's disease (AD), yet it remains under-recognized and frequently misdiagnosed due to heterogenous clinical presentations, the presence of co-pathology, and the lack of specific diagnostic tools. Pathologically, DLB is characterized by the accumulation of misfolded alpha-synuclein (aSyn) aggregates, known as Lewy bodies. Recent advancements have improved in vivo detection of aSyn pathology through techniques such as seed amplification assays, monoclonal antibodies, and positron emission tomography using novel small-molecule ligands. The ability to detect aSyn in vivo has sparked dialogue about using biomarkers to identify individuals with aSyn, similar to the approach influencing the field of AD. Proponents argue that biological staging could facilitate the detection of preclinical disease stages, allowing for earlier intervention and targets for disease modification, and could improve diagnostic sensitivity and accuracy in selecting patients for clinical trials. However, critics caution that this method may oversimplify the complexity of DLB and overlook its clinical heterogeneity, also highlighting practical challenges related to implementation, cost, and global access to advanced diagnostic technologies. Importantly, although significant progress has been made in detecting aSyn for diagnostic purposes, disease-modifying therapies targeting aSyn have yet to demonstrate clear efficacy in slowing disease progression. Elucidating the physiological and pathophysiological roles of aSyn remains an urgent priority in neurodegenerative research. Other experimental research priorities for DLB include developing improved cellular and animal models that reflect epigenetic and environmental factors, mapping post-translational modifications, and systematically characterizing neurons that are vulnerable and resistant to lewy pathology using a multi-omic approach. Clinically, there is an urgent need for international, prospective, longitudinal studies and for validated, disease-specific outcome measures. Addressing these priorities is essential for advancing our understanding of DLB and developing effective therapies.
Collapse
Affiliation(s)
- Bianca Palushaj
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | | | - Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
19
|
Kovacs GG, Grinberg LT, Halliday G, Alafuzoff I, Dugger BN, Murayama S, Forrest SL, Martinez‐Valbuena I, Tanaka H, Kon T, Yoshida K, Jaunmuktane Z, Spina S, Nelson PT, Gentleman S, Alegre‐Abarrategui J, Serrano GE, Paes VR, Takao M, Wakabayashi K, Uchihara T, Yoshida M, Saito Y, Kofler J, Rodriguez RD, Gelpi E, Attems J, Crary JF, Seeley WW, Duda JE, Keene CD, Woulfe J, Munoz D, Smith C, Lee EB, Neumann M, White CL, McKee AC, Thal DR, Jellinger K, Ghetti B, Mackenzie IRA, Dickson DW, Beach TG. Biomarker-Based Approach to α-Synucleinopathies: Lessons from Neuropathology. Mov Disord 2024; 39:2173-2179. [PMID: 39360851 PMCID: PMC11657033 DOI: 10.1002/mds.30028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 12/20/2024] Open
Affiliation(s)
- Gabor G. Kovacs
- Tanz Centre for Research in Neurodegenerative Diseases, University of TorontoTorontoOntarioCanada
| | - Lea T. Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco (UCSF)San FranciscoCaliforniaUSA
- Department of PathologyUniversity of São Paulo Medical SchoolSão PauloBrazil
| | - Glenda Halliday
- Brain and Mind Centre, The University of SydneySydneyAustralia
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | | | | | - Shigeo Murayama
- Brain Bank for Aging Research, Tokyo Metropolitan Institute for Geriatrics and GerontologyTokyoJapan
| | - Shelley L. Forrest
- Tanz Centre for Research in Neurodegenerative Diseases, University of TorontoTorontoOntarioCanada
| | - Ivan Martinez‐Valbuena
- Tanz Centre for Research in Neurodegenerative Diseases, University of TorontoTorontoOntarioCanada
| | - Hidetomo Tanaka
- Tanz Centre for Research in Neurodegenerative Diseases, University of TorontoTorontoOntarioCanada
| | - Tomoya Kon
- Department of NeurologyHirosaki University Graduate School of MedicineHirosakiJapan
| | - Koji Yoshida
- Tanz Centre for Research in Neurodegenerative Diseases, University of TorontoTorontoOntarioCanada
| | - Zane Jaunmuktane
- Department of Clinical and Movement NeurosciencesQueen Square Brain Bank for Neurological Disorders, Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
| | - Salvatore Spina
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco (UCSF)San FranciscoCaliforniaUSA
| | - Peter T. Nelson
- Sanders‐Brown Center on Aging, University of KentuckyLexingtonKentuckyUSA
| | - Steve Gentleman
- Imperial College London, Parkinson's UK Tissue BankLondonUnited Kingdom
| | | | | | - Vitor Ribeiro Paes
- Department of PathologyUniversity of São Paulo Medical SchoolSão PauloBrazil
| | - Masaki Takao
- Department of Clinical Laboratory and Internal MedicineNational Center of Neurology and Psychiatry (NCNP), National Center HospitalTokyoJapan
| | - Koichi Wakabayashi
- Department of NeuropathologyHirosaki University Graduate School of MedicineHirosakiJapan
| | - Toshiki Uchihara
- University of Hawaii Postgraduate Medical Education Program at Okinawa Chubu HospitalUrumaJapan
| | - Mari Yoshida
- Department of NeuropathologyInstitute for Medical Science of Aging, Aichi Medical UniversityAichiJapan
| | - Yuko Saito
- Brain Bank for Aging Research, Tokyo Metropolitan Institute for Geriatrics and GerontologyTokyoJapan
| | - Julia Kofler
- Department of PathologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Roberta Diehl Rodriguez
- Laboratorio de Ressonancia Magnetica em Neurorradiologia (LIM‐44), HCFMUSP, Faculdade de MedicinaUniversidade de São PauloSão PauloBrazil
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of NeurologyMedical University of ViennaViennaAustria
| | - Johannes Attems
- Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - John F. Crary
- Departments of Pathology, Neuroscience, and Artificial Intelligence and Human HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - William W. Seeley
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco (UCSF)San FranciscoCaliforniaUSA
| | - John E. Duda
- Michael J. Crescenz VA Medical CenterPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - C. Dirk Keene
- Department of Laboratory Medicine and PathologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - John Woulfe
- Ottawa Hospital Research Institute and University of OttawaOttawaOntarioCanada
| | | | - Colin Smith
- University of Edinburgh, Academic Department of Neuropathology, Centre for Clinical Brain SciencesEdinburghUnited Kingdom
| | - Edward B. Lee
- Department of Pathology and Laboratory MedicinePerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Manuela Neumann
- Department of NeuropathologyUniversity Hospital Tübingen and DZNE TübingenTübingenGermany
| | - Charles L. White
- Neuropathology Section, Department of PathologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Ann C. McKee
- Boston University Alzheimer's Disease Center and CTE CenterBostonMassachusettsUSA
| | - Dietmar R. Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology and Leuven Brain InstituteKU‐LeuvenLeuvenBelgium
- Department of PathologyUniversity Hospital LeuvenLeuvenBelgium
| | | | - Bernardino Ghetti
- School of Medicine, Department of Pathology and Laboratory MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Ian R. A. Mackenzie
- Department of PathologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | | | | |
Collapse
|
20
|
Moisoi N. Mitochondrial proteases modulate mitochondrial stress signalling and cellular homeostasis in health and disease. Biochimie 2024; 226:165-179. [PMID: 38906365 DOI: 10.1016/j.biochi.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/16/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Maintenance of mitochondrial homeostasis requires a plethora of coordinated quality control and adaptations' mechanisms in which mitochondrial proteases play a key role. Their activation or loss of function reverberate beyond local mitochondrial biochemical and metabolic remodelling into coordinated cellular pathways and stress responses that feedback onto the mitochondrial functionality and adaptability. Mitochondrial proteolysis modulates molecular and organellar quality control, metabolic adaptations, lipid homeostasis and regulates transcriptional stress responses. Defective mitochondrial proteolysis results in disease conditions most notably, mitochondrial diseases, neurodegeneration and cancer. Here, it will be discussed how mitochondrial proteases and mitochondria stress signalling impact cellular homeostasis and determine the cellular decision to survive or die, how these processes may impact disease etiopathology, and how modulation of proteolysis may offer novel therapeutic strategies.
Collapse
Affiliation(s)
- Nicoleta Moisoi
- Leicester School of Pharmacy, Leicester Institute for Pharmaceutical Health and Social Care Innovations, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Hawthorn Building 1.03, LE1 9BH, Leicester, UK.
| |
Collapse
|
21
|
Vatsa N, Brynildsen JK, Goralski TM, Kurgat K, Meyerdirk L, Breton L, DeWeerd D, Brasseur L, Turner L, Becker K, Gallik KL, Bassett DS, Henderson MX. Network analysis of α-synuclein pathology progression reveals p21-activated kinases as regulators of vulnerability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619411. [PMID: 39484617 PMCID: PMC11526907 DOI: 10.1101/2024.10.22.619411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
α-Synuclein misfolding and progressive accumulation drives a pathogenic process in Parkinson's disease. To understand cellular and network vulnerability to α-synuclein pathology, we developed a framework to quantify network-level vulnerability and identify new therapeutic targets at the cellular level. Full brain α-synuclein pathology was mapped in mice over 9 months. Empirical pathology data was compared to theoretical pathology estimates from a diffusion model of pathology progression along anatomical connections. Unexplained variance in the model enabled us to derive regional vulnerability that we compared to regional gene expression. We identified gene expression patterns that relate to regional vulnerability, including 12 kinases that were enriched in vulnerable regions. Among these, an inhibitor of group II PAKs demonstrated protection from neuron death and α-synuclein pathology, even after delayed compound treatment. This study provides a framework for the derivation of cellular vulnerability from network-based studies and identifies a promising therapeutic pathway for Parkinson's disease.
Collapse
Affiliation(s)
- Naman Vatsa
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Julia K. Brynildsen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas M. Goralski
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kevin Kurgat
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Lindsay Meyerdirk
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Libby Breton
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniella DeWeerd
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura Brasseur
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | | | | | - Dani S. Bassett
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Michael X. Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Lead Contact
| |
Collapse
|
22
|
Priyanka, Qamar SH, Visanji NP. Toward an animal model of Progressive Supranuclear Palsy. Front Neurosci 2024; 18:1433465. [PMID: 39420986 PMCID: PMC11484047 DOI: 10.3389/fnins.2024.1433465] [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: 05/15/2024] [Accepted: 08/19/2024] [Indexed: 10/19/2024] Open
Abstract
Progressive Supranuclear Palsy (PSP) is a rare and fatal neurodegenerative tauopathy which, with a rapid clinical progression coupled to a strong degree of clinico-pathologic correlation, has been suggested to be a "frontrunner" in translational development for neurodegenerative proteinopathies. Elegant studies in animals have contributed greatly to our understanding of disease pathogenesis in PSP. However, presently no animal model replicates the key anatomical and cytopathologic hallmarks, the spatiotemporal spread of pathology, progressive neurodegeneration, or locomotor and cognitive symptoms that characterize PSP. Current models therefore likely fail to recapitulate the key mechanisms that underly the pathological progression of PSP, impeding their translational value. Here we review what we have learned about PSP from work in animals to date, examine the gaps in modeling the disease and discuss strategies for the development of refined animal models that will improve our understanding of disease pathogenesis and provide a critical platform for the testing of novel therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Priyanka
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Syeda Hania Qamar
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Naomi P. Visanji
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Rossy Progressive Supranuclear Palsy Centre, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
23
|
Wu S, Schekman RW. Intercellular transmission of alpha-synuclein. Front Mol Neurosci 2024; 17:1470171. [PMID: 39324117 PMCID: PMC11422390 DOI: 10.3389/fnmol.2024.1470171] [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: 07/25/2024] [Accepted: 08/28/2024] [Indexed: 09/27/2024] Open
Abstract
An emerging theme in Parkinson's disease (PD) is the propagation of α-synuclein pathology as the disease progresses. Research involving the injection of preformed α-synuclein fibrils (PFFs) in animal models has recapitulated the pathological spread observed in PD patients. At the cellular and molecular levels, this intercellular spread requires the translocation of α-synuclein across various membrane barriers. Recent studies have identified subcellular organelles and protein machineries that facilitate these processes. In this review, we discuss the proposed pathways for α-synuclein intercellular transmission, including unconventional secretion, receptor-mediated uptake, endosome escape and nanotube-mediated transfer. In addition, we advocate for a rigorous examination of the evidence for the localization of α-synuclein in extracellular vesicles.
Collapse
Affiliation(s)
| | - Randy W. Schekman
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
24
|
Chen L, Chehade HD, Chu HY. Motor Cortical Neuronal Hyperexcitability Associated with α-Synuclein Aggregation. RESEARCH SQUARE 2024:rs.3.rs-4797540. [PMID: 39281856 PMCID: PMC11398582 DOI: 10.21203/rs.3.rs-4797540/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Dysfunction of the cerebral cortex is thought to underlie motor and cognitive impairments in Parkinson disease (PD). While cortical function is known to be suppressed by abnormal basal ganglia output following dopaminergic degeneration, it remains to be determined how the deposition of Lewy pathology disrupts cortical circuit integrity and function. Moreover, it is also unknown whether cortical Lewy pathology and midbrain dopaminergic degeneration interact to disrupt cortical function in late-stage. To begin to address these questions, we injected α-synuclein (αSyn) preformed fibrils (PFFs) into the dorsolateral striatum of mice to seed αSyn pathology in the cortical cortex and induce degeneration of midbrain dopaminergic neurons. Using this model system, we reported that αSyn aggregates accumulate in the motor cortex in a layer- and cell-subtype-specific pattern. Particularly, intratelencephalic neurons (ITNs) showed earlier accumulation and greater extent of αSyn aggregates relative to corticospinal neurons (CSNs). Moreover, we demonstrated that the intrinsic excitability and inputs resistance of αSyn aggregates-bearing ITNs in the secondary motor cortex (M2) are increased, along with a noticeable shrinkage of cell bodies and loss of dendritic spines. Last, neither the intrinsic excitability of CSNs nor their thalamocortical input was altered by a partial striatal dopamine depletion associated with αSyn pathology. Our results documented motor cortical neuronal hyperexcitability associated with αSyn aggregation and provided a novel mechanistic understanding of cortical circuit dysfunction in PD.
Collapse
Affiliation(s)
- Liqiang Chen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| | - Hiba Douja Chehade
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| | - Hong-Yuan Chu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| |
Collapse
|
25
|
Gcwensa NZ, Russell DL, Long KY, Brzozowski CF, Liu X, Gamble KL, Cowell RM, Volpicelli-Daley LA. Excitatory synaptic structural abnormalities produced by templated aggregation of α-syn in the basolateral amygdala. Neurobiol Dis 2024; 199:106595. [PMID: 38972360 PMCID: PMC11632701 DOI: 10.1016/j.nbd.2024.106595] [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: 05/15/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024] Open
Abstract
Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) are characterized by neuronal α-synuclein (α-syn) inclusions termed Lewy Pathology, which are abundant in the amygdala. The basolateral amygdala (BLA), in particular, receives projections from the thalamus and cortex. These projections play a role in cognition and emotional processing, behaviors which are impaired in α-synucleinopathies. To understand if and how pathologic α-syn impacts the BLA requires animal models of α-syn aggregation. Injection of α-syn pre-formed fibrils (PFFs) into the striatum induces robust α-syn aggregation in excitatory neurons in the BLA that corresponds with reduced contextual fear conditioning. At early time points after aggregate formation, cortico-amygdala excitatory transmission is abolished. The goal of this project was to determine if α-syn inclusions in the BLA induce synaptic degeneration and/or morphological changes. In this study, we used C57BL/6 J mice injected bilaterally with PFFs in the dorsal striatum to induce α-syn aggregate formation in the BLA. A method was developed using immunofluorescence and three-dimensional reconstruction to analyze excitatory cortico-amygdala and thalamo-amygdala presynaptic terminals closely juxtaposed to postsynaptic densities. The abundance and morphology of synapses were analyzed at 6- or 12-weeks post-injection of PFFs. α-Syn aggregate formation in the BLA did not cause a significant loss of synapses, but cortico-amygdala and thalamo-amygdala presynaptic terminals and postsynaptic densities with aggregates of α-syn show increased volumes, similar to previous findings in human DLB cortex, and in non-human primate models of PD. Transmission electron microscopy showed that asymmetric synapses in mice with PFF-induced α-syn aggregates have reduced synaptic vesicle intervesicular distances, similar to a recent study showing phospho-serine-129 α-syn increases synaptic vesicle clustering. Thus, pathologic α-syn causes major alterations to synaptic architecture in the BLA, potentially contributing to behavioral impairment and amygdala dysfunction observed in synucleinopathies.
Collapse
Affiliation(s)
- Nolwazi Z Gcwensa
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Dreson L Russell
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Khaliah Y Long
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Charlotte F Brzozowski
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Xinran Liu
- Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Karen L Gamble
- Department of Psychiatry and Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Rita M Cowell
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Laura A Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
26
|
Chen L, Chehade HD, Chu HY. Motor Cortical Neuronal Hyperexcitability Associated with α-Synuclein Aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604995. [PMID: 39091827 PMCID: PMC11291145 DOI: 10.1101/2024.07.24.604995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Dysfunction of the cerebral cortex is thought to underlie motor and cognitive impairments in Parkinson disease (PD). While cortical function is known to be suppressed by abnormal basal ganglia output following dopaminergic degeneration, it remains to be determined how the deposition of Lewy pathology disrupts cortical circuit integrity and function. Moreover, it is also unknown whether cortical Lewy pathology and midbrain dopaminergic degeneration interact to disrupt cortical function in late-stage. To begin to address these questions, we injected α-synuclein (αSyn) preformed fibrils (PFFs) into the dorsolateral striatum of mice to seed αSyn pathology in the cortical cortex and induce degeneration of midbrain dopaminergic neurons. Using this model system, we reported that αSyn aggregates accumulate in the motor cortex in a layer- and cell-subtype-specific pattern. Particularly, intratelencephalic neurons (ITNs) showed earlier accumulation and greater extent of αSyn aggregates relative to corticospinal neurons (CSNs). Moreover, we demonstrated that the intrinsic excitability and inputs resistance of αSyn aggregates-bearing ITNs in the secondary motor cortex (M2) are increased, along with a noticeable shrinkage of cell bodies and loss of dendritic spines. Last, neither the intrinsic excitability of CSNs nor their thalamocortical input was altered by a partial striatal dopamine depletion associated with αSyn pathology. Our results documented motor cortical neuronal hyperexcitability associated with αSyn aggregation and provided a novel mechanistic understanding of cortical circuit dysfunction in PD.
Collapse
Affiliation(s)
- Liqiang Chen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| | - Hiba Douja Chehade
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| | - Hong-Yuan Chu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| |
Collapse
|
27
|
Guo J, You L, Zhou Y, Hu J, Li J, Yang W, Tang X, Sun Y, Gu Y, Dong Y, Chen X, Sato C, Zinman L, Rogaeva E, Wang J, Chen Y, Zhang M. Spatial enrichment and genomic analyses reveal the link of NOMO1 with amyotrophic lateral sclerosis. Brain 2024; 147:2826-2841. [PMID: 38643019 DOI: 10.1093/brain/awae123] [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: 10/21/2023] [Revised: 03/07/2024] [Accepted: 03/24/2024] [Indexed: 04/22/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a severe motor neuron disease with uncertain genetic predisposition in most sporadic cases. The spatial architecture of cell types and gene expression are the basis of cell-cell interactions, biological function and disease pathology, but are not well investigated in the human motor cortex, a key ALS-relevant brain region. Recent studies indicated single nucleus transcriptomic features of motor neuron vulnerability in ALS motor cortex. However, the brain regional vulnerability of ALS-associated genes and the genetic link between region-specific genes and ALS risk remain largely unclear. Here, we developed an entropy-weighted differential gene expression matrix-based tool (SpatialE) to identify the spatial enrichment of gene sets in spatial transcriptomics. We benchmarked SpatialE against another enrichment tool (multimodal intersection analysis) using spatial transcriptomics data from both human and mouse brain tissues. To investigate regional vulnerability, we analysed three human motor cortex and two dorsolateral prefrontal cortex tissues for spatial enrichment of ALS-associated genes. We also used Cell2location to estimate the abundance of cell types in ALS-related cortex layers. To dissect the link of regionally expressed genes and ALS risk, we performed burden analyses of rare loss-of-function variants detected by whole-genome sequencing in ALS patients and controls, then analysed differential gene expression in the TargetALS RNA-sequencing dataset. SpatialE showed more accurate and specific spatial enrichment of regional cell type markers than multimodal intersection analysis in both mouse brain and human dorsolateral prefrontal cortex. Spatial transcriptomic analyses of human motor cortex showed heterogeneous cell types and spatial gene expression profiles. We found that 260 manually curated ALS-associated genes are significantly enriched in layer 5 of the motor cortex, with abundant expression of upper motor neurons and layer 5 excitatory neurons. Burden analyses of rare loss-of-function variants in Layer 5-associated genes nominated NOMO1 as a novel ALS-associated gene in a combined sample set of 6814 ALS patients and 3324 controls (P = 0.029). Gene expression analyses in CNS tissues revealed downregulation of NOMO1 in ALS, which is consistent with a loss-of-function disease mechanism. In conclusion, our integrated spatial transcriptomics and genomic analyses identified regional brain vulnerability in ALS and the association of a layer 5 gene (NOMO1) with ALS risk.
Collapse
Affiliation(s)
- Jingyan Guo
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Linya You
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
- Key Laboratory of Medical Computing and Computer Assisted Intervention of Shanghai, 200032, Shanghai, China
| | - Yu Zhou
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Jiali Hu
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Jiahao Li
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
| | - Wanli Yang
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
| | - Xuelin Tang
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 0S8, Canada
| | - Yimin Sun
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Yuqi Gu
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
| | - Yi Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Xi Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Christine Sato
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 0S8, Canada
| | - Lorne Zinman
- Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurology, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 0S8, Canada
- Division of Neurology, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Jian Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Yan Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, 200040, Shanghai, China
| | - Ming Zhang
- The First Rehabilitation Hospital of Shanghai, Department of Medical Genetics, School of Medicine, Tongji University, 200090, Shanghai, China
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, School of Medicine, Tongji University, 200331, Shanghai, China
- Institute for Advanced Study, Tongji University, 200092, Shanghai, China
| |
Collapse
|
28
|
Gcwensa NZ, Russell DL, Long KY, Brzozowski CF, Liu X, Gamble KL, Cowell RM, Volpicelli-Daley LA. Cortico-amygdala synaptic structural abnormalities produced by templated aggregation of α-synuclein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594419. [PMID: 38798467 PMCID: PMC11118572 DOI: 10.1101/2024.05.15.594419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Parkinson's disease (PD) and Dementia with Lewy bodies (DLB) are characterized by neuronal α-synuclein (α-syn) inclusions termed Lewy Pathology, which are abundant in the amygdala. The basolateral amygdala (BLA), in particular, receives projections from the thalamus and cortex. These projections play a role in cognition and emotional processing, behaviors which are impaired in α-synucleinopathies. To understand if and how pathologic α-syn impacts the BLA requires animal models of α-syn aggregation. Injection of α-synuclein pre-formed fibrils (PFFs) into the striatum induces robust α-synuclein aggregation in excitatory neurons in the BLA that corresponds with reduced contextual fear conditioning. At early time points after aggregate formation, cortico-amygdala excitatory transmission is abolished. The goal of this project was to determine if α-syn inclusions in the BLA induce synaptic degeneration and/or morphological changes. In this study, we used C57BL/6J mice injected bilaterally with PFFs in the dorsal striatum to induce α-syn aggregate formation in the BLA. A method was developed using immunofluorescence and three-dimensional reconstruction to analyze excitatory cortico-amygdala and thalamo-amygdala presynaptic terminals closely juxtaposed to postsynaptic densities. The abundance and morphology of synapses were analyzed at 6- or 12-weeks post-injection of PFFs. α-Syn aggregate formation in the BLA did not cause a significant loss of synapses, but cortico-amygdala and thalamo-amygdala presynaptic terminals and postsynaptic densities with aggregates of α-synuclein show increased volumes, similar to previous findings in human DLB cortex, and in non-human primate models of PD. Transmission electron microscopy showed that PFF-injected mice showed reduced intervesicular distances similar to a recent study showing phospho-serine-129 α-synuclein increases synaptic vesicle clustering. Thus, pathologic α-synuclein causes major alterations to synaptic architecture in the BLA, potentially contributing to behavioral impairment and amygdala dysfunction observed in synucleinopathies.
Collapse
|
29
|
Woerman AL, Luk KC. Are Preformed Fibrils a Model of Parkinson's Disease? JOURNAL OF PARKINSON'S DISEASE 2024; 14:1095-1103. [PMID: 39031387 PMCID: PMC11380230 DOI: 10.3233/jpd-240228] [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] [Accepted: 06/19/2024] [Indexed: 07/22/2024]
Abstract
Pre-formed fibrils (PFFs) made from recombinant α-synuclein are broadly used throughout the field in cellular and animal models of Parkinson's disease. However, their ability to successfully recapitulate disease biology is a controversial topic. In this article, two researchers debate this issue with Amanda Woerman taking the view that PFFs are a model of synucleinopathy but not Parkinson's disease, while Kelvin Luk defends their use as an important tool in the field.
Collapse
Affiliation(s)
- Amanda L. Woerman
- Department of Microbiology, Immunology, and Pathology, Prion Research Center, Colorado State University, Fort Collins, CO, USA
| | - Kelvin C. Luk
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
Mercado G, Kaeufer C, Richter F, Peelaerts W. Infections in the Etiology of Parkinson's Disease and Synucleinopathies: A Renewed Perspective, Mechanistic Insights, and Therapeutic Implications. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1301-1329. [PMID: 39331109 PMCID: PMC11492057 DOI: 10.3233/jpd-240195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Increasing evidence suggests a potential role for infectious pathogens in the etiology of synucleinopathies, a group of age-related neurodegenerative disorders including Parkinson's disease (PD), multiple system atrophy and dementia with Lewy bodies. In this review, we discuss the link between infections and synucleinopathies from a historical perspective, present emerging evidence that supports this link, and address current research challenges with a focus on neuroinflammation. Infectious pathogens can elicit a neuroinflammatory response and modulate genetic risk in PD and related synucleinopathies. The mechanisms of how infections might be linked with synucleinopathies as well as the overlap between the immune cellular pathways affected by virulent pathogens and disease-related genetic risk factors are discussed. Here, an important role for α-synuclein in the immune response against infections is emerging. Critical methodological and knowledge gaps are addressed, and we provide new future perspectives on how to address these gaps. Understanding how infections and neuroinflammation influence synucleinopathies will be essential for the development of early diagnostic tools and novel therapies.
Collapse
Affiliation(s)
- Gabriela Mercado
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Kaeufer
- Center for Systems Neuroscience, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wouter Peelaerts
- Laboratory for Virology and Gene Therapy, Department of Pharmacy and Pharmaceutical Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|