1
|
Daniel Estrella L, Trease AJ, Sheldon L, Roland NJ, Fox HS, Stauch KL. Tau association with synaptic mitochondria coincides with energetic dysfunction and excitatory synapse loss in the P301S tauopathy mouse model. Neurobiol Aging 2025; 147:163-175. [PMID: 39778459 DOI: 10.1016/j.neurobiolaging.2024.12.006] [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: 09/06/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Neurodegenerative Tauopathies are a part of several neurological disorders and aging-related diseases including, but not limited to, Alzheimer's Disease, Frontotemporal Dementia with Parkinsonism, and Chronic Traumatic Encephalopathy. The major hallmarks present in these conditions include Tau pathology (composed of hyperphosphorylated Tau tangles) and synaptic loss. in vivo studies linking Tau pathology and mitochondrial alterations at the synapse, an avenue that could lead to synaptic loss, remain predominantly scarce. For this reason, using 3-month-old wild-type and human mutant Tau P301S transgenic mice, we investigated the association of Tau with mitochondria, synaptosome bioenergetics, and characterized excitatory synaptic loss across hippocampal regions (Dentate Gyrus, perisomatic CA3, and perisomatic CA1) and in the parietal cortex. We found a significant loss of excitatory synapses in the parietal cortex and hippocampal Dentate Gyrus (DG) of Tau P301S mice. Furthermore, we found that Tau (total and disease-relevant phosphorylated Tau) associates with both the non-synaptic and synaptic mitochondria of Tau P301S mice and this coincided with synaptic mitochondrial dysfunction. The findings presented here suggest that Tau associates with mitochondria at the synapse, leading to synaptic mitochondrial dysfunction, and likely contributing to synaptic loss.
Collapse
Affiliation(s)
- L Daniel Estrella
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Andrew J Trease
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Lexi Sheldon
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Nashanthea J Roland
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Howard S Fox
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA
| | - Kelly L Stauch
- University of Nebraska Medical Center, College of Medicine, Department of Neurological Sciences, Omaha, NE, USA.
| |
Collapse
|
2
|
Majewski S, Klein P, Boillée S, Clarke BE, Patani R. Towards an integrated approach for understanding glia in Amyotrophic Lateral Sclerosis. Glia 2025; 73:591-607. [PMID: 39318236 PMCID: PMC11784848 DOI: 10.1002/glia.24622] [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: 04/17/2024] [Revised: 09/03/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Substantial advances in technology are permitting a high resolution understanding of the salience of glia, and have helped us to transcend decades of predominantly neuron-centric research. In particular, recent advances in 'omic' technologies have enabled unique insights into glial biology, shedding light on the cellular and molecular aspects of neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). Here, we review studies using omic techniques to attempt to understand the role of glia in ALS across different model systems and post mortem tissue. We also address caveats that should be considered when interpreting such studies, and how some of these may be mitigated through either using a multi-omic approach and/or careful low throughput, high fidelity orthogonal validation with particular emphasis on functional validation. Finally, we consider emerging technologies and their potential relevance in deepening our understanding of glia in ALS.
Collapse
Affiliation(s)
- Stanislaw Majewski
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Pierre Klein
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, APHPHôpital de la Pitié‐SalpêtrièreParisFrance
| | - Benjamin E. Clarke
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| | - Rickie Patani
- Department of Neuromuscular Diseases, Queen Square Institute of NeurologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| |
Collapse
|
3
|
Sun Q, Zhu J, Zhao X, Huang X, Qu W, Tang X, Ma D, Shu Q, Li X. Mettl3-m 6A-NPY axis governing neuron-microglia interaction regulates sleep amount of mice. Cell Discov 2025; 11:10. [PMID: 39905012 PMCID: PMC11794856 DOI: 10.1038/s41421-024-00756-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/13/2024] [Indexed: 02/06/2025] Open
Abstract
Sleep behavior is regulated by diverse mechanisms including genetics, neuromodulation and environmental signals. However, it remains completely unknown regarding the roles of epitranscriptomics in regulating sleep behavior. In the present study, we showed that the deficiency of RNA m6A methyltransferase Mettl3 in excitatory neurons specifically induces microglia activation, neuroinflammation and neuronal loss in thalamus of mice. Mettl3 deficiency remarkably disrupts sleep rhythm and reduces the amount of non-rapid eye movement sleep. We also showed that Mettl3 regulates neuropeptide Y (NPY) via m6A modification and Mettl3 conditional knockout (cKO) mice displayed significantly decreased expression of NPY in thalamus. In addition, the dynamic distribution pattern of NPY is observed during wake-sleep cycle in cKO mice. Ectopic expression of Mettl3 and NPY significantly inhibits microglia activation and neuronal loss in thalamus, and restores the disrupted sleep behavior of cKO mice. Collectively, our study has revealed the critical function of Mettl3-m6A-NPY axis in regulating sleep behavior.
Collapse
Affiliation(s)
- Qihang Sun
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinpiao Zhu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xingsen Zhao
- Institute of Biotechnology, Xianghu Laboratory, Hangzhou, Zhejiang, China
| | - Xiaoli Huang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Wenzheng Qu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xia Tang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Daqing Ma
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Division of Anesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| | - Qiang Shu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xuekun Li
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
McCallister TX, Lim CKW, Singh M, Zhang S, Ahsan NS, Terpstra WM, Xiong AY, Zeballos C MA, Powell JE, Drnevich J, Kang Y, Gaj T. A high-fidelity CRISPR-Cas13 system improves abnormalities associated with C9ORF72-linked ALS/FTD. Nat Commun 2025; 16:460. [PMID: 39779681 PMCID: PMC11711314 DOI: 10.1038/s41467-024-55548-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
An abnormal expansion of a GGGGCC (G4C2) hexanucleotide repeat in the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two debilitating neurodegenerative disorders driven in part by gain-of-function mechanisms involving transcribed forms of the repeat expansion. By utilizing a Cas13 variant with reduced collateral effects, we develop here a high-fidelity RNA-targeting CRISPR-based system for C9ORF72-linked ALS/FTD. When delivered to the brain of a transgenic rodent model, this Cas13-based platform curbed the expression of the G4C2 repeat-containing RNA without affecting normal C9ORF72 levels, which in turn decreased the formation of RNA foci, reduced the production of a dipeptide repeat protein, and reversed transcriptional deficits. This high-fidelity system possessed improved transcriptome-wide specificity compared to its native form and mediated targeting in motor neuron-like cells derived from a patient with ALS. These results lay the foundation for the implementation of RNA-targeting CRISPR technologies for C9ORF72-linked ALS/FTD.
Collapse
Affiliation(s)
- Tristan X McCallister
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Colin K W Lim
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Mayuri Singh
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Sijia Zhang
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Najah S Ahsan
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - William M Terpstra
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Alisha Y Xiong
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - M Alejandra Zeballos C
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jackson E Powell
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jenny Drnevich
- High-Performance Biological Computing, Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Yifei Kang
- High-Performance Biological Computing, Roy J. Carver Biotechnology Center, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Thomas Gaj
- Department of Bioengineering, The Grainger College of Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
5
|
Kloske CM, Mahinrad S, Barnum CJ, Batista AF, Bradshaw EM, Butts B, Carrillo MC, Chakrabarty P, Chen X, Craft S, Da Mesquita S, Dabin LC, Devanand D, Duran‐Laforet V, Elyaman W, Evans EE, Fitzgerald‐Bocarsly P, Foley KE, Harms AS, Heneka MT, Hong S, Huang YA, Jackvony S, Lai L, Guen YL, Lemere CA, Liddelow SA, Martin‐Peña A, Orr AG, Quintana FJ, Ramey GD, Rexach JE, Rizzo SJS, Sexton C, Tang AS, Torrellas JG, Tsai AP, van Olst L, Walker KA, Wharton W, Tansey MG, Wilcock DM. Advancements in Immunity and Dementia Research: Highlights from the 2023 AAIC Advancements: Immunity Conference. Alzheimers Dement 2025; 21:e14291. [PMID: 39692624 PMCID: PMC11772715 DOI: 10.1002/alz.14291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/23/2024] [Accepted: 09/07/2024] [Indexed: 12/19/2024]
Abstract
The immune system is a key player in the onset and progression of neurodegenerative disorders. While brain resident immune cell-mediated neuroinflammation and peripheral immune cell (eg, T cell) infiltration into the brain have been shown to significantly contribute to Alzheimer's disease (AD) pathology, the nature and extent of immune responses in the brain in the context of AD and related dementias (ADRD) remain unclear. Furthermore, the roles of the peripheral immune system in driving ADRD pathology remain incompletely elucidated. In March of 2023, the Alzheimer's Association convened the Alzheimer's Association International Conference (AAIC), Advancements: Immunity, to discuss the roles of the immune system in ADRD. A wide range of topics were discussed, such as animal models that replicate human pathology, immune-related biomarkers and clinical trials, and lessons from other fields describing immune responses in neurodegeneration. This manuscript presents highlights from the conference and outlines avenues for future research on the roles of immunity in neurodegenerative disorders. HIGHLIGHTS: The immune system plays a central role in the pathogenesis of Alzheimer's disease. The immune system exerts numerous effects throughout the brain on amyloid-beta, tau, and other pathways. The 2023 AAIC, Advancements: Immunity, encouraged discussions and collaborations on understanding the role of the immune system.
Collapse
|
6
|
Wu M, Fletcher EL, Chinnery HR, Downie LE, Mueller SN. Redefining our vision: an updated guide to the ocular immune system. Nat Rev Immunol 2024; 24:896-911. [PMID: 39215057 DOI: 10.1038/s41577-024-01064-y] [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: 07/01/2024] [Indexed: 09/04/2024]
Abstract
Balanced immune responses in the eyes are crucial to preserve vision. The ocular immune system has long been considered distinct, owing to the so-called 'immune privilege' of its component tissues. More recently, intravital imaging and transcriptomic techniques have reshaped scientific understanding of the ocular immune landscape, such as revealing the specialization of immune cell populations in the various tissues of the eye. As knowledge of the phenotypes of corneal and retinal immune cells has evolved, links to both the systemic immune system, and the central and peripheral nervous systems, have been identified. Using intravital imaging, T cells have recently been found to reside in, and actively patrol, the healthy human cornea. Disease-associated retinal microglia with links to retinal degeneration have also been identified. This Review provides an updated guide to the ocular immune system, highlighting current knowledge of the immune cells that are present in steady-state and specific diseased ocular tissues, as well as evidence for their relationship to systemic disease. In addition, we discuss emerging intravital imaging techniques that can be used to visualize immune cell morphology and dynamics in living human eyes and how these could be applied to advance understanding of the human immune system.
Collapse
Affiliation(s)
- Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Physiology, The University of Melbourne, Carlton, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
- Lions Eye Institute, Nedlands, Western Australia, Australia.
- Optometry, The University of Western Australia, Crawley, Western Australia, Australia.
| | - Laura E Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, Victoria, Australia.
| | - Scott N Mueller
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [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: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
8
|
Ito D, Okada K. Rethinking antisense oligonucleotide therapeutics for amyotrophic lateral sclerosis. Ann Clin Transl Neurol 2024; 11:3054-3063. [PMID: 39473221 DOI: 10.1002/acn3.52234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/18/2024] Open
Abstract
Antisense oligonucleotides, which are used to silence target genes, are gaining attention as a novel drug discovery modality for proteinopathies. However, while clinical trials for neurodegenerative diseases like amyotrophic lateral sclerosis have been conducted in recent years, the results have not always been favorable. The results from a Phase III trial of the antisense oligonucleotide, that is, tofersen, which targets SOD1 mRNA, showed decreased levels of cerebrospinal fluid SOD1 and plasma neurofilament light chain but no improvements in primary clinical endpoint. Moreover, case reports pertaining to patients with amyotrophic lateral sclerosis carrying FUS and C9orf72 mutations who received antisense oligonucleotide-based treatments have demonstrated a notable reduction in the targeted protein (thus providing the proof of mechanism) but with no discernible clinical benefits. There are several possible reasons why antisense oligonucleotides knockdown fails to achieve proof of concept, which need to be addressed: on-target adverse effects resulting from the loss of function of target gene and irreversible neuronal death cascade due to toxic protein accumulation, among other factors. This review provides an overview of the current status and discusses the prospects of antisense oligonucleotides treatment for amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Daisuke Ito
- Memory Center, Keio University School of Medicine, Tokyo, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Kensuke Okada
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
9
|
Tuddenham JF, Fujita M, Khairallah A, Harbison C, Flowers XE, Coronas-Samano G, Maniatis S, Daly A, Schneider JA, Teich AF, Vonsattel JPG, Sims PA, Elyaman W, Bradshaw EM, Phatnani H, Shneider N, Bennett DA, De Jager PL, Przedborski S, Menon V, Olah M. Single-cell transcriptomic landscape of the neuroimmune compartment in amyotrophic lateral sclerosis brain and spinal cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623183. [PMID: 39605399 PMCID: PMC11601279 DOI: 10.1101/2024.11.12.623183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Development of therapeutic approaches that target specific microglia responses in amyotrophic lateral sclerosis (ALS) is crucial due to the involvement of microglia in ALS progression. Our study identifies the predominant microglia subset in human ALS primary motor cortex and spinal cord as an undifferentiated phenotype with dysregulated respiratory electron transport. Moreover, we find that the interferon response microglia subset is enriched in donors with aggressive disease progression, while a previously described potentially protective microglia phenotype is depleted in ALS. Additionally, we observe an enrichment of non-microglial immune cell, mainly NK/T cells, in ALS central nervous system, primarily in the spinal cord. These findings pave the way for the development of microglia subset-specific therapeutic interventions to slow or even stop ALS progression.
Collapse
|
10
|
Trainor AR, MacDonald DS, Penney J. Microglia: roles and genetic risk in Parkinson's disease. Front Neurosci 2024; 18:1506358. [PMID: 39554849 PMCID: PMC11564156 DOI: 10.3389/fnins.2024.1506358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
The prevalence of neurodegenerative disorders such as Parkinson's disease are increasing as world populations age. Despite this growing public health concern, the precise molecular and cellular mechanisms that culminate in neurodegeneration remain unclear. Effective treatment options for Parkinson's disease and other neurodegenerative disorders remain very limited, due in part to this uncertain disease etiology. One commonality across neurodegenerative diseases is sustained neuroinflammation, mediated in large part by microglia, the innate immune cells of the brain. Initially thought to simply react to neuron-derived pathology, genetic and functional studies in recent years suggest that microglia play a more active role in the neurodegenerative process than previously appreciated. Here, we review evidence for the roles of microglia in Parkinson's disease pathogenesis and progression, with a particular focus on microglial functions that are perturbed by disease associated genes and mutations.
Collapse
Affiliation(s)
| | | | - Jay Penney
- Department of Biomedical Sciences, AVC, University of Prince Edward Island, Charlottetown, PE, Canada
| |
Collapse
|
11
|
Mubeen H, Masood A, Zafar A, Khan ZQ, Khan MQ, Nisa AU. Insights into AlphaFold's breakthrough in neurodegenerative diseases. Ir J Med Sci 2024; 193:2577-2588. [PMID: 38833116 DOI: 10.1007/s11845-024-03721-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/19/2024] [Indexed: 06/06/2024]
Abstract
Neurodegenerative diseases (ND) are disorders of the central nervous system (CNS) characterized by impairment in neurons' functions, and complete loss, leading to memory loss, and difficulty in learning, language, and movement processes. The most common among these NDs are Alzheimer's disease (AD) and Parkinson's disease (PD), although several other disorders also exist. These are frontotemporal dementia (FTD), amyotrophic lateral syndrome (ALS), Huntington's disease (HD), and others; the major pathological hallmark of NDs is the proteinopathies, either of amyloid-β (Aβ), tauopathies, or synucleinopathies. Aggregation of proteins that do not undergo normal configuration, either due to mutations or through some disturbance in cellular pathway contributes to the diseases. Artificial Intelligence (AI) and deep learning (DL) have proven to be successful in the diagnosis and treatment of various congenital diseases. DL approaches like AlphaFold (AF) are a major leap towards success in CNS disorders. This 3D protein geometry modeling algorithm developed by DeepMind has the potential to revolutionize biology. AF has the potential to predict 3D-protein confirmation at an accuracy level comparable to experimentally predicted one, with the additional advantage of precisely estimating protein interactions. This breakthrough will be beneficial to identify diseases' advancement and the disturbance of signaling pathways stimulating impaired functions of proteins. Though AlphaFold has solved a major problem in structural biology, it cannot predict membrane proteins-a beneficial approach for drug designing.
Collapse
Affiliation(s)
- Hira Mubeen
- Department of Biotechnology, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan.
| | - Ammara Masood
- Department of Biotechnology, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan
| | - Asma Zafar
- Department of Biotechnology, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan
| | - Zohaira Qayyum Khan
- Department of Biotechnology, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan
| | - Muneeza Qayyum Khan
- Department of Biotechnology, Faculty of Science & Technology, University of Central Punjab, Lahore, Pakistan
| | - Alim Un Nisa
- Pakistan Council of Scientific and Industrial Research, Lahore, Pakistan
| |
Collapse
|
12
|
van der Geest AT, Jakobs CE, Ljubikj T, Huffels CFM, Cañizares Luna M, Vieira de Sá R, Adolfs Y, de Wit M, Rutten DH, Kaal M, Zwartkruis MM, Carcolé M, Groen EJN, Hol EM, Basak O, Isaacs AM, Westeneng HJ, van den Berg LH, Veldink JH, Schlegel DK, Pasterkamp RJ. Molecular pathology, developmental changes and synaptic dysfunction in (pre-) symptomatic human C9ORF72-ALS/FTD cerebral organoids. Acta Neuropathol Commun 2024; 12:152. [PMID: 39289761 PMCID: PMC11409520 DOI: 10.1186/s40478-024-01857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
A hexanucleotide repeat expansion (HRE) in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Human brain imaging and experimental studies indicate early changes in brain structure and connectivity in C9-ALS/FTD, even before symptom onset. Because these early disease phenotypes remain incompletely understood, we generated iPSC-derived cerebral organoid models from C9-ALS/FTD patients, presymptomatic C9ORF72-HRE (C9-HRE) carriers, and controls. Our work revealed the presence of all three C9-HRE-related molecular pathologies and developmental stage-dependent size phenotypes in cerebral organoids from C9-ALS/FTD patients. In addition, single-cell RNA sequencing identified changes in cell type abundance and distribution in C9-ALS/FTD organoids, including a reduction in the number of deep layer cortical neurons and the distribution of neural progenitors. Further, molecular and cellular analyses and patch-clamp electrophysiology detected various changes in synapse structure and function. Intriguingly, organoids from all presymptomatic C9-HRE carriers displayed C9-HRE molecular pathology, whereas the extent to which more downstream cellular defects, as found in C9-ALS/FTD models, were detected varied for the different presymptomatic C9-HRE cases. Together, these results unveil early changes in 3D human brain tissue organization and synaptic connectivity in C9-ALS/FTD that likely constitute initial pathologies crucial for understanding disease onset and the design of therapeutic strategies.
Collapse
Affiliation(s)
- Astrid T van der Geest
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Channa E Jakobs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tijana Ljubikj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan F M Huffels
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Renata Vieira de Sá
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Daan H Rutten
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marthe Kaal
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maria M Zwartkruis
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Ewout J N Groen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henk-Jan Westeneng
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Domino K Schlegel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
13
|
Steffke C, Agarwal S, Kabashi E, Catanese A. Overexpression of Toxic Poly(Glycine-Alanine) Aggregates in Primary Neuronal Cultures Induces Time-Dependent Autophagic and Synaptic Alterations but Subtle Activity Impairments. Cells 2024; 13:1300. [PMID: 39120329 PMCID: PMC11311834 DOI: 10.3390/cells13151300] [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: 07/04/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
The pathogenic expansion of the intronic GGGGCC hexanucleotide located in the non-coding region of the C9orf72 gene represents the most frequent genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This mutation leads to the accumulation of toxic RNA foci and dipeptide repeats (DPRs), as well as reduced levels of the C9orf72 protein. Thus, both gain and loss of function are coexisting pathogenic aspects linked to C9orf72-ALS/FTD. Synaptic alterations have been largely described in C9orf72 models, but it is still not clear which aspect of the pathology mostly contributes to these impairments. To address this question, we investigated the dynamic changes occurring over time at the synapse upon accumulation of poly(GA), the most abundant DPR. Overexpression of this toxic form induced a drastic loss of synaptic proteins in primary neuron cultures, anticipating autophagic defects. Surprisingly, the dramatic impairment characterizing the synaptic proteome was not fully matched by changes in network properties. In fact, high-density multi-electrode array analysis highlighted only minor reductions in the spike number and firing rate of poly(GA) neurons. Our data show that the toxic gain of function linked to C9orf72 affects the synaptic proteome but exerts only minor effects on the network activity.
Collapse
Affiliation(s)
- Christina Steffke
- Institute of Anatomy and Cell Biology, University of Ulm, 89069 Ulm, Germany; (C.S.); (S.A.)
- Department of Neurology, University of Ulm, 89069 Ulm, Germany
- International Graduate School in Molecular Medicine of Ulm (IGraDU), University of Ulm, 89069 Ulm, Germany
| | - Shreya Agarwal
- Institute of Anatomy and Cell Biology, University of Ulm, 89069 Ulm, Germany; (C.S.); (S.A.)
| | - Edor Kabashi
- Institute Imagine, Necker-Enfants Malades Hospital, University Paris Descartes, 75015 Paris, France;
| | - Alberto Catanese
- Institute of Anatomy and Cell Biology, University of Ulm, 89069 Ulm, Germany; (C.S.); (S.A.)
- Institute Imagine, Necker-Enfants Malades Hospital, University Paris Descartes, 75015 Paris, France;
| |
Collapse
|
14
|
Clayton EL, Huggon L, Cousin MA, Mizielinska S. Synaptopathy: presynaptic convergence in frontotemporal dementia and amyotrophic lateral sclerosis. Brain 2024; 147:2289-2307. [PMID: 38451707 PMCID: PMC11224618 DOI: 10.1093/brain/awae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Frontotemporal dementia and amyotrophic lateral sclerosis are common forms of neurodegenerative disease that share overlapping genetics and pathologies. Crucially, no significantly disease-modifying treatments are available for either disease. Identifying the earliest changes that initiate neuronal dysfunction is important for designing effective intervention therapeutics. The genes mutated in genetic forms of frontotemporal dementia and amyotrophic lateral sclerosis have diverse cellular functions, and multiple disease mechanisms have been proposed for both. Identification of a convergent disease mechanism in frontotemporal dementia and amyotrophic lateral sclerosis would focus research for a targetable pathway, which could potentially effectively treat all forms of frontotemporal dementia and amyotrophic lateral sclerosis (both familial and sporadic). Synaptopathies are diseases resulting from physiological dysfunction of synapses, and define the earliest stages in multiple neuronal diseases, with synapse loss a key feature in dementia. At the presynapse, the process of synaptic vesicle recruitment, fusion and recycling is necessary for activity-dependent neurotransmitter release. The unique distal location of the presynaptic terminal means the tight spatio-temporal control of presynaptic homeostasis is dependent on efficient local protein translation and degradation. Recently, numerous publications have shown that mutations associated with frontotemporal dementia and amyotrophic lateral sclerosis present with synaptopathy characterized by presynaptic dysfunction. This review will describe the complex local signalling and membrane trafficking events that occur at the presynapse to facilitate neurotransmission and will summarize recent publications linking frontotemporal dementia/amyotrophic lateral sclerosis genetic mutations to presynaptic function. This evidence indicates that presynaptic synaptopathy is an early and convergent event in frontotemporal dementia and amyotrophic lateral sclerosis and illustrates the need for further research in this area, to identify potential therapeutic targets with the ability to impact this convergent pathomechanism.
Collapse
Affiliation(s)
- Emma L Clayton
- UK Dementia Research Institute at King’s College London, London SE5 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| | - Laura Huggon
- UK Dementia Research Institute at King’s College London, London SE5 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sarah Mizielinska
- UK Dementia Research Institute at King’s College London, London SE5 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| |
Collapse
|
15
|
Xie M, Miller AS, Pallegar PN, Umpierre A, Liang Y, Wang N, Zhang S, Nagaraj NK, Fogarty ZC, Ghayal NB, Oskarsson B, Zhao S, Zheng J, Qi F, Nguyen A, Dickson DW, Wu LJ. Rod-shaped microglia interact with neuronal dendrites to regulate cortical excitability in TDP-43 related neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.30.601396. [PMID: 39005475 PMCID: PMC11244918 DOI: 10.1101/2024.06.30.601396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Motor cortical hyperexcitability is well-documented in the presymptomatic stage of amyotrophic lateral sclerosis (ALS). However, the mechanisms underlying this early dysregulation are not fully understood. Microglia, as the principal immune cells of the central nervous system, have emerged as important players in sensing and regulating neuronal activity. Here we investigated the role of microglia in the motor cortical circuits in a mouse model of TDP-43 neurodegeneration (rNLS8). Utilizing multichannel probe recording and longitudinal in vivo calcium imaging in awake mice, we observed neuronal hyperactivity at the initial stage of disease progression. Spatial and single-cell RNA sequencing revealed that microglia are the primary responders to motor cortical hyperactivity. We further identified a unique subpopulation of microglia, rod-shaped microglia, which are characterized by a distinct morphology and transcriptional profile. Notably, rod-shaped microglia predominantly interact with neuronal dendrites and excitatory synaptic inputs to attenuate motor cortical hyperactivity. The elimination of rod-shaped microglia through TREM2 deficiency increased neuronal hyperactivity, exacerbated motor deficits, and further decreased survival rates of rNLS8 mice. Together, our results suggest that rod-shaped microglia play a neuroprotective role by attenuating cortical hyperexcitability in the mouse model of TDP-43 related neurodegeneration.
Collapse
|
16
|
Jaroszynska N, Salzinger A, Tsarouchas TM, Becker CG, Becker T, Lyons DA, MacDonald RB, Keatinge M. C9ORF72 Deficiency Results in Neurodegeneration in the Zebrafish Retina. J Neurosci 2024; 44:e2128232024. [PMID: 38658168 PMCID: PMC11209673 DOI: 10.1523/jneurosci.2128-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/19/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
Hexanucleotide repeat expansions within the gene C9ORF72 are the most common cause of the neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This disease-causing expansion leads to a reduction in C9ORF72 expression levels in patients, suggesting loss of C9ORF72 function could contribute to disease. To further understand the consequences of C9ORF72 deficiency in vivo, we generated a c9orf72 mutant zebrafish line. Analysis of the adult female spinal cords revealed no appreciable neurodegenerative pathology such as loss of motor neurons or increased levels of neuroinflammation. However, detailed examination of adult female c9orf72-/- retinas showed prominent neurodegenerative features, including a decrease in retinal thickness, gliosis, and an overall reduction in neurons of all subtypes. Analysis of rod and cone cells within the photoreceptor layer showed a disturbance in their outer segment structure and rhodopsin mislocalization from rod outer segments to their cell bodies and synaptic terminals. Thus, C9ORF72 may play a previously unappreciated role in retinal homeostasis and suggests C9ORF72 deficiency can induce tissue specific neuronal loss.
Collapse
Affiliation(s)
- Natalia Jaroszynska
- Institute of Ophthalmology, University College London, London EC1Y 0AD, United Kingdom
| | - Andrea Salzinger
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Themistoklis M Tsarouchas
- Department of Psychiatry and Behavioural Sciences, Stanford University School of Medicine, Palo Alto, California 94305
| | - Catherina G Becker
- Center for Regenerative Therapies Dresden (CRTD), Dresden 01307, Germany
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, United Kingdom
| | - Thomas Becker
- Center for Regenerative Therapies Dresden (CRTD), Dresden 01307, Germany
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, United Kingdom
| | - David A Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4SB, United Kingdom
| | - Ryan B MacDonald
- Institute of Ophthalmology, University College London, London EC1Y 0AD, United Kingdom
| | - Marcus Keatinge
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| |
Collapse
|
17
|
Beiter RM, Sheehan PW, Schafer DP. Microglia phagocytic mechanisms: Development informing disease. Curr Opin Neurobiol 2024; 86:102877. [PMID: 38631077 PMCID: PMC11162951 DOI: 10.1016/j.conb.2024.102877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024]
Abstract
Microglia are tissue-resident macrophages and professional phagocytes of the central nervous system (CNS). In development, microglia-mediated phagocytosis is important for sculpting the cellular architecture. This includes the engulfment of dead/dying cells, pruning extranumerary synapses and axons, and phagocytosing fragments of myelin sheaths. Intriguingly, these developmental phagocytic mechanisms by which microglia sculpt the CNS are now appreciated as important for eliminating synapses, myelin, and proteins during neurodegeneration. Here, we discuss parallels between neurodevelopment and neurodegeneration, which highlights how development is informing disease. We further discuss recent advances and challenges towards therapeutically targeting these phagocytic pathways and how we can leverage development to overcome these challenges.
Collapse
Affiliation(s)
- Rebecca M Beiter
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Patrick W Sheehan
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
18
|
Sellier C, Corcia P, Vourc'h P, Dupuis L. C9ORF72 hexanucleotide repeat expansion: From ALS and FTD to a broader pathogenic role? Rev Neurol (Paris) 2024; 180:417-428. [PMID: 38609750 DOI: 10.1016/j.neurol.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024]
Abstract
The major gene underlying monogenic forms of amyotrophic lateral sclerosis (ALS) and fronto-temporal dementia (FTD) is C9ORF72. The causative mutation in C9ORF72 is an abnormal hexanucleotide (G4C2) repeat expansion (HRE) located in the first intron of the gene. The aim of this review is to propose a comprehensive update on recent developments on clinical, biological and therapeutics aspects related to C9ORF72 in order to highlight the current understanding of genotype-phenotype correlations, and also on biological machinery leading to neuronal death. We will particularly focus on the broad phenotypic presentation of C9ORF72-related diseases, that goes well beyond the classical phenotypes observed in ALS and FTD patients. Last, we will comment the possible therapeutical hopes for patients carrying a C9ORF72 HRE.
Collapse
Affiliation(s)
- C Sellier
- Centre de recherches en biomédecine de Strasbourg, UMR-S1329, Inserm, université de Strasbourg, Strasbourg, France
| | - P Corcia
- UMR 1253 iBrain, Inserm, université de Tours, Tours, France; Centre constitutif de coordination SLA, CHU de Bretonneau, 2, boulevard Tonnelle, 37044 Tours cedex 1, France
| | - P Vourc'h
- UMR 1253 iBrain, Inserm, université de Tours, Tours, France; Service de biochimie et biologie moléculaire, CHU de Tours, Tours, France
| | - L Dupuis
- Centre de recherches en biomédecine de Strasbourg, UMR-S1329, Inserm, université de Strasbourg, Strasbourg, France.
| |
Collapse
|
19
|
Ounissi M, Latouche M, Racoceanu D. PhagoStat a scalable and interpretable end to end framework for efficient quantification of cell phagocytosis in neurodegenerative disease studies. Sci Rep 2024; 14:6482. [PMID: 38499658 PMCID: PMC10948879 DOI: 10.1038/s41598-024-56081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 03/01/2024] [Indexed: 03/20/2024] Open
Abstract
Quantifying the phagocytosis of dynamic, unstained cells is essential for evaluating neurodegenerative diseases. However, measuring rapid cell interactions and distinguishing cells from background make this task very challenging when processing time-lapse phase-contrast video microscopy. In this study, we introduce an end-to-end, scalable, and versatile real-time framework for quantifying and analyzing phagocytic activity. Our proposed pipeline is able to process large data-sets and includes a data quality verification module to counteract potential perturbations such as microscope movements and frame blurring. We also propose an explainable cell segmentation module to improve the interpretability of deep learning methods compared to black-box algorithms. This includes two interpretable deep learning capabilities: visual explanation and model simplification. We demonstrate that interpretability in deep learning is not the opposite of high performance, by additionally providing essential deep learning algorithm optimization insights and solutions. Besides, incorporating interpretable modules results in an efficient architecture design and optimized execution time. We apply this pipeline to quantify and analyze microglial cell phagocytosis in frontotemporal dementia (FTD) and obtain statistically reliable results showing that FTD mutant cells are larger and more aggressive than control cells. The method has been tested and validated on several public benchmarks by generating state-of-the art performances. To stimulate translational approaches and future studies, we release an open-source end-to-end pipeline and a unique microglial cells phagocytosis dataset for immune system characterization in neurodegenerative diseases research. This pipeline and the associated dataset will consistently crystallize future advances in this field, promoting the development of efficient and effective interpretable algorithms dedicated to the critical domain of neurodegenerative diseases' characterization. https://github.com/ounissimehdi/PhagoStat .
Collapse
Affiliation(s)
- Mehdi Ounissi
- CNRS, Inserm, AP-HP, Inria, Paris Brain Institute-ICM, Sorbonne University, 75013, Paris, France
| | - Morwena Latouche
- Inserm, CNRS, AP-HP, Institut du Cerveau, ICM, Sorbonne Université, 75013, Paris, France
- PSL Research university, EPHE, Paris, France
| | - Daniel Racoceanu
- CNRS, Inserm, AP-HP, Inria, Paris Brain Institute-ICM, Sorbonne University, 75013, Paris, France.
| |
Collapse
|
20
|
Limone F, Couto A, Wang JY, Zhang Y, McCourt B, Huang C, Minkin A, Jani M, McNeer S, Keaney J, Gillet G, Gonzalez RL, Goodman WA, Kadiu I, Eggan K, Burberry A. Myeloid and lymphoid expression of C9orf72 regulates IL-17A signaling in mice. Sci Transl Med 2024; 16:eadg7895. [PMID: 38295187 PMCID: PMC11247723 DOI: 10.1126/scitranslmed.adg7895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
A mutation in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Patients with ALS or FTD often develop autoimmunity and inflammation that precedes or coincides with the onset of neurological symptoms, but the underlying mechanisms are poorly understood. Here, we knocked out murine C9orf72 in seven hematopoietic progenitor compartments by conditional mutagenesis and found that myeloid lineage C9orf72 prevents splenomegaly, loss of tolerance, and premature mortality. Furthermore, we demonstrated that C9orf72 plays a role in lymphoid cells to prevent interleukin-17A (IL-17A) production and neutrophilia. Mass cytometry identified early and sustained elevation of the costimulatory molecule CD80 expressed on C9orf72-deficient mouse macrophages, monocytes, and microglia. Enrichment of CD80 was similarly observed in human spinal cord microglia from patients with C9ORF72-mediated ALS compared with non-ALS controls. Single-cell RNA sequencing of murine spinal cord, brain cortex, and spleen demonstrated coordinated induction of gene modules related to antigen processing and presentation and antiviral immunity in C9orf72-deficient endothelial cells, microglia, and macrophages. Mechanistically, C9ORF72 repressed the trafficking of CD80 to the cell surface in response to Toll-like receptor agonists, interferon-γ, and IL-17A. Deletion of Il17a in C9orf72-deficient mice prevented CD80 enrichment in the spinal cord, reduced neutrophilia, and reduced gut T helper type 17 cells. Last, systemic delivery of an IL-17A neutralizing antibody augmented motor performance and suppressed neuroinflammation in C9orf72-deficient mice. Altogether, we show that C9orf72 orchestrates myeloid costimulatory potency and provide support for IL-17A as a therapeutic target for neuroinflammation associated with ALS or FTD.
Collapse
Affiliation(s)
- Francesco Limone
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Leiden University Medical Center, LUMC, 2333 ZA Leiden, The Netherlands
| | - Alexander Couto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Jin-Yuan Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Yingying Zhang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Blake McCourt
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Cerianne Huang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Adina Minkin
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Marghi Jani
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sarah McNeer
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - James Keaney
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Gaëlle Gillet
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Rodrigo Lopez Gonzalez
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44196, USA
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Irena Kadiu
- Neuroinflammation Focus Area, UCB Biopharma SRL, Braine-l’Alleud, 1420, Belgium
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Aaron Burberry
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
21
|
Chong ZZ, Menkes DL, Souayah N. Pathogenesis underlying hexanucleotide repeat expansions in C9orf72 gene in amyotrophic lateral sclerosis. Rev Neurosci 2024; 35:85-97. [PMID: 37525497 DOI: 10.1515/revneuro-2023-0060] [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: 05/29/2023] [Accepted: 07/07/2023] [Indexed: 08/02/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disorder. Mutations in C9orf72 and the resulting hexanucleotide repeat (GGGGCC) expansion (HRE) has been identified as a major cause of familial ALS, accounting for about 40 % of familial and 6 % of sporadic cases of ALS in Western patients. The pathological outcomes of HRE expansion in ALS have been recognized as the results of two mechanisms that include both the toxic gain-of-function and loss-of-function of C9ORF72. The gain of toxicity results from RNA and dipeptide repeats (DPRs). The HRE can be bidirectionally transcribed into RNA foci, which can bind to and disrupt RNA splicing, transport, and translation. The DPRs that include poly-glycine-alanine, poly-glycine-proline, poly-glycine- arginine, poly-proline-alanine, and poly-proline-arginine can induce toxicity by direct binding and sequestrating other proteins to interfere rRNA synthesis, ribosome biogenesis, translation, and nucleocytoplasmic transport. The C9ORF72 functions through binding to its partners-Smith-Magenis chromosome regions 8 (SMCR8) and WD repeat-containing protein (WDR41). Loss of C9ORF72 function results in impairment of autophagy, deregulation of autoimmunity, increased stress, and disruption of nucleocytoplasmic transport. Further insight into the mechanism in C9ORF72 HRE pathogenesis will facilitate identifying novel and effective therapeutic targets for ALS.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Department of Neurology, Rutgers University, New Jersey Medical School, 185 S. Orange Ave, Newark, NJ 07103, USA
| | - Daniel L Menkes
- Department of Neurology, Oakland University William Beaumont School of Medicine, 3555 West 13 Mile Road, Suite N120, Royal Oak, MI 48073, USA
| | - Nizar Souayah
- Department of Neurology, Rutgers University, New Jersey Medical School, 90 Bergen Street DOC 8100, Newark, NJ 07101, USA
| |
Collapse
|
22
|
Bottillo I, Laino L, Azzarà A, Lintas C, Cassano I, Di Lazzaro V, Ursini F, Motolese F, Bargiacchi S, Formicola D, Grammatico P, Gurrieri F. A pathogenic variant in the FLCN gene presenting with pure dementia: is autophagy at the intersection between neurodegeneration and cancer? Front Neurosci 2024; 17:1304080. [PMID: 38249578 PMCID: PMC10796570 DOI: 10.3389/fnins.2023.1304080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction Folliculin, encoded by FLCN gene, plays a role in the mTORC1 autophagy cascade and its alterations are responsible for the Birt-Hogg-Dubé (BHD) syndrome, characterized by follicle hamartomas, kidney tumors and pneumothorax. Patient and results We report a 74-years-old woman diagnosed with dementia and carrying a FLCN alteration in absence of any sign of BHD. She also carried an alteration of MAT1A gene, which is also implicated in the regulation of mTORC1. Discussion The MAT1A variant could have prevented the development of a FLCN-related oncological phenotype. Conversely, our patient presented with dementia that, to date, has yet to be documented in BHD. Folliculin belongs to the DENN family proteins, which includes C9orf72 whose alteration has been associated to neurodegeneration. The folliculin perturbation could affect the C9orf72 activity and our patient could represent the first human model of a relationship between FLCN and C9orf72 across the path of autophagy.
Collapse
Affiliation(s)
- Irene Bottillo
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| | - Luigi Laino
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| | - Alessia Azzarà
- Research Unit of Medical Genetics, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Ilaria Cassano
- Research Unit of Medical Genetics, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Vincenzo Di Lazzaro
- Department of Medicine and Surgery, Unit of Neurology, Neurophysiology, Neurobiology and Psychiatry, Università Campus Bio-Medico di Roma, Rome, Italy
- Unit of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesca Ursini
- Unit of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesco Motolese
- Department of Medicine and Surgery, Unit of Neurology, Neurophysiology, Neurobiology and Psychiatry, Università Campus Bio-Medico di Roma, Rome, Italy
- Unit of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Simone Bargiacchi
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| | - Daniela Formicola
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| | - Paola Grammatico
- Division of Medical Genetics, Department of Experimental Medicine, San Camillo-Forlanini Hospital, Sapienza University, Rome, Italy
| | - Fiorella Gurrieri
- Research Unit of Medical Genetics, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
23
|
Tang D, Zheng K, Zhu J, Jin X, Bao H, Jiang L, Li H, Wang Y, Lu Y, Liu J, Liu H, Tang C, Feng S, Dong X, Xu L, Yin Y, Dang S, Wei X, Ren H, Dong B, Dai L, Cheng W, Wan M, Li Z, Chen J, Li H, Kong E, Wang K, Lu K, Qi S. ALS-linked C9orf72-SMCR8 complex is a negative regulator of primary ciliogenesis. Proc Natl Acad Sci U S A 2023; 120:e2220496120. [PMID: 38064514 PMCID: PMC10723147 DOI: 10.1073/pnas.2220496120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 10/25/2023] [Indexed: 12/17/2023] Open
Abstract
Massive GGGGCC (G4C2) repeat expansion in C9orf72 and the resulting loss of C9orf72 function are the key features of ~50% of inherited amyotrophic lateral sclerosis and frontotemporal dementia cases. However, the biological function of C9orf72 remains unclear. We previously found that C9orf72 can form a stable GTPase activating protein (GAP) complex with SMCR8 (Smith-Magenis chromosome region 8). Herein, we report that the C9orf72-SMCR8 complex is a major negative regulator of primary ciliogenesis, abnormalities in which lead to ciliopathies. Mechanistically, the C9orf72-SMCR8 complex suppresses the primary cilium as a RAB8A GAP. Moreover, based on biochemical analysis, we found that C9orf72 is the RAB8A binding subunit and that SMCR8 is the GAP subunit in the complex. We further found that the C9orf72-SMCR8 complex suppressed the primary cilium in multiple tissues from mice, including but not limited to the brain, kidney, and spleen. Importantly, cells with C9orf72 or SMCR8 knocked out were more sensitive to hedgehog signaling. These results reveal the unexpected impact of C9orf72 on primary ciliogenesis and elucidate the pathogenesis of diseases caused by the loss of C9orf72 function.
Collapse
Affiliation(s)
- Dan Tang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Kaixuan Zheng
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Jiangli Zhu
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang453000, People’s Republic of China
| | - Xi Jin
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Hui Bao
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Lan Jiang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Huihui Li
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Yichang Wang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| | - Ying Lu
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Jiaming Liu
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Hang Liu
- Division of Life Science, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou511458, People’s Republic of China
- HKUST-Shenzhen Research Institute, Nanshan, Shenzhen518057, People’s Republic of China
| | - Chengbing Tang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Shijian Feng
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Xiuju Dong
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Liangting Xu
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Yike Yin
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Shangyu Dang
- Division of Life Science, Center of Systems Biology and Human Health, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou511458, People’s Republic of China
- HKUST-Shenzhen Research Institute, Nanshan, Shenzhen518057, People’s Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| | - Haiyan Ren
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Biao Dong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
- Sichuan Real & Best Biotech Co., Ltd., Chengdu610219, People’s Republic of China
| | - Lunzhi Dai
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Wei Cheng
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Meihua Wan
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Zhonghan Li
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Jing Chen
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Hong Li
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Eryan Kong
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang453000, People’s Republic of China
| | - Kunjie Wang
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Kefeng Lu
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
| | - Shiqian Qi
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu610041, People’s Republic of China
- National Health Commission Key Lab of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| |
Collapse
|
24
|
McCallister TX, Lim CKW, Terpstra WM, Alejandra Zeballos C M, Zhang S, Powell JE, Gaj T. A high-fidelity CRISPR-Cas13 system improves abnormalities associated with C9ORF72-linked ALS/FTD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571328. [PMID: 38168370 PMCID: PMC10760048 DOI: 10.1101/2023.12.12.571328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
An abnormal expansion of a GGGGCC hexanucleotide repeat in the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two debilitating neurodegenerative disorders driven in part by gain-of-function mechanisms involving transcribed forms of the repeat expansion. By utilizing a Cas13 variant with reduced collateral effects, we developed a high-fidelity RNA-targeting CRISPR-based system for C9ORF72-linked ALS/FTD. When delivered to the brain of a transgenic rodent model, this Cas13-based platform effectively curbed the expression of the GGGGCC repeat-containing RNA without affecting normal C9ORF72 levels, which in turn decreased the formation of RNA foci and reversed transcriptional deficits. This high-fidelity Cas13 variant possessed improved transcriptome-wide specificity compared to its native form and mediated efficient targeting in motor neuron-like cells derived from a patient with ALS. Our results lay the foundation for the implementation of RNA-targeting CRISPR technologies for C9ORF72-linked ALS/FTD.
Collapse
Affiliation(s)
- Tristan X. McCallister
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Colin K. W. Lim
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - William M. Terpstra
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - M. Alejandra Zeballos C
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Sijia Zhang
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Jackson E. Powell
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Thomas Gaj
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
25
|
Sattler R, Traynor BJ, Robertson J, Van Den Bosch L, Barmada SJ, Svendsen CN, Disney MD, Gendron TF, Wong PC, Turner MR, Boxer A, Babu S, Benatar M, Kurnellas M, Rohrer JD, Donnelly CJ, Bustos LM, Van Keuren-Jensen K, Dacks PA, Sabbagh MN. Roadmap for C9ORF72 in Frontotemporal Dementia and Amyotrophic Lateral Sclerosis: Report on the C9ORF72 FTD/ALS Summit. Neurol Ther 2023; 12:1821-1843. [PMID: 37847372 PMCID: PMC10630271 DOI: 10.1007/s40120-023-00548-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/14/2023] [Indexed: 10/18/2023] Open
Abstract
A summit held March 2023 in Scottsdale, Arizona (USA) focused on the intronic hexanucleotide expansion in the C9ORF72 gene and its relevance in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS; C9ORF72-FTD/ALS). The goal of this summit was to connect basic scientists, clinical researchers, drug developers, and individuals affected by C9ORF72-FTD/ALS to evaluate how collaborative efforts across the FTD-ALS disease spectrum might break down existing disease silos. Presentations and discussions covered recent discoveries in C9ORF72-FTD/ALS disease mechanisms, availability of disease biomarkers and recent advances in therapeutic development, and clinical trial design for prevention and treatment for individuals affected by C9ORF72-FTD/ALS and asymptomatic pathological expansion carriers. The C9ORF72-associated hexanucleotide repeat expansion is an important locus for both ALS and FTD. C9ORF72-FTD/ALS may be characterized by loss of function of the C9ORF72 protein and toxic gain of functions caused by both dipeptide repeat (DPR) proteins and hexanucleotide repeat RNA. C9ORF72-FTD/ALS therapeutic strategies discussed at the summit included the use of antisense oligonucleotides, adeno-associated virus (AAV)-mediated gene silencing and gene delivery, and engineered small molecules targeting RNA structures associated with the C9ORF72 expansion. Neurofilament light chain, DPR proteins, and transactive response (TAR) DNA-binding protein 43 (TDP-43)-associated molecular changes were presented as biomarker candidates. Similarly, brain imaging modalities (i.e., magnetic resonance imaging [MRI] and positron emission tomography [PET]) measuring structural, functional, and metabolic changes were discussed as important tools to monitor individuals affected with C9ORF72-FTD/ALS, at both pre-symptomatic and symptomatic disease stages. Finally, summit attendees evaluated current clinical trial designs available for FTD or ALS patients and concluded that therapeutics relevant to FTD/ALS patients, such as those specifically targeting C9ORF72, may need to be tested with composite endpoints covering clinical symptoms of both FTD and ALS. The latter will require novel clinical trial designs to be inclusive of all patient subgroups spanning the FTD/ALS spectrum.
Collapse
Affiliation(s)
- Rita Sattler
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ludo Van Den Bosch
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology and KU Leuven, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
| | - Sami J Barmada
- Department of Neurology, Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Matthew D Disney
- Department of Chemistry, The Herbert Wertheim UF-Scripps Institute for Biomedical Research and Innovation, The Scripps Research Institute, Jupiter, FL, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Philip C Wong
- Departments of Pathology and Neuroscience, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adam Boxer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of San Francisco, San Francisco, CA, USA
| | - Suma Babu
- Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital-Harvard Medical School, Boston, MA, USA
| | - Michael Benatar
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33129, USA
| | | | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher J Donnelly
- LiveLikeLou Center for ALS Research, Brain Institute, University of Pittsburgh, Pittsburgh, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lynette M Bustos
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA
| | | | - Penny A Dacks
- The Association for Frontotemporal Degeneration and FTD Disorders Registry, King of Prussia, PA, USA
| | - Marwan N Sabbagh
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| |
Collapse
|
26
|
Pelaez MC, Desmeules A, Gelon PA, Glasson B, Marcadet L, Rodgers A, Phaneuf D, Pozzi S, Dutchak PA, Julien JP, Sephton CF. Neuronal dysfunction caused by FUSR521G promotes ALS-associated phenotypes that are attenuated by NF-κB inhibition. Acta Neuropathol Commun 2023; 11:182. [PMID: 37974279 PMCID: PMC10652582 DOI: 10.1186/s40478-023-01671-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/09/2023] [Indexed: 11/19/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are related neurodegenerative diseases that belong to a common disease spectrum based on overlapping clinical, pathological and genetic evidence. Early pathological changes to the morphology and synapses of affected neuron populations in ALS/FTD suggest a common underlying mechanism of disease that requires further investigation. Fused in sarcoma (FUS) is a DNA/RNA-binding protein with known genetic and pathological links to ALS/FTD. Expression of ALS-linked FUS mutants in mice causes cognitive and motor defects, which correlate with loss of motor neuron dendritic branching and synapses, in addition to other pathological features of ALS/FTD. The role of ALS-linked FUS mutants in causing ALS/FTD-associated disease phenotypes is well established, but there are significant gaps in our understanding of the cell-autonomous role of FUS in promoting structural changes to motor neurons, and how these changes relate to disease progression. Here we generated a neuron-specific FUS-transgenic mouse model expressing the ALS-linked human FUSR521G variant, hFUSR521G/Syn1, to investigate the cell-autonomous role of FUSR521G in causing loss of dendritic branching and synapses of motor neurons, and to understand how these changes relate to ALS-associated phenotypes. Longitudinal analysis of mice revealed that cognitive impairments in juvenile hFUSR521G/Syn1 mice coincide with reduced dendritic branching of cortical motor neurons in the absence of motor impairments or changes in the neuromorphology of spinal motor neurons. Motor impairments and dendritic attrition of spinal motor neurons developed later in aged hFUSR521G/Syn1 mice, along with FUS cytoplasmic mislocalisation, mitochondrial abnormalities and glial activation. Neuroinflammation promotes neuronal dysfunction and drives disease progression in ALS/FTD. The therapeutic effects of inhibiting the pro-inflammatory nuclear factor kappa B (NF-κB) pathway with an analog of Withaferin A, IMS-088, were assessed in symptomatic hFUSR521G/Syn1 mice and were found to improve cognitive and motor function, increase dendritic branches and synapses of motor neurons, and attenuate other ALS/FTD-associated pathological features. Treatment of primary cortical neurons expressing FUSR521G with IMS-088 promoted the restoration of dendritic mitochondrial numbers and mitochondrial activity to wild-type levels, suggesting that inhibition of NF-κB permits the restoration of mitochondrial stasis in our models. Collectively, this work demonstrates that FUSR521G has a cell-autonomous role in causing early pathological changes to dendritic and synaptic structures of motor neurons, and that these changes precede motor defects and other well-known pathological features of ALS/FTD. Finally, these findings provide further support that modulation of the NF-κB pathway in ALS/FTD is an important therapeutic approach to attenuate disease.
Collapse
Affiliation(s)
- Mari Carmen Pelaez
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Antoine Desmeules
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Pauline A Gelon
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Bastien Glasson
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Laetitia Marcadet
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Alicia Rodgers
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Daniel Phaneuf
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Silvia Pozzi
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Paul A Dutchak
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
27
|
Litwiniuk A, Juszczak GR, Stankiewicz AM, Urbańska K. The role of glial autophagy in Alzheimer's disease. Mol Psychiatry 2023; 28:4528-4539. [PMID: 37679471 DOI: 10.1038/s41380-023-02242-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
Although Alzheimer's disease is the most pervasive neurodegenerative disorder, the mechanism underlying its development is still not precisely understood. Available data indicate that pathophysiology of this disease may involve impaired autophagy in glial cells. The dysfunction is manifested as reduced ability of astrocytes and microglia to clear abnormal protein aggregates. Consequently, excessive accumulation of amyloid beta plaques and neurofibrillary tangles activates microglia and astrocytes leading to decreased number of mature myelinated oligodendrocytes and death of neurons. These pathologic effects of autophagy dysfunction can be rescued by pharmacological activation of autophagy. Therefore, a deeper understanding of the molecular mechanisms involved in autophagy dysfunction in glial cells in Alzheimer's disease may lead to the development of new therapeutic strategies. However, such strategies need to take into consideration differences in regulation of autophagy in different types of neuroglia.
Collapse
Affiliation(s)
- Anna Litwiniuk
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Mazovia, Poland
| | - Grzegorz Roman Juszczak
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, Mazovia, Poland
| | - Adrian Mateusz Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, Mazovia, Poland.
| | - Kaja Urbańska
- Department of Morphological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Mazovia, Poland.
| |
Collapse
|
28
|
Yeo S, Jang J, Jung HJ, Lee H, Choe Y. Primary cilia-mediated regulation of microglial secretion in Alzheimer's disease. Front Mol Biosci 2023; 10:1250335. [PMID: 37942288 PMCID: PMC10627801 DOI: 10.3389/fmolb.2023.1250335] [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: 06/30/2023] [Accepted: 09/28/2023] [Indexed: 11/10/2023] Open
Abstract
Alzheimer's disease (AD) is a brain disorder manifested by a gradual decline in cognitive function due to the accumulation of extracellular amyloid plaques, disruptions in neuronal substance transport, and the degeneration of neurons. In affected neurons, incomplete clearance of toxic proteins by neighboring microglia leads to irreversible brain inflammation, for which cellular signaling is poorly understood. Through single-cell transcriptomic analysis, we discovered distinct regional differences in the ability of microglia to clear damaged neurites. Specifically, microglia in the septal region of wild type mice exhibited a transcriptomic signature resembling disease-associated microglia (DAM). These lateral septum (LS)-enriched microglia were associated with dense axonal bundles originating from the hippocampus. Further transcriptomic and proteomic approaches revealed that primary cilia, small hair-like structures found on cells, played a role in the regulation of microglial secretory function. Notably, primary cilia were transiently observed in microglia, and their presence was significantly reduced in microglia from AD mice. We observed significant changes in the secretion and proteomic profiles of the secretome after inhibiting the primary cilia gene intraflagellar transport particle 88 (Ift88) in microglia. Intriguingly, inhibiting primary cilia in the septal microglia of AD mice resulted in the expansion of extracellular amyloid plaques and damage to adjacent neurites. These results indicate that DAM-like microglia are present in the LS, a critical target region for hippocampal nerve bundles, and that the primary ciliary signaling system regulates microglial secretion, affecting extracellular proteostasis. Age-related primary ciliopathy probably contributes to the selective sensitivity of microglia, thereby exacerbating AD. Targeting the primary ciliary signaling system could therefore be a viable strategy for modulating neuroimmune responses in AD treatments.
Collapse
Affiliation(s)
- Seungeun Yeo
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jaemyung Jang
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyun Jin Jung
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hyeyoung Lee
- Division of Applied Bioengineering, Dong-eui University, Busan, Republic of Korea
| | - Youngshik Choe
- Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
29
|
Xie M, Pallegar PN, Parusel S, Nguyen AT, Wu LJ. Regulation of cortical hyperexcitability in amyotrophic lateral sclerosis: focusing on glial mechanisms. Mol Neurodegener 2023; 18:75. [PMID: 37858176 PMCID: PMC10585818 DOI: 10.1186/s13024-023-00665-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder characterized by the loss of both upper and lower motor neurons, resulting in muscle weakness, atrophy, paralysis, and eventually death. Motor cortical hyperexcitability is a common phenomenon observed at the presymptomatic stage of ALS. Both cell-autonomous (the intrinsic properties of motor neurons) and non-cell-autonomous mechanisms (cells other than motor neurons) are believed to contribute to cortical hyperexcitability. Decoding the pathological relevance of these dynamic changes in motor neurons and glial cells has remained a major challenge. This review summarizes the evidence of cortical hyperexcitability from both clinical and preclinical research, as well as the underlying mechanisms. We discuss the potential role of glial cells, particularly microglia, in regulating abnormal neuronal activity during the disease progression. Identifying early changes such as neuronal hyperexcitability in the motor system may provide new insights for earlier diagnosis of ALS and reveal novel targets to halt the disease progression.
Collapse
Affiliation(s)
- Manling Xie
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Praveen N Pallegar
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | - Sebastian Parusel
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Aivi T Nguyen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
30
|
Pérez-Acuña D, Shin SJ, Rhee KH, Kim SJ, Lee SJ. α-Synuclein propagation leads to synaptic abnormalities in the cortex through microglial synapse phagocytosis. Mol Brain 2023; 16:72. [PMID: 37848910 PMCID: PMC10580656 DOI: 10.1186/s13041-023-01059-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023] Open
Abstract
The major neuropathologic feature of Parkinson's disease is the presence of widespread intracellular inclusions of α-synuclein known as Lewy bodies. Evidence suggests that these misfolded protein inclusions spread through the brain with disease progression. Changes in synaptic function precede neurodegeneration, and this extracellular α-synuclein can affect synaptic transmission. However, whether and how the spreading of α-synuclein aggregates modulates synaptic function before neuronal loss remains unknown. In the present study, we investigated the effect of intrastriatal injection of α-synuclein preformed fibrils (PFFs) on synaptic activity in the somatosensory cortex using a combination of whole-cell patch-clamp electrophysiology, histology, and Golgi-Cox staining. Intrastriatal PFF injection was followed by formation of phosphorylated α-synuclein inclusions in layer 5 of the somatosensory cortex, leading to a decrease in synapse density, dendritic spines, and spontaneous excitatory post-synaptic currents, without apparent neuronal loss. Additionally, three-dimensional reconstruction of microglia using confocal imaging showed an increase in the engulfment of synapses. Collectively, our data indicate that propagation of α-synuclein through neural networks causes abnormalities in synaptic structure and dynamics prior to neuronal loss.
Collapse
Affiliation(s)
- Dayana Pérez-Acuña
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Republic of Korea
| | - Soo Jean Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Republic of Korea
| | - Ka Hyun Rhee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Republic of Korea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sang Jeong Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University, College of Medicine, Seoul, 03080, Republic of Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-Ro, Jongro-Gu, Seoul, 03080, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- , Neuramedy, Seoul, 04796, Republic of Korea.
| |
Collapse
|
31
|
You J, Youssef MMM, Santos JR, Lee J, Park J. Microglia and Astrocytes in Amyotrophic Lateral Sclerosis: Disease-Associated States, Pathological Roles, and Therapeutic Potential. BIOLOGY 2023; 12:1307. [PMID: 37887017 PMCID: PMC10603852 DOI: 10.3390/biology12101307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Microglial and astrocytic reactivity is a prominent feature of amyotrophic lateral sclerosis (ALS). Microglia and astrocytes have been increasingly appreciated to play pivotal roles in disease pathogenesis. These cells can adopt distinct states characterized by a specific molecular profile or function depending on the different contexts of development, health, aging, and disease. Accumulating evidence from ALS rodent and cell models has demonstrated neuroprotective and neurotoxic functions from microglia and astrocytes. In this review, we focused on the recent advancements of knowledge in microglial and astrocytic states and nomenclature, the landmark discoveries demonstrating a clear contribution of microglia and astrocytes to ALS pathogenesis, and novel therapeutic candidates leveraging these cells that are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Justin You
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.); (M.M.M.Y.); (J.R.S.); (J.L.)
| | - Mohieldin M. M. Youssef
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.); (M.M.M.Y.); (J.R.S.); (J.L.)
| | - Jhune Rizsan Santos
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.); (M.M.M.Y.); (J.R.S.); (J.L.)
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Jooyun Lee
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.); (M.M.M.Y.); (J.R.S.); (J.L.)
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Jeehye Park
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.); (M.M.M.Y.); (J.R.S.); (J.L.)
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
32
|
Vahsen BF, Nalluru S, Morgan GR, Farrimond L, Carroll E, Xu Y, Cramb KML, Amein B, Scaber J, Katsikoudi A, Candalija A, Carcolé M, Dafinca R, Isaacs AM, Wade-Martins R, Gray E, Turner MR, Cowley SA, Talbot K. C9orf72-ALS human iPSC microglia are pro-inflammatory and toxic to co-cultured motor neurons via MMP9. Nat Commun 2023; 14:5898. [PMID: 37736756 PMCID: PMC10517114 DOI: 10.1038/s41467-023-41603-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive motor neuron loss, with additional pathophysiological involvement of non-neuronal cells such as microglia. The commonest ALS-associated genetic variant is a hexanucleotide repeat expansion (HRE) mutation in C9orf72. Here, we study its consequences for microglial function using human iPSC-derived microglia. By RNA-sequencing, we identify enrichment of pathways associated with immune cell activation and cyto-/chemokines in C9orf72 HRE mutant microglia versus healthy controls, most prominently after LPS priming. Specifically, LPS-primed C9orf72 HRE mutant microglia show consistently increased expression and release of matrix metalloproteinase-9 (MMP9). LPS-primed C9orf72 HRE mutant microglia are toxic to co-cultured healthy motor neurons, which is ameliorated by concomitant application of an MMP9 inhibitor. Finally, we identify release of dipeptidyl peptidase-4 (DPP4) as a marker for MMP9-dependent microglial dysregulation in co-culture. These results demonstrate cellular dysfunction of C9orf72 HRE mutant microglia, and a non-cell-autonomous role in driving C9orf72-ALS pathophysiology in motor neurons through MMP9 signaling.
Collapse
Affiliation(s)
- Björn F Vahsen
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Sumedha Nalluru
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Georgia R Morgan
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Lucy Farrimond
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Emily Carroll
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Yinyan Xu
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
- Chinese Academy of Medical Sciences (CAMS), CAMS Oxford Institute (COI), Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Kaitlyn M L Cramb
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QX, UK
| | - Benazir Amein
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Jakub Scaber
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Antigoni Katsikoudi
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
- Molecular Neurodegeneration Research Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Ana Candalija
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WCIN 3BG, UK
| | - Ruxandra Dafinca
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, WCIN 3BG, UK
| | - Richard Wade-Martins
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QX, UK
| | - Elizabeth Gray
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Martin R Turner
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Sally A Cowley
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| | - Kevin Talbot
- Oxford Motor Neuron Disease Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK.
| |
Collapse
|
33
|
Gao C, Jiang J, Tan Y, Chen S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct Target Ther 2023; 8:359. [PMID: 37735487 PMCID: PMC10514343 DOI: 10.1038/s41392-023-01588-0] [Citation(s) in RCA: 209] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/11/2023] [Accepted: 08/03/2023] [Indexed: 09/23/2023] Open
Abstract
Microglia activation is observed in various neurodegenerative diseases. Recent advances in single-cell technologies have revealed that these reactive microglia were with high spatial and temporal heterogeneity. Some identified microglia in specific states correlate with pathological hallmarks and are associated with specific functions. Microglia both exert protective function by phagocytosing and clearing pathological protein aggregates and play detrimental roles due to excessive uptake of protein aggregates, which would lead to microglial phagocytic ability impairment, neuroinflammation, and eventually neurodegeneration. In addition, peripheral immune cells infiltration shapes microglia into a pro-inflammatory phenotype and accelerates disease progression. Microglia also act as a mobile vehicle to propagate protein aggregates. Extracellular vesicles released from microglia and autophagy impairment in microglia all contribute to pathological progression and neurodegeneration. Thus, enhancing microglial phagocytosis, reducing microglial-mediated neuroinflammation, inhibiting microglial exosome synthesis and secretion, and promoting microglial conversion into a protective phenotype are considered to be promising strategies for the therapy of neurodegenerative diseases. Here we comprehensively review the biology of microglia and the roles of microglia in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, corticobasal degeneration, dementia with Lewy bodies and Huntington's disease. We also summarize the possible microglia-targeted interventions and treatments against neurodegenerative diseases with preclinical and clinical evidence in cell experiments, animal studies, and clinical trials.
Collapse
Affiliation(s)
- Chao Gao
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Jingwen Jiang
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yuyan Tan
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
34
|
Li J, Jaiswal MK, Chien JF, Kozlenkov A, Jung J, Zhou P, Gardashli M, Pregent LJ, Engelberg-Cook E, Dickson DW, Belzil VV, Mukamel EA, Dracheva S. Divergent single cell transcriptome and epigenome alterations in ALS and FTD patients with C9orf72 mutation. Nat Commun 2023; 14:5714. [PMID: 37714849 PMCID: PMC10504300 DOI: 10.1038/s41467-023-41033-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 08/21/2023] [Indexed: 09/17/2023] Open
Abstract
A repeat expansion in the C9orf72 (C9) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we investigate single nucleus transcriptomics (snRNA-seq) and epigenomics (snATAC-seq) in postmortem motor and frontal cortices from C9-ALS, C9-FTD, and control donors. C9-ALS donors present pervasive alterations of gene expression with concordant changes in chromatin accessibility and histone modifications. The greatest alterations occur in upper and deep layer excitatory neurons, as well as in astrocytes. In neurons, the changes imply an increase in proteostasis, metabolism, and protein expression pathways, alongside a decrease in neuronal function. In astrocytes, the alterations suggest activation and structural remodeling. Conversely, C9-FTD donors have fewer high-quality neuronal nuclei in the frontal cortex and numerous gene expression changes in glial cells. These findings highlight a context-dependent molecular disruption in C9-ALS and C9-FTD, indicating unique effects across cell types, brain regions, and diseases.
Collapse
Affiliation(s)
- Junhao Li
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, 92037, US
| | - Manoj K Jaiswal
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Jo-Fan Chien
- Department of Physics, University of California San Diego, La Jolla, CA, 92037, US
| | - Alexey Kozlenkov
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Jinyoung Jung
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Ping Zhou
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | | | - Luc J Pregent
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, US
| | | | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, US
| | | | - Eran A Mukamel
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, 92037, US.
| | - Stella Dracheva
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US.
- Research & Development and VISN2 MIREC, James J, Peters VA Medical Center, Bronx, NY, 10468, US.
| |
Collapse
|
35
|
Guan PP, Ge TQ, Wang P. As a Potential Therapeutic Target, C1q Induces Synapse Loss Via Inflammasome-activating Apoptotic and Mitochondria Impairment Mechanisms in Alzheimer's Disease. J Neuroimmune Pharmacol 2023; 18:267-284. [PMID: 37386257 DOI: 10.1007/s11481-023-10076-9] [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: 08/11/2022] [Accepted: 06/16/2023] [Indexed: 07/01/2023]
Abstract
C1q, the initiator of the classical pathway of the complement system, is activated during Alzheimer's disease (AD) development and progression and is especially associated with the production and deposition of β-amyloid protein (Aβ) and phosphorylated tau in β-amyloid plaques (APs) and neurofibrillary tangles (NFTs). Activation of C1q is responsible for induction of synapse loss, leading to neurodegeneration in AD. Mechanistically, C1q could activate glial cells, which results in the loss of synapses via regulation of synapse pruning and phagocytosis in AD. In addition, C1q induces neuroinflammation by inducing proinflammatory cytokine secretion, which is partially mediated by inflammasome activation. Activation of inflammasomes might mediate the effects of C1q on induction of synapse apoptosis. On the other hand, activation of C1q impairs mitochondria, which hinders the renovation and regeneration of synapses. All these actions of C1q contribute to the loss of synapses during neurodegeneration in AD. Therefore, pharmacological, or genetic interventions targeting C1q may provide potential therapeutic strategies for combating AD.
Collapse
Affiliation(s)
- Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China
| | - Tong-Qi Ge
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China.
| |
Collapse
|
36
|
De Marchi F, Tondo G, Corrado L, Menegon F, Aprile D, Anselmi M, D’Alfonso S, Comi C, Mazzini L. Neuroinflammatory Pathways in the ALS-FTD Continuum: A Focus on Genetic Variants. Genes (Basel) 2023; 14:1658. [PMID: 37628709 PMCID: PMC10454262 DOI: 10.3390/genes14081658] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal dementia (FDT) are progressive neurodegenerative disorders that, in several cases, overlap in clinical presentation, and genetic and pathological disease mechanisms. About 10-15% of ALS cases and up to 40% of FTD are familial, usually with dominant traits. ALS and FTD, in several cases, share common gene mutations, such as in C9ORF72, TARDBP, SQSTM-1, FUS, VCP, CHCHD10, and TBK-1. Also, several mechanisms are involved in ALS and FTD pathogenesis, such as protein misfolding, oxidative stress, and impaired axonal transport. In addition, neuroinflammation and neuroinflammatory cells, such as astrocytes, oligodendrocytes, microglia, and lymphocytes and, overall, the cellular microenvironment, have been proposed as pivotal players in the pathogenesis the ALS-FTD spectrum disorders. This review overviews the current evidence regarding neuroinflammatory markers in the ALS/FTD continuum, focusing on the neuroinflammatory pathways involved in the genetic cases, moving from post-mortem reports to in vivo biofluid and neuroimaging data. We further discuss the potential link between genetic and autoimmune disorders and potential therapeutic implications.
Collapse
Affiliation(s)
- Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy; (G.T.); (D.A.); (C.C.)
| | - Lucia Corrado
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy; (L.C.); (S.D.)
| | - Federico Menegon
- Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (F.M.); (M.A.)
| | - Davide Aprile
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy; (G.T.); (D.A.); (C.C.)
| | - Matteo Anselmi
- Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (F.M.); (M.A.)
| | - Sandra D’Alfonso
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy; (L.C.); (S.D.)
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy; (G.T.); (D.A.); (C.C.)
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
| |
Collapse
|
37
|
Todd BP, Luo Z, Gilkes N, Chimenti MS, Peterson Z, Mix MR, Harty JT, Nickl-Jockschat T, Ferguson PJ, Bassuk AG, Newell EA. Selective neuroimmune modulation by type I interferon drives neuropathology and neurologic dysfunction following traumatic brain injury. Acta Neuropathol Commun 2023; 11:134. [PMID: 37596685 PMCID: PMC10436463 DOI: 10.1186/s40478-023-01635-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/05/2023] [Indexed: 08/20/2023] Open
Abstract
Accumulating evidence suggests that type I interferon (IFN-I) signaling is a key contributor to immune cell-mediated neuropathology in neurodegenerative diseases. Recently, we demonstrated a robust upregulation of type I interferon-stimulated genes in microglia and astrocytes following experimental traumatic brain injury (TBI). The specific molecular and cellular mechanisms by which IFN-I signaling impacts the neuroimmune response and neuropathology following TBI remains unknown. Using the lateral fluid percussion injury model (FPI) in adult male mice, we demonstrated that IFN α/β receptor (IFNAR) deficiency resulted in selective and sustained blockade of type I interferon-stimulated genes following TBI as well as decreased microgliosis and monocyte infiltration. Molecular alteration of reactive microglia also occurred with diminished expression of genes needed for MHC class I antigen processing and presentation following TBI. This was associated with decreased accumulation of cytotoxic T cells in the brain. The IFNAR-dependent modulation of the neuroimmune response was accompanied by protection from secondary neuronal death, white matter disruption, and neurobehavioral dysfunction. These data support further efforts to leverage the IFN-I pathway for novel, targeted therapy of TBI.
Collapse
Affiliation(s)
- Brittany P Todd
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, USA
| | - Zili Luo
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Noah Gilkes
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Michael S Chimenti
- Bioinformatics Division, Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA
| | - Zeru Peterson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Madison R Mix
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - John T Harty
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Thomas Nickl-Jockschat
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Polly J Ferguson
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Elizabeth A Newell
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA.
| |
Collapse
|
38
|
Li Y, Dou X, Liu J, Xiao Y, Zhang Z, Hayes L, Wu R, Fu X, Ye Y, Yang B, Ostrow LW, He C, Sun S. Globally reduced N 6-methyladenosine (m 6A) in C9ORF72-ALS/FTD dysregulates RNA metabolism and contributes to neurodegeneration. Nat Neurosci 2023; 26:1328-1338. [PMID: 37365312 PMCID: PMC11361766 DOI: 10.1038/s41593-023-01374-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
Repeat expansion in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we show that N6-methyladenosine (m6A), the most prevalent internal mRNA modification, is downregulated in C9ORF72-ALS/FTD patient-derived induced pluripotent stem cell (iPSC)-differentiated neurons and postmortem brain tissues. The global m6A hypomethylation leads to transcriptome-wide mRNA stabilization and upregulated gene expression, particularly for genes involved in synaptic activity and neuronal function. Moreover, the m6A modification in the C9ORF72 intron sequence upstream of the expanded repeats enhances RNA decay via the nuclear reader YTHDC1, and the antisense RNA repeats can also be regulated through m6A modification. The m6A reduction increases the accumulation of repeat RNAs and the encoded poly-dipeptides, contributing to disease pathogenesis. We further demonstrate that, by elevating m6A methylation, we could significantly reduce repeat RNA levels from both strands and the derived poly-dipeptides, rescue global mRNA homeostasis and improve survival of C9ORF72-ALS/FTD patient iPSC-derived neurons.
Collapse
Affiliation(s)
- Yini Li
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaoyang Dou
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, Chicago, IL, USA
| | - Jun Liu
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, Chicago, IL, USA
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yu Xiao
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, Chicago, IL, USA
| | - Zhe Zhang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lindsey Hayes
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rong Wu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiujuan Fu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yingzhi Ye
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cellular and Molecular Physiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bing Yang
- Laboratory of Cellular and Developmental Biology, NIDDK Intramural Research Program, Bethesda, MD, USA
| | - Lyle W Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Chuan He
- Department of Chemistry and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA.
- Howard Hughes Medical Institute, Chicago, IL, USA.
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA.
| | - Shuying Sun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
39
|
Ye L, Shu S, Jia J, Sun M, Xu S, Bao X, Bian H, Liu Y, Zhang M, Zhu X, Bai F, Xu Y. Absent in melanoma 2 mediates aging-related cognitive dysfunction by acting on complement-dependent microglial phagocytosis. Aging Cell 2023; 22:e13860. [PMID: 37177836 PMCID: PMC10352562 DOI: 10.1111/acel.13860] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023] Open
Abstract
Pattern separation (PS) dysfunction is a type of cognitive impairment that presents early during the aging process, and this deficit has been attributed to structural and functional alterations in the dentate gyrus (DG) of the hippocampus. Absent in melanoma 2 (AIM2) is an essential component of the inflammasome. However, whether AIM2 plays a role in aging-associated cognitive dysfunction remains unclear. Here, we found that PS function was impaired in aging mice and was accompanied by marked synaptic loss and increased expression of AIM2 in the DG. Subsequently, we used an AIM2 overexpression virus and mice with AIM2 deletion to investigate the role of AIM2 in regulating PS function and synaptic plasticity and the mechanisms involved. Our study revealed that AIM2 regulates microglial activation during synaptic pruning in the DG region via the complement pathway, leading to impaired synaptic plasticity and PS function in aging mice. These results suggest a critical role for AIM2 in regulating synaptic plasticity and PS function and provide a new direction for ameliorating aging-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Lei Ye
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Shu Shu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Junqiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Huijie Bian
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Yi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Feng Bai
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjingChina
- Jiangsu Provincial Key Discipline of NeurologyNanjingChina
- Nanjing Neurology Medical CenterNanjingChina
- Nanjing Neuropsychiatry Clinic Medical CenterNanjingChina
| |
Collapse
|
40
|
Ghaffari LT, Trotti D, Haeusler AR. Differential response of C9orf72 transcripts following neuronal depolarization. iScience 2023; 26:106959. [PMID: 37332610 PMCID: PMC10272498 DOI: 10.1016/j.isci.2023.106959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
The (G4C2)n nucleotide repeat expansion (NRE) mutation in C9orf72 is the most common genetic cause of ALS and FTD. The biological functions of C9orf72 are becoming understood, but it is unclear if this gene is regulated in a neural-specific manner. Neuronal activity is a crucial modifier of biological processes in health and neurodegenerative disease contexts. Here, we show that prolonged membrane depolarization in healthy human iPSC-cortical neurons leads to a significant downregulation of a transcript variant 3 (V3) of C9orf72, with a concomitant increase in variant 2 (V2), which leads to total C9orf72 RNA transcript levels remaining unchanged. However, the same response is not observed in cortical neurons derived from patients with the C9-NRE mutation. These findings reveal the impact of depolarization on C9orf72 transcripts, and how this response diverges in C9-NRE-carriers, which may have important implications in the underlying unique clinical associations of C9-NRE transcripts and disease pathogenesis.
Collapse
Affiliation(s)
- Layla T. Ghaffari
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Aaron R. Haeusler
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
41
|
Todd BP, Luo Z, Gilkes N, Chimenti MS, Peterson Z, Mix M, Harty JT, Nickl-Jockschat T, Ferguson PJ, Bassuk AG, Newell EA. Selective neuroimmune modulation by type I interferon drives neuropathology and neurologic dysfunction following traumatic brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543774. [PMID: 37333385 PMCID: PMC10274693 DOI: 10.1101/2023.06.06.543774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Accumulating evidence suggests that type I interferon (IFN-I) signaling is a key contributor to immune cell-mediated neuropathology in neurodegenerative diseases. Recently, we demonstrated a robust upregulation of type I interferon-stimulated genes in microglia and astrocytes following experimental traumatic brain injury (TBI). The specific molecular and cellular mechanisms by which IFN-I signaling impacts the neuroimmune response and neuropathology following TBI remains unknown. Using the lateral fluid percussion injury model (FPI) in adult male mice, we demonstrated that IFN α/β receptor (IFNAR) deficiency resulted in selective and sustained blockade of type I interferon-stimulated genes following TBI as well as decreased microgliosis and monocyte infiltration. Phenotypic alteration of reactive microglia also occurred with diminished expression of molecules needed for MHC class I antigen processing and presentation following TBI. This was associated with decreased accumulation of cytotoxic T cells in the brain. The IFNAR-dependent modulation of the neuroimmune response was accompanied by protection from secondary neuronal death, white matter disruption, and neurobehavioral dysfunction. These data support further efforts to leverage the IFN-I pathway for novel, targeted therapy of TBI.
Collapse
|
42
|
Lorenzini I, Alsop E, Levy J, Gittings LM, Lall D, Rabichow BE, Moore S, Pevey R, Bustos LM, Burciu C, Bhatia D, Singer M, Saul J, McQuade A, Tzioras M, Mota TA, Logemann A, Rose J, Almeida S, Gao FB, Marks M, Donnelly CJ, Hutchins E, Hung ST, Ichida J, Bowser R, Spires-Jones T, Blurton-Jones M, Gendron TF, Baloh RH, Van Keuren-Jensen K, Sattler R. Moderate intrinsic phenotypic alterations in C9orf72 ALS/FTD iPSC-microglia despite the presence of C9orf72 pathological features. Front Cell Neurosci 2023; 17:1179796. [PMID: 37346371 PMCID: PMC10279871 DOI: 10.3389/fncel.2023.1179796] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
While motor and cortical neurons are affected in C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD), it remains largely unknown if and how non-neuronal cells induce or exacerbate neuronal damage. We differentiated C9orf72 ALS/FTD patient-derived induced pluripotent stem cells into microglia (iPSC-MG) and examined their intrinsic phenotypes. Similar to iPSC motor neurons, C9orf72 ALS/FTD iPSC-MG mono-cultures form G4C2 repeat RNA foci, exhibit reduced C9orf72 protein levels, and generate dipeptide repeat proteins. Healthy control and C9orf72 ALS/FTD iPSC-MG equally express microglial specific genes and perform microglial functions, including inflammatory cytokine release and phagocytosis of extracellular cargos, such as synthetic amyloid beta peptides and healthy human brain synaptoneurosomes. RNA sequencing analysis revealed select transcriptional changes of genes associated with neuroinflammation or neurodegeneration in diseased microglia yet no significant differentially expressed microglial-enriched genes. Moderate molecular and functional differences were observed in C9orf72 iPSC-MG mono-cultures despite the presence of C9orf72 pathological features suggesting that a diseased microenvironment may be required to induce phenotypic changes in microglial cells and the associated neuronal dysfunction seen in C9orf72 ALS/FTD neurodegeneration.
Collapse
Affiliation(s)
- Ileana Lorenzini
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Eric Alsop
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Jennifer Levy
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Lauren M. Gittings
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Deepti Lall
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Regenerative Medicine Institute, Los Angeles, CA, United States
| | - Benjamin E. Rabichow
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Stephen Moore
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Ryan Pevey
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Lynette M. Bustos
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Camelia Burciu
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Divya Bhatia
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Mo Singer
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Justin Saul
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Amanda McQuade
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| | - Makis Tzioras
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Discovery Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas A. Mota
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Regenerative Medicine Institute, Los Angeles, CA, United States
| | - Amber Logemann
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Jamie Rose
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Discovery Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Michael Marks
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Christopher J. Donnelly
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Shu-Ting Hung
- Department of Stem Cell Biology Regenerative Medicine, USC Keck School of Medicine, Los Angeles, CA, United States
| | - Justin Ichida
- Department of Stem Cell Biology Regenerative Medicine, USC Keck School of Medicine, Los Angeles, CA, United States
| | - Robert Bowser
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Tara Spires-Jones
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Brain Discovery Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mathew Blurton-Jones
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| | - Tania F. Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Robert H. Baloh
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Regenerative Medicine Institute, Los Angeles, CA, United States
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | | - Rita Sattler
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
43
|
Huseby CJ, Delvaux E, Brokaw DL, Coleman PD. Blood RNA transcripts reveal similar and differential alterations in fundamental cellular processes in Alzheimer's disease and other neurodegenerative diseases. Alzheimers Dement 2023; 19:2618-2632. [PMID: 36541444 PMCID: PMC11633037 DOI: 10.1002/alz.12880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dysfunctional processes in Alzheimer's disease and other neurodegenerative diseases lead to neural degeneration in the central and peripheral nervous system. Research demonstrates that neurodegeneration of any kind is a systemic disease that may even begin outside of the region vulnerable to the disease. Neurodegenerative diseases are defined by the vulnerabilities and pathology occurring in the regions affected. METHOD A random forest machine learning analysis on whole blood transcriptomes from six neurodegenerative diseases generated unbiased disease-classifying RNA transcripts subsequently subjected to pathway analysis. RESULTS We report that transcripts of the blood transcriptome selected for each of the neurodegenerative diseases represent fundamental biological cell processes including transcription regulation, degranulation, immune response, protein synthesis, apoptosis, cytoskeletal components, ubiquitylation/proteasome, and mitochondrial complexes that are also affected in the brain and reveal common themes across six neurodegenerative diseases. CONCLUSION Neurodegenerative diseases share common dysfunctions in fundamental cellular processes. Identifying regional vulnerabilities will reveal unique disease mechanisms. HIGHLIGHTS Transcriptomics offer information about dysfunctional processes. Comparing multiple diseases will expose unique malfunctions within diseases. Blood RNA can be used ante mortem to track expression changes in neurodegenerative diseases. Protocol standardization will make public datasets compatible.
Collapse
Affiliation(s)
- Carol J. Huseby
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Elaine Delvaux
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Danielle L. Brokaw
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paul D. Coleman
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
44
|
Berriat F, Lobsiger CS, Boillée S. The contribution of the peripheral immune system to neurodegeneration. Nat Neurosci 2023:10.1038/s41593-023-01323-6. [PMID: 37231108 DOI: 10.1038/s41593-023-01323-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 04/05/2023] [Indexed: 05/27/2023]
Abstract
Microglial cells are the major immune cells of the central nervous system (CNS), and directly react to neurodegeneration, but other immune cell types are also able to react to pathology and can modify the course of neurodegenerative processes. These mainly include monocytes/macrophages and lymphocytes. While these peripheral immune cells were initially considered to act only after infiltrating the CNS, recent evidence suggests that some of them can also act directly from the periphery. We will review the existing and emerging evidence for a role of peripheral immune cells in neurodegenerative diseases, both with and without CNS infiltration. Our focus will be on amyotrophic lateral sclerosis, but we will also compare to Alzheimer's disease and Parkinson's disease to highlight similarities or differences. Peripheral immune cells are easily accessible, and therefore may be an attractive therapeutic target for neurodegenerative diseases. Thus, understanding how these peripheral immune cells communicate with the CNS deserves deeper investigation.
Collapse
Affiliation(s)
- Félix Berriat
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christian S Lobsiger
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
45
|
Ecarnot F, Boccardi V, Calcagno A, Franceschi C, Fülop T, Itzhaki RF, Michel JP, Panza F, Rainero I, Solfrizzi V, Ticinesi A, Veronese N, Maggi S. Dementia, infections and vaccines: 30 years of controversy. Aging Clin Exp Res 2023; 35:1145-1160. [PMID: 37160649 PMCID: PMC10169152 DOI: 10.1007/s40520-023-02409-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 03/21/2023] [Indexed: 05/11/2023]
Abstract
This paper reports the proceedings of a virtual meeting convened by the European Interdisciplinary Council on Ageing (EICA), to discuss the involvement of infectious disorders in the pathogenesis of dementia and neurological disorders leading to dementia. We recap how our view of the infectious etiology of dementia has changed over the last 30 years in light of emerging evidence, and we present evidence in support of the implication of infection in dementia, notably Alzheimer's disease (AD). The bacteria and viruses thought to be responsible for neuroinflammation and neurological damage are reviewed. We then review the genetic basis for neuroinflammation and dementia, highlighting the genes that are currently the focus of investigation as potential targets for therapy. Next, we describe the antimicrobial hypothesis of dementia, notably the intriguing possibility that amyloid beta may itself possess antimicrobial properties. We further describe the clinical relevance of the gut-brain axis in dementia, the mechanisms by which infection can move from the intestine to the brain, and recent findings regarding dysbiosis patterns in patients with AD. We review the involvement of specific pathogens in neurological disorders, i.e. SARS-CoV-2, human immunodeficiency virus (HIV), herpes simplex virus type 1 (HSV1), and influenza. Finally, we look at the role of vaccination to prevent dementia. In conclusion, there is a large body of evidence supporting the involvement of various infectious pathogens in the pathogenesis of dementia, but large-scale studies with long-term follow-up are needed to elucidate the role that infection may play, especially before subclinical or clinical disease is present.
Collapse
Affiliation(s)
- Fiona Ecarnot
- EA3920, University of Franche-Comté, 25000, Besancon, France
- Department of Cardiology, University Hospital Besancon, 3-8 Boulevard Fleming, 25000, Besancon, France
| | - Virginia Boccardi
- Institute of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Santa Maria Della Misericordia Hospital, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Claudio Franceschi
- Laboratory of Systems Medicine of Healthy Aging, Institute of Biology and Biomedicine and Institute of Information Technology, Mathematics and Mechanics, Department of Applied Mathematics, N. I. Lobachevsky State University, Nizhny Novgorod, Russia
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Tamas Fülop
- Department of Medicine, Geriatrics Division, Research Center on Aging, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC, J1H 5N4, Canada
| | - Ruth F Itzhaki
- Institute of Population Ageing, University of Oxford and Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | - Francesco Panza
- Unit of Research Methodology and Data Sciences for Population Health, National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Innocenzo Rainero
- Dementia Center, Department of Neuroscience "Rita Levi Montalcini", University of Torino, Turin, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Andrea Ticinesi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Geriatric-Rehabilitation Department, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Nicola Veronese
- Geriatrics Section, Department of Internal Medicine, University of Palermo, Palermo, Italy.
| | - Stefania Maggi
- National Research Council, Neuroscience Institute, Aging Branch, Padua, Italy
| |
Collapse
|
46
|
Banerjee P, Mehta AR, Nirujogi RS, Cooper J, James OG, Nanda J, Longden J, Burr K, McDade K, Salzinger A, Paza E, Newton J, Story D, Pal S, Smith C, Alessi DR, Selvaraj BT, Priller J, Chandran S. Cell-autonomous immune dysfunction driven by disrupted autophagy in C9orf72-ALS iPSC-derived microglia contributes to neurodegeneration. SCIENCE ADVANCES 2023; 9:eabq0651. [PMID: 37083530 PMCID: PMC10121169 DOI: 10.1126/sciadv.abq0651] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/20/2023] [Indexed: 05/03/2023]
Abstract
Although microglial activation is widely found in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), the underlying mechanism(s) are poorly understood. Here, using human-induced pluripotent stem cell-derived microglia-like cells (hiPSC-MG) harboring the most common ALS/FTD mutation (C9orf72, mC9-MG), gene-corrected isogenic controls (isoC9-MG), and C9orf72 knockout hiPSC-MG (C9KO-MG), we show that reduced C9ORF72 protein is associated with impaired phagocytosis and an exaggerated immune response upon stimulation with lipopolysaccharide. Analysis of the C9ORF72 interactome revealed that C9ORF72 interacts with regulators of autophagy and functional studies showed impaired initiation of autophagy in mC9-MG and C9KO-MG. Coculture studies with motor neurons (MNs) demonstrated that the autophagy deficit in mC9-MG drives increased vulnerability of mC9-MNs to excitotoxic stimulus. Pharmacological activation of autophagy ameliorated both cell-autonomous functional deficits in hiPSC-MG and MN death in MG-MN coculture. Together, these findings reveal an important role for C9ORF72 in regulating immune homeostasis and identify dysregulation in myeloid cells as a contributor to neurodegeneration in ALS/FTD.
Collapse
Affiliation(s)
- Poulomi Banerjee
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Arpan R. Mehta
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Raja S. Nirujogi
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - James Cooper
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Owen G. James
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Jyoti Nanda
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - James Longden
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Karen Burr
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Karina McDade
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Edinburgh Brain Bank, Academic Department of Neuropathology, University of Edinburgh, Edinburgh, UK
- Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Andrea Salzinger
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Evdokia Paza
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Judith Newton
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - David Story
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Suvankar Pal
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
- Edinburgh Brain Bank, Academic Department of Neuropathology, University of Edinburgh, Edinburgh, UK
- Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Dario R. Alessi
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Bhuvaneish T. Selvaraj
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Josef Priller
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Department of Psychiatry and Psychotherapy; School of Medicine, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
- Neuropsychiatry, Charité–Universitätsmedizin Berlin and DZNE, Charitéplatz 1, 10117 Berlin, Germany
| | - Siddharthan Chandran
- UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh bioQuarter, Chancellor’s Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
47
|
Vinceti G, Gallingani C, Zucchi E, Martinelli I, Gianferrari G, Simonini C, Bedin R, Chiari A, Zamboni G, Mandrioli J. Young Onset Alzheimer's Disease Associated with C9ORF72 Hexanucleotide Expansion: Further Evidence for a Still Unsolved Association. Genes (Basel) 2023; 14:genes14040930. [PMID: 37107688 PMCID: PMC10138077 DOI: 10.3390/genes14040930] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are recognized as part of a disease continuum (FTD-ALS spectrum), in which the most common genetic cause is chromosome 9 open reading frame 72 (C9ORF72) gene hexanucleotide repeat expansion. The clinical phenotype of patients carrying this expansion varies widely and includes diseases beyond the FTD-ALS spectrum. Although a few cases of patients with C9ORF72 expansion and a clinical or biomarker-supported diagnosis of Alzheimer's disease (AD) have been described, they have been considered too sparse to establish a definite association between the C9ORF72 expansion and AD pathology. Here, we describe a C9ORF72 family with pleomorphic phenotypical expressions: a 54-year-old woman showing cognitive impairment and behavioral disturbances with both neuroimaging and cerebrospinal fluid (CSF) biomarkers consistent with AD pathology, her 49-year-old brother with typical FTD-ALS, and their 63-year-old mother with the behavioral variant of FTD and CSF biomarkers suggestive of AD pathology. The young onset of disease in all three family members and their different phenotypes and biomarker profiles make the simple co-occurrence of different diseases an extremely unlikely explanation. Our report adds to previous findings and may contribute to further expanding the spectrum of diseases associated with C9ORF72 expansion.
Collapse
Affiliation(s)
- Giulia Vinceti
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
| | - Chiara Gallingani
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Elisabetta Zucchi
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Ilaria Martinelli
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Gianferrari
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Cecilia Simonini
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Roberta Bedin
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
| | - Annalisa Chiari
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
| | - Giovanna Zamboni
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Jessica Mandrioli
- Neurology Unit, Azienda Ospedaliero Universitaria di Modena, 41126 Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
48
|
Marian OC, Teo JD, Lee JY, Song H, Kwok JB, Landin-Romero R, Halliday G, Don AS. Disrupted myelin lipid metabolism differentiates frontotemporal dementia caused by GRN and C9orf72 gene mutations. Acta Neuropathol Commun 2023; 11:52. [PMID: 36967384 PMCID: PMC10041703 DOI: 10.1186/s40478-023-01544-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/12/2023] [Indexed: 03/29/2023] Open
Abstract
Heterozygous mutations in the GRN gene and hexanucleotide repeat expansions in C9orf72 are the two most common genetic causes of Frontotemporal Dementia (FTD) with TDP-43 protein inclusions. The triggers for neurodegeneration in FTD with GRN (FTD-GRN) or C9orf72 (FTD-C9orf72) gene abnormalities are unknown, although evidence from mouse and cell culture models suggests that GRN mutations disrupt lysosomal lipid catabolism. To determine how brain lipid metabolism is affected in familial FTD with TDP-43 inclusions, and how this is related to myelin and lysosomal markers, we undertook comprehensive lipidomic analysis, enzyme activity assays, and western blotting on grey and white matter samples from the heavily-affected frontal lobe and less-affected parietal lobe of FTD-GRN cases, FTD-C9orf72 cases, and age-matched neurologically-normal controls. Substantial loss of myelin-enriched sphingolipids (sulfatide, galactosylceramide, sphingomyelin) and myelin proteins was observed in frontal white matter of FTD-GRN cases. A less-pronounced, yet statistically significant, loss of sphingolipids was also observed in FTD-C9orf72. FTD-GRN was distinguished from FTD-C9orf72 and control cases by increased acylcarnitines in frontal grey matter and marked accumulation of cholesterol esters in both frontal and parietal white matter, indicative of myelin break-down. Both FTD-GRN and FTD-C9orf72 cases showed significantly increased lysosomal and phagocytic protein markers, however galactocerebrosidase activity, required for lysosomal catabolism of galactosylceramide and sulfatide, was selectively increased in FTD-GRN. We conclude that both C9orf72 and GRN mutations are associated with disrupted lysosomal homeostasis and white matter lipid loss, but GRN mutations cause a more pronounced disruption to myelin lipid metabolism. Our findings support the hypothesis that hyperactive myelin lipid catabolism is a driver of gliosis and neurodegeneration in FTD-GRN. Since FTD-GRN is associated with white matter hyperintensities by MRI, our data provides important biochemical evidence supporting the use of MRI measures of white matter integrity in the diagnosis and management of FTD.
Collapse
Affiliation(s)
- Oana C Marian
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Jonathan D Teo
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Jun Yup Lee
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Huitong Song
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - John B Kwok
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Ramon Landin-Romero
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- School of Health Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Glenda Halliday
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Anthony S Don
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
- School of Medical Sciences, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
49
|
Wilson DM, Cookson MR, Van Den Bosch L, Zetterberg H, Holtzman DM, Dewachter I. Hallmarks of neurodegenerative diseases. Cell 2023; 186:693-714. [PMID: 36803602 DOI: 10.1016/j.cell.2022.12.032] [Citation(s) in RCA: 503] [Impact Index Per Article: 251.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 11/23/2022] [Accepted: 12/19/2022] [Indexed: 02/18/2023]
Abstract
Decades of research have identified genetic factors and biochemical pathways involved in neurodegenerative diseases (NDDs). We present evidence for the following eight hallmarks of NDD: pathological protein aggregation, synaptic and neuronal network dysfunction, aberrant proteostasis, cytoskeletal abnormalities, altered energy homeostasis, DNA and RNA defects, inflammation, and neuronal cell death. We describe the hallmarks, their biomarkers, and their interactions as a framework to study NDDs using a holistic approach. The framework can serve as a basis for defining pathogenic mechanisms, categorizing different NDDs based on their primary hallmarks, stratifying patients within a specific NDD, and designing multi-targeted, personalized therapies to effectively halt NDDs.
Collapse
Affiliation(s)
- David M Wilson
- Hasselt University, Biomedical Research Institute, BIOMED, 3500 Hasselt, Belgium.
| | - Mark R Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ludo Van Den Bosch
- KU Leuven, University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), 3000 Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China; UW Department of Medicine, School of Medicine and Public Health, Madison, WI, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Ilse Dewachter
- Hasselt University, Biomedical Research Institute, BIOMED, 3500 Hasselt, Belgium.
| |
Collapse
|
50
|
Pathak D, Sriram K. Molecular Mechanisms Underlying Neuroinflammation Elicited by Occupational Injuries and Toxicants. Int J Mol Sci 2023; 24:2272. [PMID: 36768596 PMCID: PMC9917383 DOI: 10.3390/ijms24032272] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Occupational injuries and toxicant exposures lead to the development of neuroinflammation by activating distinct mechanistic signaling cascades that ultimately culminate in the disruption of neuronal function leading to neurological and neurodegenerative disorders. The entry of toxicants into the brain causes the subsequent activation of glial cells, a response known as 'reactive gliosis'. Reactive glial cells secrete a wide variety of signaling molecules in response to neuronal perturbations and thus play a crucial role in the progression and regulation of central nervous system (CNS) injury. In parallel, the roles of protein phosphorylation and cell signaling in eliciting neuroinflammation are evolving. However, there is limited understanding of the molecular underpinnings associated with toxicant- or occupational injury-mediated neuroinflammation, gliosis, and neurological outcomes. The activation of signaling molecules has biological significance, including the promotion or inhibition of disease mechanisms. Nevertheless, the regulatory mechanisms of synergism or antagonism among intracellular signaling pathways remain elusive. This review highlights the research focusing on the direct interaction between the immune system and the toxicant- or occupational injury-induced gliosis. Specifically, the role of occupational injuries, e.g., trips, slips, and falls resulting in traumatic brain injury, and occupational toxicants, e.g., volatile organic compounds, metals, and nanoparticles/nanomaterials in the development of neuroinflammation and neurological or neurodegenerative diseases are highlighted. Further, this review recapitulates the recent advancement related to the characterization of the molecular mechanisms comprising protein phosphorylation and cell signaling, culminating in neuroinflammation.
Collapse
Affiliation(s)
| | - Krishnan Sriram
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| |
Collapse
|