1
|
Taddei RN, Duff KE. Synapse vulnerability and resilience across the clinical spectrum of dementias. Nat Rev Neurol 2025:10.1038/s41582-025-01094-7. [PMID: 40404832 DOI: 10.1038/s41582-025-01094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2025] [Indexed: 05/24/2025]
Abstract
Preservation of synapses is crucial for healthy cognitive ageing, and synapse loss is one of the closest anatomical correlates of cognitive decline in Alzheimer disease, dementia with Lewy bodies and frontotemporal dementia. In these conditions, some synapses seem particularly vulnerable to degeneration whereas others are resilient and remain preserved. Evidence has highlighted that vulnerability and resilience are intrinsically distinct phenomena linked to specific brain structural and/or functional signatures, yet the key features of vulnerable and resilient synapses in the dementias remain incompletely understood. Defining the characteristics of vulnerable and resilient synapses in each form of dementia could offer novel insight into the mechanisms of synapse preservation and of synapse loss that underlies cognitive decline, thereby facilitating the discovery of targeted biomarkers and disease-modifying therapies. In this Review, we consider the concepts of synapse vulnerability and resilience, and provide an overview of our current understanding of the associations between synaptic protein changes, neuropathology and cognitive decline. We also consider how understanding of the underlying mechanisms could identify novel strategies to mitigate the cognitive dysfunction associated with dementias.
Collapse
Affiliation(s)
- Raquel N Taddei
- Neurology Department, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- UK Dementia Research Institute at UCL, Institute of Neurology, University College London, London, UK.
| | - Karen E Duff
- UK Dementia Research Institute at UCL, Institute of Neurology, University College London, London, UK
| |
Collapse
|
2
|
Kedia S, Fertan E, Wu Y, Zhang YP, Meisl G, Lam JYL, K Wiseman F, McEwan WA, Quaegebeur A, Spillantini MG, Danial JSH, Klenerman D. SynPull: An advanced method for studying neurodegeneration-related aggregates in synaptosomes using super-resolution microscopy. Cell Chem Biol 2025; 32:338-351.e4. [PMID: 39862866 DOI: 10.1016/j.chembiol.2025.01.001] [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: 09/11/2024] [Revised: 11/09/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025]
Abstract
Synaptic dysfunction is a primary hallmark of both Alzheimer's and Parkinson's disease, leading to cognitive and behavioral decline. While alpha-synuclein, beta-amyloid, and tau are involved in the physiological functioning of synapses, their pathological aggregation has been linked to synaptopathology. The methodology for studying the small-soluble protein aggregates formed by these proteins is limited. Here we describe SynPull, a method combining single-molecule pull-down, super-resolution microscopy, and advanced computational analyses to characterize the protein aggregates in human and mouse synaptosomes. We show that AT8-positive tau aggregates are the predominant aggregate type in synaptosomes from postmortem Alzheimer's disease brain, although the aggregate size does not change in disease. Meanwhile, the relatively smaller amount of alpha-synuclein and beta-amyloid aggregates found in the synapses are larger than the extra-synaptic ones. Collectively, these results show the utility of SynPull to study pathological aggregates in neurodegeneration, elucidating the disease mechanisms causing synaptic dysfunction.
Collapse
Affiliation(s)
- Shekhar Kedia
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK
| | - Emre Fertan
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK; Department of Clinical Neurosciences, University of Cambridge Cambridge, UK
| | - Yunzhao Wu
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK
| | - Yu P Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK
| | - Georg Meisl
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK
| | - Jeff Y L Lam
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK
| | - Frances K Wiseman
- UK Dementia Research Institute at University College London London NW1 3BT, UK; Queen Square Institute of Neurology, University College London London WC1N 3BG, UK
| | - William A McEwan
- UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK; Department of Clinical Neurosciences, University of Cambridge Cambridge, UK
| | | | | | - John S H Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK; School of Physics and Astronomy, University of St Andrews St Andrews KY16 9SS, UK.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge Cambridge CB2 1EW, UK; UK Dementia Research Institute at University of Cambridge Cambridge CB2 0XY, UK.
| |
Collapse
|
3
|
Micheva KD, Simhal AK, Schardt J, Smith SJ, Weinberg RJ, Owen SF. Data-driven synapse classification reveals a logic of glutamate receptor diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.11.628056. [PMID: 39713368 PMCID: PMC11661198 DOI: 10.1101/2024.12.11.628056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The rich diversity of synapses facilitates the capacity of neural circuits to transmit, process and store information. We used multiplex super-resolution proteometric imaging through array tomography to define features of single synapses in mouse neocortex. We find that glutamatergic synapses cluster into subclasses that parallel the distinct biochemical and functional categories of receptor subunits: GluA1/4, GluA2/3 and GluN1/GluN2B. Two of these subclasses align with physiological expectations based on synaptic plasticity: large AMPAR-rich synapses may represent potentiated synapses, whereas small NMDAR-rich synapses suggest "silent" synapses. The NMDA receptor content of large synapses correlates with spine neck diameter, and thus the potential for coupling to the parent dendrite. Overall, ultrastructural features predict receptor content of synapses better than parent neuron identity does, suggesting synapse subclasses act as fundamental elements of neuronal circuits. No barriers prevent future generalization of this approach to other species, or to study of human disorders and therapeutics.
Collapse
Affiliation(s)
- Kristina D. Micheva
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Anish K. Simhal
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Jenna Schardt
- Allen Institute for Brain Science, Seattle, WA 98109
| | - Stephen J Smith
- Allen Institute for Brain Science, Seattle, WA 98109
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | - Richard J. Weinberg
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27514
| | - Scott F. Owen
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305
- Lead contact
| |
Collapse
|
4
|
Dong J, Tong W, Liu M, Liu M, Liu J, Jin X, Chen J, Jia H, Gao M, Wei M, Duan Y, Zhong X. Endosomal traffic disorders: a driving force behind neurodegenerative diseases. Transl Neurodegener 2024; 13:66. [PMID: 39716330 DOI: 10.1186/s40035-024-00460-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/05/2024] [Indexed: 12/25/2024] Open
Abstract
Endosomes are crucial sites for intracellular material sorting and transportation. Endosomal transport is a critical process involved in the selective uptake, processing, and intracellular transport of substances. The equilibrium between endocytosis and circulation mediated by the endosome-centered transport pathway plays a significant role in cell homeostasis, signal transduction, and immune response. In recent years, there have been hints linking endosomal transport abnormalities to neurodegenerative diseases, including Alzheimer's disease. Nonetheless, the related mechanisms remain unclear. Here, we provide an overview of endosomal-centered transport pathways and highlight potential physiological processes regulated by these pathways, with a particular focus on the correlation of endosomal trafficking disorders with common pathological features of neurodegenerative diseases. Additionally, we summarize potential therapeutic agents targeting endosomal trafficking for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jianru Dong
- School of Pharmacy, China Medical University, Shenyang, 110122, China
- Weifang Hospital of Traditional Chinese Medicine, Weifang, 261000, China
| | - Weiwei Tong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, 110069, China
| | - Mingyan Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Mengyu Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Jinyue Liu
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ju Chen
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Huachao Jia
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Menglin Gao
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, 110167, China.
| | - Ying Duan
- Liaoning Maternal and Child Health Hospital, Shenyang, 110005, China.
| | - Xin Zhong
- School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
5
|
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
|
6
|
Gu YY, Zhao XR, Zhang N, Yang Y, Yi Y, Shao QH, Liu MX, Zhang XL. Mitochondrial dysfunction as a therapeutic strategy for neurodegenerative diseases: Current insights and future directions. Ageing Res Rev 2024; 102:102577. [PMID: 39528070 DOI: 10.1016/j.arr.2024.102577] [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: 09/19/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative diseases, as common diseases in the elderly, tend to become younger due to environmental changes, social development and other factors. They are mainly characterized by progressive loss or dysfunction of neurons in the central or peripheral nervous system, and common diseases include Parkinson's disease, Alzheimer's disease, Huntington's disease and so on. Mitochondria are important organelles for adenosine triphosphate (ATP) production in the brain. In recent years, a large amount of evidence has shown that mitochondrial dysfunction plays a direct role in neurodegenerative diseases, which is expected to provide new ideas for the treatment of related diseases. This review will summarize the main mechanisms of mitochondrial dysfunction in neurodegenerative diseases, as well as collating recent advances in the study of mitochondrial disorders and new therapies.
Collapse
Affiliation(s)
- Ying-Ying Gu
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Xin-Ru Zhao
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Nan Zhang
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Yuan Yang
- Department of Gastroenterology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, PR China
| | - Ying Yi
- College of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Qian-Hang Shao
- Department of Pharmacy, Peking University People's Hospital, Beijing 100871, P R China
| | - Ming-Xuan Liu
- College of Pharmacy, Nantong University, Nantong 226001, PR China.
| | - Xiao-Ling Zhang
- College of Pharmacy, Nantong University, Nantong 226001, PR China.
| |
Collapse
|
7
|
Alharbi KS. Non-coding RNAs as therapeutic targets in Parkinson's Disease: A focus on dopamine. Pathol Res Pract 2024; 263:155641. [PMID: 39395297 DOI: 10.1016/j.prp.2024.155641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/14/2024]
Abstract
Parkinson's Disease is a highly complicated neurological disorder, with a key manifestation of loss of dopaminergic neurons. Despite the plethora of medicines that alleviate the symptoms, there is an urgent need for new treatments acting on the fundamental pathology of PD. Non-coding RNAs are becoming increasingly important in gene regulation and various cellular processes and are found to play a role in PD pathophysiology. This review analyzes the cross-talk of distinct ncRNAs with dopamine signaling. We attempt to constrain the various ncRNA networks that can activate dopamine production. First, we describe the deregulation of miRNAs that target dopamine receptors and have been implicated in PD. Next, we turn to the functions of lncRNAs in dopaminergic neurons and the connections to susceptibility genes for PD. Finally, we will analyze the novel circRNAs, such as ciRS-7, which may modulate dopamine-linked processes and serve as possible PD biomarkers. In this review, we describe recent progress in dopamine neuron revival to treat PD and the therapeutic potential of ncRNA. This review critically evaluates the available data, and we predict the role of some ncRNAs, such as PTBP1, to become candidate treatment targets in the future. Thus, this review aims to summarize the molecular causes for the deficit in dopamine signaling in PD and point to novel ncRNAs-linked therapeutic directions in neuroscience.
Collapse
Affiliation(s)
- Khalid Saad Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, AL Qassim 51452, Saudi Arabia.
| |
Collapse
|
8
|
Wang L, Wang Q, Wang X, Yang C, Wang X, Liu H, Wang H. Intermittent fasting alleviates postoperative cognitive dysfunction by reducing neuroinflammation in aged mice. Brain Res Bull 2024; 216:111034. [PMID: 39053649 DOI: 10.1016/j.brainresbull.2024.111034] [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: 04/27/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Elderly individuals undergoing surgical procedures are often confronted with the peril of experiencing postoperative cognitive dysfunction (POCD). Prior research has demonstrated the exacerbating effect of sevoflurane anesthesia on neuroinflammation, which can further deteriorate the condition of POCD in elderly patients. Intermittent fasting (IF) restricts food consumption to a specific time window and has been demonstrated to ameliorate cognitive dysfunction induced by neuropathic inflammation. We subjected 18-month-old male mice to 16 hours of fasting and 8 hours of unrestricted eating over a 24-hour period for 0, 1, 2, and 4 weeks, followed by abdominal exploration under sevoflurane anesthesia. In this study, we aim to explore the potential impact of IF on postoperative cognitive function in aged mice undergoing sevoflurane surgery through the preoperative implementation of IF measures. The findings indicate two weeks of IF leads to a significant enhancement of learning and memory capabilities in mice following surgery. The cognitive performance, as determined by the novel object recognition and Morris water maze tests, as well as the synaptic plasticity, as measured by in vivo electrophysiological recordings, has demonstrated marked improvements. Furthermore, the administration of IF markedly enhances the expression of synaptic-associated proteins in hippocampal neurons, concomitant with a decreasing expression of pro-inflammatory factors and a reduced density of microglial cells within the hippocampal brain region. To summarize, the results of this study indicate that IF may mitigate inflammation in the hippocampal area of the brain. Furthermore, IF appears to provide a safeguard against cognitive impairment and synaptic plasticity impairment brought on by sevoflurane anesthesia.
Collapse
Affiliation(s)
- Lei Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, China
| | - Qiang Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xiaoqing Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Chenyi Yang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China
| | - Xinyi Wang
- Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China
| | - Huan Liu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Haiyun Wang
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Anesthesiology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Nankai University 300071, Tianjin, China; Nankai University Affinity the Third Central Hospital, Tianjin 300170, China.
| |
Collapse
|
9
|
Liu Z, Cheng L, Cao W, Shen C, Qiu Y, Li C, Xiong Y, Yang SB, Chen Z, Yin X, Zhang X. Present and future use of exosomes containing proteins and RNAs in neurodegenerative diseases for synaptic function regulation: A comprehensive review. Int J Biol Macromol 2024; 280:135826. [PMID: 39322147 DOI: 10.1016/j.ijbiomac.2024.135826] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Neurodegenerative diseases (NDDs) are increasingly prevalent with global aging, demanding effective treatments. Exosomes, which contain biological macromolecules such as RNA (including miRNAs) and proteins like α-synuclein, tau, and amyloid-beta, are gaining attention as innovative therapeutics. This comprehensive review systematically explores the potential roles of exosomes in NDDs, with a particular focus on their role in synaptic dysfunction. We present the synaptic pathophysiology of NDDs and discuss the mechanisms of exosome formation, secretion, and action. Subsequently, we review the roles of exosomes in different types of NDDs, such as Alzheimer's disease and Parkinson's disease, with a special focus on their regulation of synaptic function. In addition, we explore the potential use of exosomes as biomarkers, as well as the challenges and opportunities in their clinical application. We provide perspectives on future research directions and development trends to provide a more comprehensive understanding of and guidance for the application of exosomes in the treatment of NDDs. In conclusion, exosomes rich in biological macromolecules, as a novel therapeutic strategy, have opened up new possibilities for the treatment of NDDs and brought new hope to patients.
Collapse
Affiliation(s)
- Ziying Liu
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Lin Cheng
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Wa Cao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Respiratory Medicine, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Chunxiao Shen
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Yuemin Qiu
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Chuan Li
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Yinyi Xiong
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Rehabilitation, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Seung Bum Yang
- Department of Medical Non-commissioned Officer, Wonkwang Health Science University Iksan-si, Jeollabuk-do 54538, South Korea
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Xiaoping Yin
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Xiaorong Zhang
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China.
| |
Collapse
|
10
|
Cordano C, Werneburg S, Abdelhak A, Bennett DJ, Beaudry-Richard A, Duncan GJ, Oertel FC, Boscardin WJ, Yiu HH, Jabassini N, Merritt L, Nocera S, Sin JH, Samana IP, Condor Montes SY, Ananth K, Bischof A, Nourbakhsh B, Hauser SL, Cree BAC, Emery B, Schafer DP, Chan JR, Green AJ. Synaptic injury in the inner plexiform layer of the retina is associated with progression in multiple sclerosis. Cell Rep Med 2024; 5:101490. [PMID: 38574736 PMCID: PMC11031420 DOI: 10.1016/j.xcrm.2024.101490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 02/01/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
While neurodegeneration underlies the pathological basis for permanent disability in multiple sclerosis (MS), predictive biomarkers for progression are lacking. Using an animal model of chronic MS, we find that synaptic injury precedes neuronal loss and identify thinning of the inner plexiform layer (IPL) as an early feature of inflammatory demyelination-prior to symptom onset. As neuronal domains are anatomically segregated in the retina and can be monitored longitudinally, we hypothesize that thinning of the IPL could represent a biomarker for progression in MS. Leveraging our dataset with over 800 participants enrolled for more than 12 years, we find that IPL atrophy directly precedes progression and propose that synaptic loss is predictive of functional decline. Using a blood proteome-wide analysis, we demonstrate a strong correlation between demyelination, glial activation, and synapse loss independent of neuroaxonal injury. In summary, monitoring synaptic injury is a biologically relevant approach that reflects a potential driver of progression.
Collapse
Affiliation(s)
- Christian Cordano
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Sebastian Werneburg
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Ophthalmology & Visual Sciences, Michigan Neuroscience Institute, University of Michigan - Michigan Medicine, Ann Arbor, MI, USA
| | - Ahmed Abdelhak
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel J Bennett
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Alexandra Beaudry-Richard
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Greg J Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Frederike C Oertel
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - W John Boscardin
- Department of Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Hao H Yiu
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Nora Jabassini
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Lauren Merritt
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sonia Nocera
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jung H Sin
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Isaac P Samana
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Shivany Y Condor Montes
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Kirtana Ananth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Antje Bischof
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bardia Nourbakhsh
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen L Hauser
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce A C Cree
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnik Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jonah R Chan
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Ari J Green
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
11
|
Serrano GE, Walker J, Nelson C, Glass M, Arce R, Intorcia A, Cline MP, Nabaty N, Acuña A, Huppert Steed A, Sue LI, Belden C, Choudhury P, Reiman E, Atri A, Beach TG. Correlation of Presynaptic and Postsynaptic Proteins with Pathology in Alzheimer's Disease. Int J Mol Sci 2024; 25:3130. [PMID: 38542104 PMCID: PMC10970005 DOI: 10.3390/ijms25063130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 04/28/2024] Open
Abstract
Synaptic transmission is essential for nervous system function and the loss of synapses is a known major contributor to dementia. Alzheimer's disease dementia (ADD) is characterized by synaptic loss in the mesial temporal lobe and cerebral neocortex, both of which are brain areas associated with memory and cognition. The association of synaptic loss and ADD was established in the late 1980s, and it has been estimated that 30-50% of neocortical synaptic protein is lost in ADD, but there has not yet been a quantitative profiling of different synaptic proteins in different brain regions in ADD from the same individuals. Very recently, positron emission tomography (PET) imaging of synapses is being developed, accelerating the focus on the role of synaptic loss in ADD and other conditions. In this study, we quantified the densities of two synaptic proteins, the presynaptic protein Synaptosome Associated Protein 25 (SNAP25) and the postsynaptic protein postsynaptic density protein 95 (PSD95) in the human brain, using enzyme-linked immunosorbent assays (ELISA). Protein was extracted from the cingulate gyrus, hippocampus, frontal, primary visual, and entorhinal cortex from cognitively unimpaired controls, subjects with mild cognitive impairment (MCI), and subjects with dementia that have different levels of Alzheimer's pathology. SNAP25 is significantly reduced in ADD when compared to controls in the frontal cortex, visual cortex, and cingulate, while the hippocampus showed a smaller, non-significant reduction, and entorhinal cortex concentrations were not different. In contrast, all brain areas showed lower PSD95 concentrations in ADD when compared to controls without dementia, although in the hippocampus, this failed to reach significance. Interestingly, cognitively unimpaired cases with high levels of AD pathology had higher levels of both synaptic proteins in all brain regions. SNAP25 and PSD95 concentrations significantly correlated with densities of neurofibrillary tangles, amyloid plaques, and Mini Mental State Examination (MMSE) scores. Our results suggest that synaptic transmission is affected by ADD in multiple brain regions. The differences were less marked in the entorhinal cortex and the hippocampus, most likely due to a ceiling effect imposed by the very early development of neurofibrillary tangles in older people in these brain regions.
Collapse
Affiliation(s)
- Geidy E. Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Jessica Walker
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Courtney Nelson
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Michael Glass
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Richard Arce
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Anthony Intorcia
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Madison P. Cline
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Natalie Nabaty
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Amanda Acuña
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Ashton Huppert Steed
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Lucia I. Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| | - Christine Belden
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA (P.C.)
| | - Parichita Choudhury
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA (P.C.)
| | - Eric Reiman
- The Banner Alzheimer’s Institute, Phoenix, AZ 85006, USA
| | - Alireza Atri
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ 85351, USA (P.C.)
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA; (J.W.); (R.A.); (A.I.); (M.P.C.); (N.N.); (A.A.); (A.H.S.)
| |
Collapse
|
12
|
Manoj M, Sowmyanarayan S, Kowshik AV, Chatterjee J. Identification of Potentially Repurposable Drugs for Lewy Body Dementia Using a Network-Based Approach. J Mol Neurosci 2024; 74:21. [PMID: 38363395 DOI: 10.1007/s12031-024-02199-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
The conventional method of one drug being used for one target has not yielded therapeutic solutions for Lewy body dementia (LBD), which is a leading progressive neurological disorder characterized by significant loss of neurons. The age-related disease is marked by memory loss, hallucinations, sleep disorder, mental health deterioration, palsy, and cognitive impairment, all of which have no known effective cure. The present study deploys a network medicine pipeline to repurpose drugs having considerable effect on the genes and proteins related to the diseases of interest. We utilized the novel SAveRUNNER algorithm to quantify the proximity of all drugs obtained from DrugBank with the disease associated gene dataset obtained from Phenopedia and targets in the human interactome. We found that most of the 154 FDA-approved drugs predicted by SAveRUNNER were used to treat nervous system disorders, but some off-label drugs like quinapril and selegiline were interestingly used to treat hypertension and Parkinson's disease (PD), respectively. Additionally, we performed gene set enrichment analysis using Connectivity Map (CMap) and pathway enrichment analysis using EnrichR to validate the efficacy of the drug candidates obtained from the pipeline approach. The investigation enabled us to identify the significant role of the synaptic vesicle pathway in our disease and accordingly finalize 8 suitable antidepressant drugs from the 154 drugs initially predicted by SAveRUNNER. These potential anti-LBD drugs are either selective or non-selective inhibitors of serotonin, dopamine, and norepinephrine transporters. The validated selective serotonin and norepinephrine inhibitors like milnacipran, protriptyline, and venlafaxine are predicted to manage LBD along with the affecting symptomatic issues.
Collapse
Affiliation(s)
- Megha Manoj
- Department of Biotechnology, PES University, Bangalore, 560085, India
| | | | - Arjun V Kowshik
- Department of Biotechnology, PES University, Bangalore, 560085, India
| | - Jhinuk Chatterjee
- Department of Biotechnology, PES University, Bangalore, 560085, India.
| |
Collapse
|
13
|
Almeida MF, Farizatto KLG, Almeida RS, Bahr BA. Lifestyle strategies to promote proteostasis and reduce the risk of Alzheimer's disease and other proteinopathies. Ageing Res Rev 2024; 93:102162. [PMID: 38070831 DOI: 10.1016/j.arr.2023.102162] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 10/31/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Unhealthy lifestyle choices, poor diet, and aging can have negative influences on cognition, gradually increasing the risk for mild cognitive impairment (MCI) and the continuum comprising early dementia. Aging is the greatest risk factor for age-related dementias such as Alzheimer's disease, and the aging process is known to be influenced by life events that can positively or negatively affect age-related diseases. Remarkably, life experiences that make the brain vulnerable to dementia, such as seizure episodes, neurotoxin exposures, metabolic disorders, and trauma-inducing events (e.g. traumatic injuries or mild neurotrauma from a fall or blast exposure), have been associated with negative effects on proteostasis and synaptic integrity. Functional compromise of the autophagy-lysosomal pathway, a major contributor to proteostasis, has been implicated in Alzheimer's disease, Parkinson's disease, obesity-related pathology, Huntington's disease, as well as in synaptic degeneration which is the best correlate of cognitive decline. Correspondingly, pharmacological and non-pharmacological strategies that positively modulate lysosomal proteases are recognized as synaptoprotective through degradative clearance of pathogenic proteins. Here, we discuss life-associated vulnerabilities that influence key hallmarks of brain aging and the increased burden of age-related dementias. Additionally, we discuss exercise and diet among the lifestyle strategies that regulate proteostasis as well as synaptic integrity, leading to evident prevention of cognitive deficits during brain aging in pre-clinical models.
Collapse
Affiliation(s)
- Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina - Pembroke, Pembroke, NC 28372, USA; Department of Biology, University of North Carolina - Pembroke, Pembroke, NC 28372, USA; Department of Biology & Marine Biology, and the Integrative, Comparative & Marine Biology Program, University of North Carolina - Wilmington, Wilmington, NC 28409, USA
| | - Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina - Pembroke, Pembroke, NC 28372, USA
| | - Renato S Almeida
- Department of Biosciences, University of Taubate, Taubate, SP 12020-270, Brazil
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina - Pembroke, Pembroke, NC 28372, USA; Department of Biology, University of North Carolina - Pembroke, Pembroke, NC 28372, USA.
| |
Collapse
|
14
|
Shin S, Lee J, Kim YG, Ha C, Park JH, Kim JW, Lee J, Jang JH. Genetic Diagnosis of Children With Neurodevelopmental Disorders Using Whole Genome Sequencing. Pediatr Neurol 2023; 149:44-52. [PMID: 37776660 DOI: 10.1016/j.pediatrneurol.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/05/2023] [Accepted: 09/05/2023] [Indexed: 10/02/2023]
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) have diverse phenotypes. Their genetic diagnoses are often challenged by difficulties of targeting causative genes due to heterogeneous genetic etiologies. The objective of this study was to perform genetic diagnosis of children with NDDs using whole genome sequencing. METHODS This study included 78 pediatric patients with NDDs and their 152 family members for whole genome sequencing (WGS). All cases except one were families with at least two members. Seventy-five patients had previously undergone other genetic tests besides WGS. Detected variants were classified according to the guidelines of the American College of Medical Genetics and Genomics. RESULTS Among 78 probands, 26 patients were genetically diagnosed with NDDs through WGS, showing a diagnostic rate of 33.3%. Of them, 22 cases had de novo variants (DNVs) identified through trio analysis. Of these DNVs, half were novel variants. Three structural variants, including a multiexon deletion, a contiguous gene deletion involving 13 Mb, and a retrotransposon insertion, were revealed by WGS. All cases except one had defects in different genes, consistent with the phenotypically diverse nature of NDDs. In addition, three patients were inconclusive, two of them had one likely pathogenic variant in a gene associated with autosomal recessive disease and the other one had no clinical phenotypes associated with the detected DNV. CONCLUSIONS Our experience demonstrates the advantage of WGS in the diagnosis of NDDs, including detection of copy number variations and also the advantage of trio sequencing for interpretation of DNVs.
Collapse
Affiliation(s)
- Sunghwan Shin
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Laboratory Medicine, Inje University Ilsan Paik Hospital, Goyang, Korea
| | - Jiwon Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Gon Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Changhee Ha
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Ho Park
- Clinical Genomics Center, Samsung Medical Center, Seoul, Korea
| | - Jong-Won Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeehun Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
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
|
16
|
Parisi MJ, Aimino MA, Mosca TJ. A conditional strategy for cell-type-specific labeling of endogenous excitatory synapses in Drosophila. CELL REPORTS METHODS 2023; 3:100477. [PMID: 37323572 PMCID: PMC10261928 DOI: 10.1016/j.crmeth.2023.100477] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/28/2023] [Accepted: 04/19/2023] [Indexed: 06/17/2023]
Abstract
Chemical neurotransmission occurs at specialized contacts where neurotransmitter release machinery apposes neurotransmitter receptors to underlie circuit function. A series of complex events underlies pre- and postsynaptic protein recruitment to neuronal connections. To better study synaptic development in individual neurons, we need cell-type-specific strategies to visualize endogenous synaptic proteins. Although presynaptic strategies exist, postsynaptic proteins remain less studied because of a paucity of cell-type-specific reagents. To study excitatory postsynapses with cell-type specificity, we engineered dlg1[4K], a conditionally labeled marker of Drosophila excitatory postsynaptic densities. With binary expression systems, dlg1[4K] labels central and peripheral postsynapses in larvae and adults. Using dlg1[4K], we find that distinct rules govern postsynaptic organization in adult neurons, multiple binary expression systems can concurrently label pre- and postsynapse in a cell-type-specific manner, and neuronal DLG1 can sometimes localize presynaptically. These results validate our strategy for conditional postsynaptic labeling and demonstrate principles of synaptic organization.
Collapse
Affiliation(s)
- Michael J. Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Michael A. Aimino
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Timothy J. Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| |
Collapse
|
17
|
Xia L, Zhang F, Li Y, Mo Y, Zhang L, Li Q, Luo M, Hou X, Du Z, Deng J, Hao E. A new perspective on Alzheimer's disease: m6A modification. Front Genet 2023; 14:1166831. [PMID: 37255714 PMCID: PMC10225986 DOI: 10.3389/fgene.2023.1166831] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023] Open
Abstract
As a neurodegenerative disease, Alzheimer's disease (AD) is characterized by synaptic loss, extracellular plaques of amyloid accumulation, hyperphosphorylation of tau, and neuroinflammation. Various biological processes are affected by epitranscriptomic modifications, which regulate the metabolism of mRNA in cells and regulate the expression of genes. In response to changes in m6A modification levels, the nervous system becomes dysfunctional and plays a significant role in the development of Alzheimer's disease. As a result of recent research, this paper reviews advances in the understanding of the regulatory mechanisms of m6A modification in the occurrence and development of AD. In addition, the article discusses recent research techniques related to animal models of m6A and AD. Furthermore, it discusses the possibility of studying the pathogenesis of AD at the level of the epitranscriptome, identifying early diagnostic markers, and screening for effective treatment options.
Collapse
Affiliation(s)
- Lei Xia
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
| | - Fan Zhang
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi International Zhang Medicine Hospital Affiliated to Gungxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Yulu Li
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
| | - Yuemi Mo
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
| | - Lingqiu Zhang
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
| | - Qianhua Li
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
| | - Minghuang Luo
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
| | - Xiaotao Hou
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhengcai Du
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiagang Deng
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| | - Erwei Hao
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
18
|
Filipović D, Novak B, Xiao J, Yan Y, Bernardi RE, Turck CW. Chronic fluoxetine treatment in socially-isolated rats modulates the prefrontal cortex synaptoproteome. J Proteomics 2023; 282:104925. [PMID: 37164273 DOI: 10.1016/j.jprot.2023.104925] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Exposure to chronic social isolation (CSIS) and synapse dysfunction have been implicated in the etiology of major depressive disorder (MDD). Fluoxetine (Flx) has been widely used to treat MDD, but its mechanisms of action remain elusive. We employed comparative synaptoproteomics to investigate the changes in the levels of proteins and molecular signaling pathways in prefrontal cortical samples of adult male Wistar rats exposed to CSIS, a rat model of depression, and CSIS rats treated with chronic Flx and controls, using liquid chromatography coupled to tandem mass spectrometry. Flx-treated control rats showed a decreased level of proteins involved in vesicle-mediated transport, and a predominantly increased level of exocytosis-associated proteins. CSIS significantly reduced the level of proteins involved in the ATP metabolic process, clathrin-dependent endocytosis, and proteolysis. Flx treatment in CSIS rats stimulated synaptic vesicle trafficking by increasing the regulation of exo/endocytosis-associated proteins, proteins involved in synaptic plasticity including neurogenesis, Cox5a, mitochondria-associated proteins involved in oxidative phosphorylation, and ion transport proteins (Slc8a2, Atp1b2). Flx treatment resulted in an increased synaptic vesicle dynamic, plasticity and mitochondrial functionality, and a suppression of CSIS-induced impairment of these processes. BIOLOGICAL SIGNIFICANCE: Identifying biomarkers of MDD and treatment response is the goal of many studies. Contemporary studies have shown that many molecular alterations associated with the pathophysiology of MDD reside within the synapse. As part of this research, a growing importance is the use of proteomics, as monitoring the changes in protein levels enables the identification of (possible) biochemical pathways and processes of importance for the development of depressive-like behavior and the efficacy of antidepressant treatments. We profiled proteomic changes representative of the development of CSIS-induced depressive-like behavior and the antidepressant effects of Flx. Our study has identified synaptosomal proteins and altered molecular pathways that may be potential markers of prefrontal cortical synaptic dysfunction associated with depressive-like behavior, and further clarified the mechanisms of depressive-like behavior and mode of action of Flx. Our findings indicate potential PFC synaptic targets for antidepressant treatment.
Collapse
Affiliation(s)
- Dragana Filipović
- Department of Molecular Biology and Endocrinology, "VINČA", Institute of Nuclear Sciences - National Institute of thе Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Božidar Novak
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jinqiu Xiao
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Yu Yan
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph W Turck
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
19
|
Fernandes MYD, Lopes JP, Silva HB, Andrade GM, Cunha RA, Tomé AR. Caffeic acid recovers ischemia-induced synaptic dysfunction without direct effects on excitatory synaptic transmission and plasticity in mouse hippocampal slices. Neurosci Lett 2023; 808:137292. [PMID: 37156440 DOI: 10.1016/j.neulet.2023.137292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
Caffeic acid is a polyphenolic compound present in a vast array of dietary components. We previously showed that caffeic acid reduces the burden of brain ischemia joining evidence by others that it can attenuate different brain diseases. However, it is unknown if caffeic acid affects information processing in neuronal networks. Thus, we now used electrophysiological recordings in mouse hippocampal slices to test if caffeic acid directly affected synaptic transmission, plasticity and dysfunction caused by oxygen-glucose deprivation (OGD), an in vitro ischemia model. Caffeic acid (1-10 μM) was devoid of effect on synaptic transmission and paired-pulse facilitation in Schaffer collaterals-CA1 pyramidal synapses. Also, the magnitude of either hippocampal long-term potentiation (LTP) or the subsequent depotentiation were not significantly modified by 10 μM caffeic acid. However, caffeic acid (10 μM) increased the recovery of synaptic transmission upon re-oxygenation following 7 minutes of OGD. Furthermore, caffeic acid (10 μM) also recovered plasticity after OGD, as heralded by the increased magnitude of LTP after exposure. These findings show that caffeic acid does not directly affect synaptic transmission and plasticity but can indirectly affect other cellular targets to correct synaptic dysfunction. Unraveling the molecular mechanisms of action of caffeic acid may allow the design of hitherto unrecognized novel neuroprotective strategies.
Collapse
Affiliation(s)
- Mara Yone D Fernandes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Physiology and Pharmacology, Faculty of Medicine, Center for Research and Drug Development (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - João Pedro Lopes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Henrique B Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Geanne M Andrade
- Department of Physiology and Pharmacology, Faculty of Medicine, Center for Research and Drug Development (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - Angelo R Tomé
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
20
|
Saunders TS, Pozzolo FE, Heslegrave A, King D, McGeachan RI, Spires-Jones MP, Harris SE, Ritchie C, Muniz-Terrera G, Deary IJ, Cox SR, Zetterberg H, Spires-Jones TL. Predictive blood biomarkers and brain changes associated with age-related cognitive decline. Brain Commun 2023; 5:fcad113. [PMID: 37180996 PMCID: PMC10167767 DOI: 10.1093/braincomms/fcad113] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/28/2022] [Accepted: 04/05/2023] [Indexed: 04/08/2023] Open
Abstract
Growing evidence supports the use of plasma levels of tau phosphorylated at threonine 181, amyloid-β, neurofilament light and glial fibrillary acidic protein as promising biomarkers for Alzheimer's disease. While these blood biomarkers are promising for distinguishing people with Alzheimer's disease from healthy controls, their predictive validity for age-related cognitive decline without dementia remains unclear. Further, while tau phosphorylated at threonine 181 is a promising biomarker, the distribution of this phospho-epitope of tau in the brain is unknown. Here, we tested whether plasma levels of tau phosphorylated at threonine 181, amyloid-β, neurofilament light and fibrillary acidic protein predict cognitive decline between ages 72 and 82 in 195 participants in the Lothian birth cohorts 1936 study of cognitive ageing. We further examined post-mortem brain samples from temporal cortex to determine the distribution of tau phosphorylated at threonine 181 in the brain. Several forms of tau phosphorylated at threonine 181 have been shown to contribute to synapse degeneration in Alzheimer's disease, which correlates closely with cognitive decline in this form of dementia, but to date, there have not been investigations of whether tau phosphorylated at threonine 181 is found in synapses in Alzheimer's disease or healthy ageing brain. It was also previously unclear whether tau phosphorylated at threonine 181 accumulated in dystrophic neurites around plaques, which could contribute to tau leakage to the periphery due to impaired membrane integrity in dystrophies. Brain homogenate and biochemically enriched synaptic fractions were examined with western blot to examine tau phosphorylated at threonine 181 levels between groups (n = 10-12 per group), and synaptic and astrocytic localization of tau phosphorylated at threonine 181 were examined using array tomography (n = 6-15 per group), and localization of tau phosphorylated at threonine 181 in plaque-associated dystrophic neurites with associated gliosis were examined with standard immunofluorescence (n = 8-9 per group). Elevated baseline plasma tau phosphorylated at threonine 181, neurofilament light and fibrillary acidic protein predicted steeper general cognitive decline during ageing. Further, increasing tau phosphorylated at threonine 181 over time predicted general cognitive decline in females only. Change in plasma tau phosphorylated at threonine 181 remained a significant predictor of g factor decline when taking into account Alzheimer's disease polygenic risk score, indicating that the increase of blood tau phosphorylated at threonine 181 in this cohort was not only due to incipient Alzheimer's disease. Tau phosphorylated at threonine 181 was observed in synapses and astrocytes in both healthy ageing and Alzheimer's disease brain. We observed that a significantly higher proportion of synapses contain tau phosphorylated at threonine 181 in Alzheimer's disease relative to aged controls. Aged controls with pre-morbid lifetime cognitive resilience had significantly more tau phosphorylated at threonine 181 in fibrillary acidic protein-positive astrocytes than those with pre-morbid lifetime cognitive decline. Further, tau phosphorylated at threonine 181 was found in dystrophic neurites around plaques and in some neurofibrillary tangles. The presence of tau phosphorylated at threonine 181 in plaque-associated dystrophies may be a source of leakage of tau out of neurons that eventually enters the blood. Together, these data indicate that plasma tau phosphorylated at threonine 181, neurofilament light and fibrillary acidic protein may be useful biomarkers of age-related cognitive decline, and that efficient clearance of tau phosphorylated at threonine 181 by astrocytes may promote cognitive resilience.
Collapse
Affiliation(s)
- Tyler S Saunders
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Francesca E Pozzolo
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Amanda Heslegrave
- United Kingdom UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Declan King
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Robert I McGeachan
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Maxwell P Spires-Jones
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Sarah E Harris
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Craig Ritchie
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention & Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Department of Social Medicine, Ohio University, Athens, Ohio 45701, USA
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago 3485, Chile
| | - Ian J Deary
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Simon R Cox
- Lothian Birth Cohort studies, Department of Psychology, University of Edinburgh, Edinburgh, EH8 9AD, UK
| | - Henrik Zetterberg
- United Kingdom UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, S-431 80 Molndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Molndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Tara L Spires-Jones
- UK Dementia Research Institute and Centre for Discovery Brain Sciences at the University of Edinburgh, Edinburgh, EH8 9JZ, UK
| |
Collapse
|
21
|
Meftah S, Gan J. Alzheimer's disease as a synaptopathy: Evidence for dysfunction of synapses during disease progression. Front Synaptic Neurosci 2023; 15:1129036. [PMID: 36970154 PMCID: PMC10033629 DOI: 10.3389/fnsyn.2023.1129036] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
The synapse has consistently been considered a vulnerable and critical target within Alzheimer's disease, and synapse loss is, to date, one of the main biological correlates of cognitive decline within Alzheimer's disease. This occurs prior to neuronal loss with ample evidence that synaptic dysfunction precedes this, in support of the idea that synaptic failure is a crucial stage within disease pathogenesis. The two main pathological hallmarks of Alzheimer's disease, abnormal aggregates of amyloid or tau proteins, have had demonstrable effects on synaptic physiology in animal and cellular models of Alzheimer's disease. There is also growing evidence that these two proteins may have a synergistic effect on neurophysiological dysfunction. Here, we review some of the main findings of synaptic alterations in Alzheimer's disease, and what we know from Alzheimer's disease animal and cellular models. First, we briefly summarize some of the human evidence to suggest that synapses are altered, including how this relates to network activity. Subsequently, animal and cellular models of Alzheimer's disease are considered, highlighting mouse models of amyloid and tau pathology and the role these proteins may play in synaptic dysfunction, either in isolation or examining how the two pathologies may interact in dysfunction. This specifically focuses on neurophysiological function and dysfunction observed within these animal models, typically measured using electrophysiology or calcium imaging. Following synaptic dysfunction and loss, it would be impossible to imagine that this would not alter oscillatory activity within the brain. Therefore, this review also discusses how this may underpin some of the aberrant oscillatory patterns seen in animal models of Alzheimer's disease and human patients. Finally, an overview of some key directions and considerations in the field of synaptic dysfunction in Alzheimer's disease is covered. This includes current therapeutics that are targeted specifically at synaptic dysfunction, but also methods that modulate activity to rescue aberrant oscillatory patterns. Other important future avenues of note in this field include the role of non-neuronal cell types such as astrocytes and microglia, and mechanisms of dysfunction independent of amyloid and tau in Alzheimer's disease. The synapse will certainly continue to be an important target within Alzheimer's disease for the foreseeable future.
Collapse
Affiliation(s)
- Soraya Meftah
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jian Gan
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
22
|
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: 660] [Impact Index Per Article: 330.0] [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
|
23
|
Phospholipase D1 Attenuation Therapeutics Promotes Resilience against Synaptotoxicity in 12-Month-Old 3xTg-AD Mouse Model of Progressive Neurodegeneration. Int J Mol Sci 2023; 24:ijms24043372. [PMID: 36834781 PMCID: PMC9967100 DOI: 10.3390/ijms24043372] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Abrogating synaptotoxicity in age-related neurodegenerative disorders is an extremely promising area of research with significant neurotherapeutic implications in tauopathies including Alzheimer's disease (AD). Our studies using human clinical samples and mouse models demonstrated that aberrantly elevated phospholipase D1 (PLD1) is associated with amyloid beta (Aβ) and tau-driven synaptic dysfunction and underlying memory deficits. While knocking out the lipolytic PLD1 gene is not detrimental to survival across species, elevated expression is implicated in cancer, cardiovascular conditions and neuropathologies, leading to the successful development of well-tolerated mammalian PLD isoform-specific small molecule inhibitors. Here, we address the importance of PLD1 attenuation, achieved using repeated 1 mg/kg of VU0155069 (VU01) intraperitoneally every alternate day for a month in 3xTg-AD mice beginning only from ~11 months of age (with greater influence of tau-driven insults) compared to age-matched vehicle (0.9% saline)-injected siblings. A multimodal approach involving behavior, electrophysiology and biochemistry corroborate the impact of this pre-clinical therapeutic intervention. VU01 proved efficacious in preventing in later stage AD-like cognitive decline affecting perirhinal cortex-, hippocampal- and amygdala-dependent behaviors. Glutamate-dependent HFS-LTP and LFS-LTD improved. Dendritic spine morphology showed the preservation of mushroom and filamentous spine characteristics. Differential PLD1 immunofluorescence and co-localization with Aβ were noted.
Collapse
|
24
|
Effects of Chronic Caffeine Consumption on Synaptic Function, Metabolism and Adenosine Modulation in Different Brain Areas. Biomolecules 2023; 13:biom13010106. [PMID: 36671491 PMCID: PMC9855869 DOI: 10.3390/biom13010106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Adenosine receptors mainly control synaptic function, and excessive activation of adenosine receptors may worsen the onset of many neurological disorders. Accordingly, the regular intake of moderate doses of caffeine antagonizes adenosine receptors and affords robust neuroprotection. Although caffeine intake alters brain functional connectivity and multi-omics analyses indicate that caffeine intake modifies synaptic and metabolic processes, it is unclear how caffeine intake affects behavior, synaptic plasticity and its modulation by adenosine. We now report that male mice drinking caffeinated water (0.3 g/L) for 2 weeks were behaviorally indistinguishable (locomotion, mood, memory) from control mice (drinking water) and displayed superimposable synaptic plasticity (long-term potentiation) in different brain areas (hippocampus, prefrontal cortex, amygdala). Moreover, there was a general preservation of the efficiency of adenosine A1 and A2A receptors to control synaptic transmission and plasticity, although there was a tendency for lower levels of endogenous adenosine ensuring A1 receptor-mediated inhibition. In spite of similar behavioral and neurophysiological function, caffeine intake increased the energy charge and redox state of cortical synaptosomes. This increased metabolic competence likely involved a putative increase in the glycolytic rate in synapses and a prospective greater astrocyte-synapse lactate shuttling. It was concluded that caffeine intake does not trigger evident alterations of behavior or of synaptic plasticity but increases the metabolic competence of synapses, which might be related with the previously described better ability of animals consuming caffeine to cope with deleterious stimuli triggering brain dysfunction.
Collapse
|
25
|
Kromer JA, Tass PA. Synaptic reshaping of plastic neuronal networks by periodic multichannel stimulation with single-pulse and burst stimuli. PLoS Comput Biol 2022; 18:e1010568. [PMID: 36327232 PMCID: PMC9632832 DOI: 10.1371/journal.pcbi.1010568] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022] Open
Abstract
Synaptic dysfunction is associated with several brain disorders, including Alzheimer's disease, Parkinson's disease (PD) and obsessive compulsive disorder (OCD). Utilizing synaptic plasticity, brain stimulation is capable of reshaping synaptic connectivity. This may pave the way for novel therapies that specifically counteract pathological synaptic connectivity. For instance, in PD, novel multichannel coordinated reset stimulation (CRS) was designed to counteract neuronal synchrony and down-regulate pathological synaptic connectivity. CRS was shown to entail long-lasting therapeutic aftereffects in PD patients and related animal models. This is in marked contrast to conventional deep brain stimulation (DBS) therapy, where PD symptoms return shortly after stimulation ceases. In the present paper, we study synaptic reshaping by periodic multichannel stimulation (PMCS) in networks of leaky integrate-and-fire (LIF) neurons with spike-timing-dependent plasticity (STDP). During PMCS, phase-shifted periodic stimulus trains are delivered to segregated neuronal subpopulations. Harnessing STDP, PMCS leads to changes of the synaptic network structure. We found that the PMCS-induced changes of the network structure depend on both the phase lags between stimuli and the shape of individual stimuli. Single-pulse stimuli and burst stimuli with low intraburst frequency down-regulate synapses between neurons receiving stimuli simultaneously. In contrast, burst stimuli with high intraburst frequency up-regulate these synapses. We derive theoretical approximations of the stimulation-induced network structure. This enables us to formulate stimulation strategies for inducing a variety of network structures. Our results provide testable hypotheses for future pre-clinical and clinical studies and suggest that periodic multichannel stimulation may be suitable for reshaping plastic neuronal networks to counteract pathological synaptic connectivity. Furthermore, we provide novel insight on how the stimulus type may affect the long-lasting outcome of conventional DBS. This may strongly impact parameter adjustment procedures for clinical DBS, which, so far, primarily focused on acute effects of stimulation.
Collapse
Affiliation(s)
- Justus A Kromer
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| | - Peter A Tass
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| |
Collapse
|
26
|
Piscopo P, Crestini A, Carbone E, Rivabene R, Ancidoni A, Lo Giudice M, Corbo M, Vanacore N, Lacorte E. A systematic review on drugs for synaptic plasticity in the treatment of dementia. Ageing Res Rev 2022; 81:101726. [PMID: 36031056 DOI: 10.1016/j.arr.2022.101726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 01/31/2023]
Abstract
The aim of the present systematic review (SR) was to provide an overview of all published and unpublished clinical trials investigating the safety and efficacy of disease-modifying drugs targeting synaptic plasticity in dementia. Searches on CT.gov and EuCT identified 27 trials (4 phase-1, 1 phase-1/2, 18 phase-2, 1 phase-2/3, 1 phase-3, 1 phase-4, and 1 not reported). Twenty of them completed, and seven are currently active or enrolling. The structured bibliographic searches yielded 3585 records. A total of 12 studies were selected on Levetiracetam, Masitinib, Saracatinib, BI 40930, Bryostatin 1, PF-04447943 and Edonerpic drugs. We used RoB tool for quality analysis of randomized studies. Efficacy was assessed as a primary outcome in all studies except one and the main scale used was ADAS-Cog (7 studies), MMSE and CDR (4 studies). Safety and tolerability were reported in eleven studies. The incidence of SAEs was similar between treatment and placebo. At the moment, only one molecule reached phase-3. This could suggest that research on these drugs is still preliminary. Of all, three studies reported promising results on Levetiracetam, Bryostatin 1 and Masitinib.
Collapse
Affiliation(s)
- P Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy.
| | - A Crestini
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - E Carbone
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - R Rivabene
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - A Ancidoni
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| | - M Lo Giudice
- Need Institute, Foundation for Cure and Rehabilitation of Neurological Diseases, Milan, Italy
| | - M Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy.
| | - N Vanacore
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| | - E Lacorte
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| |
Collapse
|
27
|
Laszlo ZI, Hindley N, Sanchez Avila A, Kline RA, Eaton SL, Lamont DJ, Smith C, Spires-Jones TL, Wishart TM, Henstridge CM. Synaptic proteomics reveal distinct molecular signatures of cognitive change and C9ORF72 repeat expansion in the human ALS cortex. Acta Neuropathol Commun 2022; 10:156. [DOI: 10.1186/s40478-022-01455-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractIncreasing evidence suggests synaptic dysfunction is a central and possibly triggering factor in Amyotrophic Lateral Sclerosis (ALS). Despite this, we still know very little about the molecular profile of an ALS synapse. To address this gap, we designed a synaptic proteomics experiment to perform an unbiased assessment of the synaptic proteome in the ALS brain. We isolated synaptoneurosomes from fresh-frozen post-mortem human cortex (11 controls and 18 ALS) and stratified the ALS group based on cognitive profile (Edinburgh Cognitive and Behavioural ALS Screen (ECAS score)) and presence of a C9ORF72 hexanucleotide repeat expansion (C9ORF72-RE). This allowed us to assess regional differences and the impact of phenotype and genotype on the synaptic proteome, using Tandem Mass Tagging-based proteomics. We identified over 6000 proteins in our synaptoneurosomes and using robust bioinformatics analysis we validated the strong enrichment of synapses. We found more than 30 ALS-associated proteins in synaptoneurosomes, including TDP-43, FUS, SOD1 and C9ORF72. We identified almost 500 proteins with altered expression levels in ALS, with region-specific changes highlighting proteins and pathways with intriguing links to neurophysiology and pathology. Stratifying the ALS cohort by cognitive status revealed almost 150 specific alterations in cognitively impaired ALS synaptic preparations. Stratifying by C9ORF72-RE status revealed 330 protein alterations in the C9ORF72-RE +ve group, with KEGG pathway analysis highlighting strong enrichment for postsynaptic dysfunction, related to glutamatergic receptor signalling. We have validated some of these changes by western blot and at a single synapse level using array tomography imaging. In summary, we have generated the first unbiased map of the human ALS synaptic proteome, revealing novel insight into this key compartment in ALS pathophysiology and highlighting the influence of cognitive decline and C9ORF72-RE on synaptic composition.
Collapse
|
28
|
Cassinotti LR, Ji L, Borges BC, Cass ND, Desai AS, Kohrman DC, Liberman MC, Corfas G. Cochlear Neurotrophin-3 overexpression at mid-life prevents age-related inner hair cell synaptopathy and slows age-related hearing loss. Aging Cell 2022; 21:e13708. [PMID: 36088647 PMCID: PMC9577954 DOI: 10.1111/acel.13708] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 08/02/2022] [Accepted: 08/21/2022] [Indexed: 01/25/2023] Open
Abstract
Age-related hearing loss (ARHL) is the most prevalent sensory deficit in the elderly. This progressive pathology often has psychological and medical comorbidities, including social isolation, depression, and cognitive decline. Despite ARHL's enormous societal and economic impact, no therapies to prevent or slow its progression exist. Loss of synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs), a.k.a. IHC synaptopathy, is an early event in cochlear aging, preceding neuronal and hair cell loss. To determine if age-related IHC synaptopathy can be prevented, and if this impacts the time-course of ARHL, we tested the effects of cochlear overexpression of neurotrophin-3 (Ntf3) starting at middle age. We chose Ntf3 because this neurotrophin regulates the formation of IHC-SGN synapses in the neonatal period. We now show that triggering Ntf3 overexpression by IHC supporting cells starting in middle age rapidly increases the amplitude of sound-evoked neural potentials compared with age-matched controls, indicating that Ntf3 produces a positive effect on cochlear function when the pathology is minimal. Furthermore, near the end of their lifespan, Ntf3-overexpressing mice have milder ARHL, with larger sound-evoked potentials along the ascending auditory pathway and reduced IHC synaptopathy compared with age-matched controls. Our results also provide evidence that an age-related decrease in cochlear Ntf3 expression contributes to ARHL and that Ntf3 supplementation could serve as a therapeutic for this prevalent disorder. Furthermore, these findings suggest that factors that regulate synaptogenesis during development could prevent age-related synaptopathy in the brain, a process involved in several central nervous system degenerative disorders.
Collapse
Affiliation(s)
- Luis R. Cassinotti
- Kresge Hearing Research Institute and Department of Otolaryngology‐Head and Neck SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Lingchao Ji
- Kresge Hearing Research Institute and Department of Otolaryngology‐Head and Neck SurgeryUniversity of MichiganAnn ArborMichiganUSA
- Present address:
Peking University Shenzhen HospitalShenzhenChina
| | - Beatriz C. Borges
- Kresge Hearing Research Institute and Department of Otolaryngology‐Head and Neck SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Nathan D. Cass
- Kresge Hearing Research Institute and Department of Otolaryngology‐Head and Neck SurgeryUniversity of MichiganAnn ArborMichiganUSA
- Present address:
The Otology Group of Vanderbilt, Vanderbilt University Medical Center Department of OtolaryngologyNashvilleTennesseeUSA
| | - Aditi S. Desai
- Kresge Hearing Research Institute and Department of Otolaryngology‐Head and Neck SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - David C. Kohrman
- Kresge Hearing Research Institute and Department of Otolaryngology‐Head and Neck SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - M. Charles Liberman
- Eaton‐Peabody Laboratories, Massachusetts Eye & Ear, and Department of Otolaryngology, Head and Neck SurgeryHarvard Medical SchoolBostonMassachusettsUSA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Department of Otolaryngology‐Head and Neck SurgeryUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
29
|
Bertoglio D, Zajicek F, Lombaerde SD, Miranda A, Stroobants S, Wang Y, Dominguez C, Munoz-Sanjuan I, Bard J, Liu L, Verhaeghe J, Staelens S. Validation, kinetic modeling, and test-retest reproducibility of [ 18F]SynVesT-1 for PET imaging of synaptic vesicle glycoprotein 2A in mice. J Cereb Blood Flow Metab 2022; 42:1867-1878. [PMID: 35570828 PMCID: PMC9536120 DOI: 10.1177/0271678x221101648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Alterations in synaptic vesicle glycoprotein 2 A (SV2A) have been associated with several neuropsychiatric and neurodegenerative disorders. Therefore, SV2A positron emission tomography (PET) imaging may provide a unique tool to investigate synaptic density dynamics during disease progression and after therapeutic intervention. This study aims to extensively characterize the novel radioligand [18F]SynVesT-1 for preclinical applications. In C57Bl/6J mice (n = 39), we assessed the plasma profile of [18F]SynVesT-1, validated the use of a noninvasive image-derived input function (IDIF) compared to an arterial input function (AIF), performed a blocking study with levetiracetam (50 and 200 mg/kg, i.p.) to verify the specificity towards SV2A, examined kinetic models for volume of distribution (VT) quantification, and explored test-retest reproducibility of [18F]SynVesT-1 in the central nervous system (CNS). Plasma availability of [18F]SynVesT-1 decreased rapidly (13.4 ± 1.5% at 30 min post-injection). VT based on AIF and IDIF showed excellent agreement (r2 = 0.95, p < 0.0001) and could be reliably estimated with a 60-min acquisition. The blocking study resulted in a complete blockade with no suitable reference region. Test-retest analysis indicated good reproducibility (mean absolute variability <10%). In conclusion, [18F]SynVesT-1 is selective for SV2A with optimal kinetics representing a candidate tool to quantify CNS synaptic density non-invasively.
Collapse
Affiliation(s)
- Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Franziska Zajicek
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Stef De Lombaerde
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Alan Miranda
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium.,Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Yuchuan Wang
- CHDI Management/CHDI Foundation, Los Angeles, California, USA
| | - Celia Dominguez
- CHDI Management/CHDI Foundation, Los Angeles, California, USA
| | | | - Jonathan Bard
- CHDI Management/CHDI Foundation, Los Angeles, California, USA
| | - Longbin Liu
- CHDI Management/CHDI Foundation, Los Angeles, California, USA
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
30
|
Li M, Cheng W, Zhang L, Zhou C, Peng X, Yu S, Zhang W. Novel Roles of RNA m6A Methylation Regulators in the Occurrence of Alzheimer’s Disease and the Subtype Classification. Int J Mol Sci 2022; 23:ijms231810766. [PMID: 36142676 PMCID: PMC9504232 DOI: 10.3390/ijms231810766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common forms of dementia, closely related to epigenetic factors. N6-methyladenosine (m6A) is the most abundant RNA modification, affecting the pathogenesis and development of neurodegenerative diseases. This study was the first exploration of the combined role of 25 common m6A RNA methylation regulators in AD through the integrated bioinformatics approaches. The 14 m6A regulators related to AD were selected by analyzing differences between AD patients and normal controls. Based on the selected m6A regulators, AD patients could be well classified into two m6A models using consensus clustering. The two clusters of patients had different immune profiles, and m6A regulators were associated with the components of immune cells. Additionally, there were 19 key AD genes obtained by screening differential genes through weighted gene co-expression network and least absolute shrinkage and selection operator regression analysis, which were highly associated with important m6A regulators during the occurrence of AD. More interestingly, NOTCH2 and NME1 could be potential targets for m6A regulation of AD. Taken together, these findings indicate that dysregulation of m6A methylation affects the occurrence of AD and is vital for the subtype classification and immune infiltration of AD.
Collapse
|
31
|
Bauer CS, Cohen RN, Sironi F, Livesey MR, Gillingwater TH, Highley JR, Fillingham DJ, Coldicott I, Smith EF, Gibson YB, Webster CP, Grierson AJ, Bendotti C, De Vos KJ. An interaction between synapsin and C9orf72 regulates excitatory synapses and is impaired in ALS/FTD. Acta Neuropathol 2022; 144:437-464. [PMID: 35876881 PMCID: PMC9381633 DOI: 10.1007/s00401-022-02470-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/17/2022] [Accepted: 07/08/2022] [Indexed: 12/16/2022]
Abstract
Dysfunction and degeneration of synapses is a common feature of amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD). A GGGGCC hexanucleotide repeat expansion in the C9ORF72 gene is the main genetic cause of ALS/FTD (C9ALS/FTD). The repeat expansion leads to reduced expression of the C9orf72 protein. How C9orf72 haploinsufficiency contributes to disease has not been resolved. Here we identify the synapsin family of synaptic vesicle proteins, the most abundant group of synaptic phosphoproteins, as novel interactors of C9orf72 at synapses and show that C9orf72 plays a cell-autonomous role in the regulation of excitatory synapses. We mapped the interaction of C9orf72 and synapsin to the N-terminal longin domain of C9orf72 and the conserved C domain of synapsin, and show interaction of the endogenous proteins in synapses. Functionally, C9orf72 deficiency reduced the number of excitatory synapses and decreased synapsin levels at remaining synapses in vitro in hippocampal neuron cultures and in vivo in the hippocampal mossy fibre system of C9orf72 knockout mice. Consistent with synaptic dysfunction, electrophysiological recordings identified impaired excitatory neurotransmission and network function in hippocampal neuron cultures with reduced C9orf72 expression, which correlated with a severe depletion of synaptic vesicles from excitatory synapses in the hippocampus of C9orf72 knockout mice. Finally, neuropathological analysis of post-mortem sections of C9ALS/FTD patient hippocampus with C9orf72 haploinsufficiency revealed a marked reduction in synapsin, indicating that disruption of the interaction between C9orf72 and synapsin may contribute to ALS/FTD pathobiology. Thus, our data show that C9orf72 plays a cell-autonomous role in the regulation of neurotransmission at excitatory synapses by interaction with synapsin and modulation of synaptic vesicle pools, and identify a novel role for C9orf72 haploinsufficiency in synaptic dysfunction in C9ALS/FTD.
Collapse
Affiliation(s)
- Claudia S Bauer
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Rebecca N Cohen
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Francesca Sironi
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Matthew R Livesey
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Hugh Robson Building, Edinburgh, EH8 9XD, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Chancellor's Building, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Daniel J Fillingham
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Ian Coldicott
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Emma F Smith
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Yolanda B Gibson
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Christopher P Webster
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Kurt J De Vos
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK.
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
32
|
Sanchez Avila A, Henstridge C. Array tomography: 15 years of synaptic analysis. Neuronal Signal 2022; 6:NS20220013. [PMID: 36187224 PMCID: PMC9512143 DOI: 10.1042/ns20220013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
Synapses are minuscule, intricate structures crucial for the correct communication between neurons. In the 125 years since the term synapse was first coined, we have advanced a long way when it comes to our understanding of how they work and what they do. Most of the fundamental discoveries have been invariably linked to advances in technology. However, due to their size, delicate structural integrity and their sheer number, our knowledge of synaptic biology has remained somewhat elusive and their role in neurodegenerative diseases still remains largely unknown. Here, we briefly discuss some of the imaging technologies used to study synapses and focus on the utility of the high-resolution imaging technique array tomography (AT). We introduce the AT technique and highlight some of the ways it is utilised with a particular focus on its power for analysing synaptic composition and pathology in human post-mortem tissue. We also discuss some of the benefits and drawbacks of techniques for imaging synapses and highlight some recent advances in the study of form and function by combining physiology and high-resolution synaptic imaging.
Collapse
Affiliation(s)
- Anna Sanchez Avila
- Euan Macdonald Centre for Motor Neuron Disease, Edinburgh, UK
- Division of Cellular and Systems Medicine, University of Dundee, Dundee, UK
| | - Christopher M. Henstridge
- Euan Macdonald Centre for Motor Neuron Disease, Edinburgh, UK
- Division of Cellular and Systems Medicine, University of Dundee, Dundee, UK
| |
Collapse
|
33
|
Chronic exposure of alcohol triggers microglia-mediated synaptic elimination inducing cognitive impairment. Exp Neurol 2022; 353:114061. [DOI: 10.1016/j.expneurol.2022.114061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/10/2022] [Accepted: 03/27/2022] [Indexed: 11/21/2022]
|
34
|
Kocurova G, Ricny J, Ovsepian SV. Autoantibodies targeting neuronal proteins as biomarkers for neurodegenerative diseases. Theranostics 2022; 12:3045-3056. [PMID: 35547759 PMCID: PMC9065204 DOI: 10.7150/thno.72126] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/09/2022] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are associated with the accumulation of a range of misfolded proteins across the central nervous system and related autoimmune responses, including the generation of antibodies and the activation of immune cells. Both innate and adaptive immunity become mobilized, leading to cellular and humoral effects. The role of humoral immunity in disease onset and progression remains to be elucidated with rising evidence suggestive of positive (protection, repair) and negative (injury, toxicity) outcomes. In this study, we review advances in research of neuron-targeting autoantibodies in the most prevalent NDDs. We discuss their biological origin, molecular diversity and changes in the course of diseases, consider their relevance to the initiation and progression of pathology as well as diagnostic and prognostic significance. It is suggested that the emerging autoimmune aspects of NDDs not only could facilitate the early detection but also might help to elucidate previously unknown facets of pathobiology with relevance to the development of precision medicine.
Collapse
Affiliation(s)
- Gabriela Kocurova
- Experimental Neurobiology Program, National Institute of Mental Health, Klecany, Czech Republic
| | - Jan Ricny
- Experimental Neurobiology Program, National Institute of Mental Health, Klecany, Czech Republic
| | - Saak V. Ovsepian
- Faculty of Science and Engineering, University of Greenwich London, Chatham Maritime, Kent, ME4 4TB, United Kingdom
| |
Collapse
|
35
|
Design, Synthesis, and Evaluation of Novel 2 H-Benzo[b][1,4]thiazin-3(4 H)-one Derivatives as New Acetylcholinesterase Inhibitors. Molecules 2022; 27:molecules27072121. [PMID: 35408519 PMCID: PMC9000418 DOI: 10.3390/molecules27072121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD) is a slowly progressive neurodegenerative disease that causes dementia in people aged 65 and over. In the present study, a series of thiadiazole hybrid compounds with benzothiazine derivatives as acetylcholinesterase inhibitors were developed and evaluated for their biological activity. The AChE and BChE inhibition potentials of all compounds were evaluated by using the in vitro Ellman method. The biological evaluation showed that compounds 3i and 3j displayed significant inhibitory activity against AChE. Compounds 3i and 3j showed IC50 values of 0.027 µM and 0.025 µM against AChE, respectively. The reference drug donepezil (IC50 = 0.021 µM) also showed significant inhibition against AChE. Further docking simulation also revealed that these compounds (3i and 3j) interacted with the active site of the enzyme similarly to donepezil. The antioxidant study revealed that compounds 3i and 3j exhibited greater antioxidant effects. An in vitro blood–brain barrier permeability study showed that compounds 3i and 3j are promising compounds against AD. The cytotoxicity study of compounds 3i and 3j showed non-cytotoxic with an IC50 value of 98.29 ± 3.98 µM and 159.68 ± 5.53 µM against NIH/3T3 cells, respectively.
Collapse
|
36
|
Bao J, Jegede SL, Hawks JW, Dade B, Guan Q, Middaugh S, Qiu Z, Levina A, Tsai TH. Detecting Cochlear Synaptopathy Through Curvature Quantification of the Auditory Brainstem Response. Front Cell Neurosci 2022; 16:851500. [PMID: 35356798 PMCID: PMC8959412 DOI: 10.3389/fncel.2022.851500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The sound-evoked electrical compound potential known as auditory brainstem response (ABR) represents the firing of a heterogenous population of auditory neurons in response to sound stimuli, and is often used for clinical diagnosis based on wave amplitude and latency. However, recent ABR applications to detect human cochlear synaptopathy have led to inconsistent results, mainly due to the high variability of ABR wave-1 amplitude. Here, rather than focusing on the amplitude of ABR wave 1, we evaluated the use of ABR wave curvature to detect cochlear synaptic loss. We first compared four curvature quantification methods using simulated ABR waves, and identified that the cubic spline method using five data points produced the most accurate quantification. We next evaluated this quantification method with ABR data from an established mouse model with cochlear synaptopathy. The data clearly demonstrated that curvature measurement is more sensitive and consistent in identifying cochlear synaptic loss in mice compared to the amplitude and latency measurements. We further tested this curvature method in a different mouse model presenting with otitis media. The change in curvature profile due to middle ear infection in otitis media is different from the profile of mice with cochlear synaptopathy. Thus, our study suggests that curvature quantification can be used to address the current ABR variability issue, and may lead to additional applications in the clinic diagnosis of hearing disorders.
Collapse
Affiliation(s)
- Jianxin Bao
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH, United States
- *Correspondence: Jianxin Bao,
| | - Segun Light Jegede
- Department of Mathematical Sciences, Kent State University, Kent, OH, United States
| | - John W. Hawks
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH, United States
| | - Bethany Dade
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH, United States
| | - Qiang Guan
- Department of Computer Science, Kent State University, Kent, OH, United States
| | - Samantha Middaugh
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Ziyu Qiu
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH, United States
| | - Anna Levina
- Department of Mathematical Sciences, Kent State University, Kent, OH, United States
| | - Tsung-Heng Tsai
- Department of Mathematical Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
37
|
Yuan M, Wang Y, Wen J, Jing F, Zou Q, Pu Y, Pan T, Cai Z. Dietary Salt Disrupts Tricarboxylic Acid Cycle and Induces Tau Hyperphosphorylation and Synapse Dysfunction during Aging. Aging Dis 2022; 13:1532-1545. [PMID: 36186135 PMCID: PMC9466974 DOI: 10.14336/ad.2022.0220] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/20/2022] [Indexed: 11/05/2022] Open
Abstract
Dietary salt causes synaptic deficits and tau hyperphosphorylation, which are detrimental to cognitive function. However, the specific effects of a high-salt diet on synapse and tau protein remain poorly understood. In this study, aged (15-month-old) C57BL/6 mice received a normal (0.5% NaCl) or high-salt (8% NaCl) diet for 3 months, and N2a cells were treated with normal culture medium or a NaCl medium (40 mM). Spatial learning and memory abilities were tested using the Morris water maze. The levels of metabolites and related enzymes in the tricarboxylic acid (TCA) cycle were confirmed using liquid chromatography-tandem mass spectrometry, western blotting, and immunofluorescence. We also investigated synapse morphology and the phosphorylation of tau protein. Under the high-salt diet, mice displayed impaired learning and memory compared to mice fed the normal diet. Furthermore, excessive salt intake disturbed the TCA cycle in both animals and cells compared to the respective normal controls. High dietary salt reduced postsynaptic density protein 95 (PSD95) and brain-derived neurotrophic factor (BDNF) expression, impaired neurons, and caused synaptic loss in the mice. We also detected tau hyperphosphorylation at different sites (Thr205, Thr231, and Thr181) without increasing total tau levels in response to high salt treatment, both in vivo and in vitro. We concluded that elevated salt intake impairs the TCA cycle and induces tau hyperphosphorylation and synapse dysfunction during aging, which ultimately results in cognitive impairment.
Collapse
Affiliation(s)
- Minghao Yuan
- Chongqing Medical University, Chongqing, China.
- Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.
- Department of Neurology, Chongqing General Hospital, Chongqing, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China.
| | - Yangyang Wang
- Department of Neurology, Chongqing General Hospital, Chongqing, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China.
| | - Jie Wen
- Guangdong Medical University, Guangdong, China.
| | - Feng Jing
- Chongqing Medical University, Chongqing, China.
- Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.
- Department of Neurology, Chongqing General Hospital, Chongqing, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China.
| | - Qian Zou
- Department of Neurology, Chongqing General Hospital, Chongqing, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China.
| | - Yinshuang Pu
- Department of Neurology, Chongqing General Hospital, Chongqing, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China.
| | - Tingyu Pan
- Department of Neurology, Chongqing General Hospital, Chongqing, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China.
| | - Zhiyou Cai
- Chongqing Medical University, Chongqing, China.
- Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.
- Department of Neurology, Chongqing General Hospital, Chongqing, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China.
- Correspondence should be addressed to: Dr. Zhiyou Cai, Department of Neurology, Chongqing Medical University, Chongqing, Chongqing, 400016, China. .
| |
Collapse
|
38
|
Systemic LPS-induced microglial activation results in increased GABAergic tone: A mechanism of protection against neuroinflammation in the medial prefrontal cortex in mice. Brain Behav Immun 2022; 99:53-69. [PMID: 34582995 DOI: 10.1016/j.bbi.2021.09.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation with excess microglial activation and synaptic dysfunction are early symptoms of most neurological diseases. However, how microglia-associated neuroinflammation regulates synaptic activity remains obscure. We report here that acute neuroinflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) results in cell-type-specific increases in inhibitory postsynaptic currents in the glutamatergic, but not the GABAergic, neurons of medial prefrontal cortex (mPFC), coinciding with excessive microglial activation. LPS causes upregulation in levels of GABAAR subunits, glutamine synthetase and vesicular GABA transporter, and downregulation in brain-derived neurotrophic factor (BDNF) and its receptor, pTrkB. Blockage of microglial activation by minocycline ameliorates LPS-induced abnormal expression of GABA signaling-related proteins and activity of synaptic and network. Moreover, minocycline prevents the mice from LPS-induced aberrant behavior, such as a reduction in total distance and time spent in the centre in the open field test; decreases in entries into the open arm of elevated-plus maze and in consumption of sucrose; increased immobility in the tail suspension test. Furthermore, upregulation of GABA signaling by tiagabine also prevents LPS-induced microglial activation and aberrant behavior. This study illustrates a mode of bidirectional constitutive signaling between the neural and immune compartments of the brain, and suggests that the mPFC is an important area for brain-immune system communication. Moreover, the present study highlights GABAergic signaling as a key therapeutic target for mitigating neuroinflammation-induced abnormal synaptic activity in the mPFC, together with the associated behavioral abnormalities.
Collapse
|
39
|
Ginsberg SD, Joshi S, Sharma S, Guzman G, Wang T, Arancio O, Chiosis G. The penalty of stress - Epichaperomes negatively reshaping the brain in neurodegenerative disorders. J Neurochem 2021; 159:958-979. [PMID: 34657288 PMCID: PMC8688321 DOI: 10.1111/jnc.15525] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/22/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023]
Abstract
Adaptation to acute and chronic stress and/or persistent stressors is a subject of wide interest in central nervous system disorders. In this context, stress is an effector of change in organismal homeostasis and the response is generated when the brain perceives a potential threat. Herein, we discuss a nuanced and granular view whereby a wide variety of genotoxic and environmental stressors, including aging, genetic risk factors, environmental exposures, and age- and lifestyle-related changes, act as direct insults to cellular, as opposed to organismal, homeostasis. These two concepts of how stressors impact the central nervous system are not mutually exclusive. We discuss how maladaptive stressor-induced changes in protein connectivity through epichaperomes, disease-associated pathologic scaffolds composed of tightly bound chaperones, co-chaperones, and other factors, impact intracellular protein functionality altering phenotypes, that in turn disrupt and remodel brain networks ranging from intercellular to brain connectome levels. We provide an evidence-based view on how these maladaptive changes ranging from stressor to phenotype provide unique precision medicine opportunities for diagnostic and therapeutic development, especially in the context of neurodegenerative disorders including Alzheimer's disease where treatment options are currently limited.
Collapse
Affiliation(s)
- Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, USA
- Departments of Psychiatry, Neuroscience & Physiology, the NYU Neuroscience Institute, New York University Grossman School of Medicine, New York City, New York, USA
| | - Suhasini Joshi
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Gianny Guzman
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Tai Wang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York City, New York, USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York City, New York, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| |
Collapse
|
40
|
Simon C, Soga T, Okano HJ, Parhar I. α-Synuclein-mediated neurodegeneration in Dementia with Lewy bodies: the pathobiology of a paradox. Cell Biosci 2021; 11:196. [PMID: 34798911 PMCID: PMC8605528 DOI: 10.1186/s13578-021-00709-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is epitomized by the pathognomonic manifestation of α-synuclein-laden Lewy bodies within selectively vulnerable neurons in the brain. By virtue of prion-like inheritance, the α-synuclein protein inexorably undergoes extensive conformational metamorphoses and culminate in the form of fibrillar polymorphs, instigating calamitous damage to the brain's neuropsychological networks. This epiphenomenon is nebulous, however, by lingering uncertainty over the quasi "pathogenic" behavior of α-synuclein conformers in DLB pathobiology. Despite numerous attempts, a monolithic "α-synuclein" paradigm that is able to untangle the enigma enshrouding the clinicopathological spectrum of DLB has failed to emanate. In this article, we review conceptual frameworks of α-synuclein dependent cell-autonomous and non-autonomous mechanisms that are likely to facilitate the transneuronal spread of degeneration through the neuraxis. In particular, we describe how the progressive demise of susceptible neurons may evolve from cellular derangements perpetrated by α-synuclein misfolding and aggregation. Where pertinent, we show how these bona fide mechanisms may mutually accentuate α-synuclein-mediated neurodegeneration in the DLB brain.
Collapse
Affiliation(s)
- Christopher Simon
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Tomoko Soga
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Hirotaka James Okano
- Division of Regenerative Medicine, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
41
|
Crapser JD, Arreola MA, Tsourmas KI, Green KN. Microglia as hackers of the matrix: sculpting synapses and the extracellular space. Cell Mol Immunol 2021; 18:2472-2488. [PMID: 34413489 PMCID: PMC8546068 DOI: 10.1038/s41423-021-00751-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia shape the synaptic environment in health and disease, but synapses do not exist in a vacuum. Instead, pre- and postsynaptic terminals are surrounded by extracellular matrix (ECM), which together with glia comprise the four elements of the contemporary tetrapartite synapse model. While research in this area is still just beginning, accumulating evidence points toward a novel role for microglia in regulating the ECM during normal brain homeostasis, and such processes may, in turn, become dysfunctional in disease. As it relates to synapses, microglia are reported to modify the perisynaptic matrix, which is the diffuse matrix that surrounds dendritic and axonal terminals, as well as perineuronal nets (PNNs), specialized reticular formations of compact ECM that enwrap neuronal subsets and stabilize proximal synapses. The interconnected relationship between synapses and the ECM in which they are embedded suggests that alterations in one structure necessarily affect the dynamics of the other, and microglia may need to sculpt the matrix to modify the synapses within. Here, we provide an overview of the microglial regulation of synapses, perisynaptic matrix, and PNNs, propose candidate mechanisms by which these structures may be modified, and present the implications of such modifications in normal brain homeostasis and in disease.
Collapse
Affiliation(s)
- Joshua D. Crapser
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Miguel A. Arreola
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kate I. Tsourmas
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kim N. Green
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| |
Collapse
|
42
|
Trujillo-Estrada L, Vanderklish PW, Nguyen MMT, Kuang RR, Nguyen C, Huynh E, da Cunha C, Javonillo DI, Forner S, Martini AC, Sarraf ST, Simmon VF, Baglietto-Vargas D, LaFerla FM. SPG302 Reverses Synaptic and Cognitive Deficits Without Altering Amyloid or Tau Pathology in a Transgenic Model of Alzheimer's Disease. Neurotherapeutics 2021; 18:2468-2483. [PMID: 34738197 PMCID: PMC8804111 DOI: 10.1007/s13311-021-01143-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/04/2022] Open
Abstract
Alzheimer's disease (AD) is conceptualized as a synaptic failure disorder in which loss of glutamatergic synapses is a major driver of cognitive decline. Thus, novel therapeutic strategies aimed at regenerating synapses may represent a promising approach to mitigate cognitive deficits in AD patients. At present, no disease-modifying drugs exist for AD, and approved therapies are palliative at best, lacking in the ability to reverse the synaptic failure. Here, we tested the efficacy of a novel synaptogenic small molecule, SPG302 - a 3rd-generation benzothiazole derivative that increases the density of axospinous glutamatergic synapses - in 3xTg-AD mice. Daily dosing of 3xTg-AD mice with SPG302 at 3 and 30 mg/kg (i.p.) for 4 weeks restored hippocampal synaptic density and improved cognitive function in hippocampal-dependent tasks. Mushroom and stubby spine profiles were increased by SPG302, and associated with enhanced expression of key postsynaptic proteins - including postsynaptic density protein 95 (PSD95), drebrin, and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) - and increased colocalization of PSD95 with synaptophysin. Notably, SPG302 proved efficacious in this model without modifying Aβ and tau pathology. Thus, our study provides preclinical support for the idea that compounds capable of restoring synaptic density offer a viable strategy to reverse cognitive decline in AD.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Peter W Vanderklish
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - Marie Minh Thu Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Run Rong Kuang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Caroline Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Eric Huynh
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Dominic Ibarra Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Alessandra C Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stella T Sarraf
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA
| | - Vincent F Simmon
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain.
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
| |
Collapse
|
43
|
Tremblay MÈ. Microglial functional alteration and increased diversity in the challenged brain: Insights into novel targets for intervention. Brain Behav Immun Health 2021; 16:100301. [PMID: 34589793 PMCID: PMC8474548 DOI: 10.1016/j.bbih.2021.100301] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) parenchyma, which perform beneficial physiological roles across life. These immune cells actively maintain CNS health by clearing toxic debris and removing dysfunctional or degenerating cells. They also modify the wiring of neuronal circuits, by acting on the formation, modification, and elimination of synapses-the connections between neurons. Microglia furthermore recently emerged as highly diverse cells comprising several structural and functional states, indicating a far more critical involvement in orchestrating brain development, plasticity, behaviour, and cognition. Various environmental factors, together with the individual genetic predispositions, confer an increased risk for neurodevelopmental and neuropsychiatric disorders, as well as neurodegenerative diseases that include autism spectrum disorders, schizophrenia, major depressive disorder, and Alzheimer's disease, across life. Microglia are highly sensitive to chronic psychological stress, inadequate diet, viral/bacterial infection, pollution, and insufficient or altered sleep, especially during critical developmental periods, but also throughout life. These environmental challenges can compromise microglial physiological functions, resulting notably in defective neuronal circuit wiring, altered brain functional connectivity, and the onset of behavioral deficits into adolescence, adulthood, and aging. This short review provides a historical and technical perspective, notably focused on my contribution to the field, on how environmental challenges affect microglia, particularly their physiological functions, and increase their diversity, which provides novel targets for intervention.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Molecular Medicine Department, Université Laval, Québec City, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- The Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
44
|
Perić I, Costina V, Djordjević S, Gass P, Findeisen P, Inta D, Borgwardt S, Filipović D. Tianeptine modulates synaptic vesicle dynamics and favors synaptic mitochondria processes in socially isolated rats. Sci Rep 2021; 11:17747. [PMID: 34493757 PMCID: PMC8423821 DOI: 10.1038/s41598-021-97186-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 08/09/2021] [Indexed: 11/09/2022] Open
Abstract
Deregulation of synaptic function and neurotransmission has been linked with the development of major depression disorder (MDD). Tianeptine (Tian) has been used as antidepressant with anxiolytic properties and recently as a nootropic to improve cognitive performance, but its mechanism of action is unknown. We conducted a proteomic study on the hippocampal synaptosomal fractions of adult male Wistar rats exposed to chronic social isolation (CSIS, 6 weeks), an animal model of depression and after chronic Tian treatment in controls (nootropic effect) and CSIS-exposed rats (lasting 3 weeks of 6-week CSIS) (therapeutic effect). Increased expression of Syn1 and Camk2-related neurotransmission, vesicle transport and energy processes in Tian-treated controls were found. CSIS led to upregulation of proteins associated with actin cytoskeleton, signaling transduction and glucose metabolism. In CSIS rats, Tian up-regulated proteins involved in mitochondrial energy production, mitochondrial transport and dynamics, antioxidative defense and glutamate clearance, while attenuating the CSIS-increased glycolytic pathway and cytoskeleton organization proteins expression and decreased the expression of proteins involved in V-ATPase and vesicle endocytosis. Our overall findings revealed that synaptic vesicle dynamics, specifically exocytosis, and mitochondria-related energy processes might be key biological pathways modulated by the effective nootropic and antidepressant treatment with Tian and be a potential target for therapeutic efficacy of the stress-related mood disorders.
Collapse
Affiliation(s)
- Ivana Perić
- Department of Molecular Biology and Endocrinology, "VINČA", Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Victor Costina
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, 68159, Mannheim, Germany
| | | | - Peter Gass
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159, Mannheim, Germany
| | - Peter Findeisen
- Institute for Clinical Chemistry, Medical Faculty Mannheim of the University of Heidelberg, University Hospital Mannheim, 68159, Mannheim, Germany
| | - Dragoš Inta
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Stefan Borgwardt
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Dragana Filipović
- Department of Molecular Biology and Endocrinology, "VINČA", Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
45
|
Genomic Aberrations Associated with the Pathophysiological Mechanisms of Neurodevelopmental Disorders. Cells 2021; 10:cells10092317. [PMID: 34571966 PMCID: PMC8470284 DOI: 10.3390/cells10092317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 12/27/2022] Open
Abstract
Genomic studies are increasingly revealing that neurodevelopmental disorders are caused by underlying genomic alterations. Chromosomal microarray testing has been used to reliably detect minute changes in genomic copy numbers. The genes located in the aberrated regions identified in patients with neurodevelopmental disorders may be associated with the phenotypic features. In such cases, haploinsufficiency is considered to be the mechanism, when the deletion of a gene is related to neurodevelopmental delay. The loss-of-function mutation in such genes may be evaluated using next-generation sequencing. On the other hand, the patients with increased copy numbers of the genes may exhibit different clinical symptoms compared to those with loss-of-function mutation in the genes. In such cases, the additional copies of the genes are considered to have a dominant negative effect, inducing cell stress. In other cases, not the copy number changes, but mutations of the genes are responsible for causing the clinical symptoms. This can be explained by the dominant negative effects of the gene mutations. Currently, the diagnostic yield of genomic alterations using comprehensive analysis is less than 50%, indicating the existence of more subtle alterations or genomic changes in the untranslated regions. Copy-neutral inversions and insertions may be related. Hence, better analytical algorithms specialized for the detection of such alterations are required for higher diagnostic yields.
Collapse
|
46
|
Cano-Astorga N, DeFelipe J, Alonso-Nanclares L. Three-Dimensional Synaptic Organization of Layer III of the Human Temporal Neocortex. Cereb Cortex 2021; 31:4742-4764. [PMID: 33999122 PMCID: PMC8408440 DOI: 10.1093/cercor/bhab120] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In the present study, we have used focused ion beam/scanning electron microscopy (FIB/SEM) to perform a study of the synaptic organization of layer III of Brodmann's area 21 in human tissue samples obtained from autopsies and biopsies. We analyzed the synaptic density, 3D spatial distribution, and type (asymmetric/symmetric), as well as the size and shape of each synaptic junction of 4945 synapses that were fully reconstructed in 3D. Significant differences in the mean synaptic density between autopsy and biopsy samples were found (0.49 and 0.66 synapses/μm3, respectively). However, in both types of samples (autopsy and biopsy), the asymmetric:symmetric ratio was similar (93:7) and most asymmetric synapses were established on dendritic spines (75%), while most symmetric synapses were established on dendritic shafts (85%). We also compared several electron microscopy methods and analysis tools to estimate the synaptic density in the same brain tissue. We have shown that FIB/SEM is much more reliable and robust than the majority of the other commonly used EM techniques. The present work constitutes a detailed description of the synaptic organization of cortical layer III. Further studies on the rest of the cortical layers are necessary to better understand the functional organization of this temporal cortical region.
Collapse
Affiliation(s)
- Nicolás Cano-Astorga
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid 28031, Spain
| | - Lidia Alonso-Nanclares
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28223, Spain
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid 28031, Spain
| |
Collapse
|
47
|
Maestú F, de Haan W, Busche MA, DeFelipe J. Neuronal excitation/inhibition imbalance: core element of a translational perspective on Alzheimer pathophysiology. Ageing Res Rev 2021; 69:101372. [PMID: 34029743 DOI: 10.1016/j.arr.2021.101372] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/16/2021] [Accepted: 05/19/2021] [Indexed: 02/08/2023]
Abstract
Our incomplete understanding of the link between Alzheimer's Disease pathology and symptomatology is a crucial obstacle for therapeutic success. Recently, translational studies have begun to connect the dots between protein alterations and deposition, brain network dysfunction and cognitive deficits. Disturbance of neuronal activity, and in particular an imbalance in underlying excitation/inhibition (E/I), appears early in AD, and can be regarded as forming a central link between structural brain pathology and cognitive dysfunction. While there are emerging (non-)pharmacological options to influence this imbalance, the complexity of human brain dynamics has hindered identification of an optimal approach. We suggest that focusing on the integration of neurophysiological aspects of AD at the micro-, meso- and macroscale, with the support of computational network modeling, can unite fundamental and clinical knowledge, provide a general framework, and suggest rational therapeutic targets.
Collapse
|
48
|
Kurucu H, Colom-Cadena M, Davies C, Wilkins L, King D, Rose J, Tzioras M, Tulloch JH, Smith C, Spires-Jones TL. Inhibitory synapse loss and accumulation of amyloid beta in inhibitory presynaptic terminals in Alzheimer's disease. Eur J Neurol 2021; 29:1311-1323. [PMID: 34331352 DOI: 10.1111/ene.15043] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Synapse degeneration in Alzheimer's disease (AD) correlates strongly with cognitive decline. There is well-established excitatory synapse loss in AD with known contributions of pathological amyloid beta (Aβ) to excitatory synapse dysfunction and loss. Despite clear changes in circuit excitability in AD and model systems, relatively little is known about pathology in inhibitory synapses. METHODS Here human postmortem brain samples (n = 5 control, 10 AD cases) from temporal and occipital cortices were examined to investigate whether inhibitory synapses and neurons are lost in AD and whether Aβ may contribute to inhibitory synapse degeneration. Inhibitory neurons were counted in all six cortical layers using stereology software, and array tomography was used to examine synapse density and the accumulation of Aβ in synaptic terminals. RESULTS Differing inhibitory neuron densities were observed in the different cortical layers. The highest inhibitory neuron density was observed in layer 4 in both brain regions and the visual cortex had a higher inhibitory neuron density than the temporal cortex. There was significantly lower inhibitory neuron density in AD than in control cases in all six cortical layers. High-resolution array tomography imaging revealed plaque-associated loss of inhibitory synapses and accumulation of Aβ in a small subset of inhibitory presynaptic terminals with the most accumulation near amyloid plaques. CONCLUSIONS Inhibitory neuron and synapse loss in AD may contribute to disrupted excitatory/inhibitory balance and cognitive decline. Future work is warranted to determine whether targeting inhibitory synapse loss could be a useful therapeutic strategy.
Collapse
Affiliation(s)
- Hatice Kurucu
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Martí Colom-Cadena
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Caitlin Davies
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Lewis Wilkins
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Declan King
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Jamie Rose
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Makis Tzioras
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Jane H Tulloch
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences and Sudden Death Brain Bank, University of Edinburgh, Edinburgh, UK
| | - Tara L Spires-Jones
- University of Edinburgh Centre for Discovery Brain Sciences and UK Dementia Research Institute, Edinburgh, UK
| |
Collapse
|
49
|
Modulation of Brain-Derived Neurotrophic Factor (BDNF) Signaling Pathway by Culinary Sage ( Salvia officinalis L.). Int J Mol Sci 2021; 22:ijms22147382. [PMID: 34299002 PMCID: PMC8303624 DOI: 10.3390/ijms22147382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 01/19/2023] Open
Abstract
Culinary sage (Salvia officinalis L.) is a common spice plant in the mint family (Lamiaceae) well known for its distinctive culinary and traditional medicinal uses. Sage tea has been used traditionally as a brain-enhancing tonic and extracts from sage have been reported to have both cognitive and memory enhancing effects. Brain-derived neurotrophic factor (BDNF) is an endogenous signaling molecule involved in cognition and memory function. In this study, activity-guided fractionation employing preparative reverse-phase high performance liquid chromatography (RP-HPLC) of culinary sage extracts led to the discovery of benzyl 6-O-β-D-apiofuranosyl-β-D-glucoside (B6AG) as a natural product that upregulates transcription of neurotrophic factors in C6 glioma cells. Purified B6AG showed a moderate dose response, with upregulation of BDNF and with EC50 at 6.46 μM. To better understand the natural variation in culinary sage, B6AG was quantitated in the leaves of several commercial varieties by liquid chromatography-mass spectrometry (LC-MS). The level of B6AG in dried culinary sage was found to range from 334 ± 14 to 698 ± 65 μg/g. This study provided a foundation for future investigations, including quantitative inquiries on the distribution of B6AG within the different plant organs, explorations in optimizing post-harvest practices, and aid in the development of sage varieties with elevated levels of B6AG.
Collapse
|
50
|
Grosso Jasutkar H, Yamamoto A. Do Changes in Synaptic Autophagy Underlie the Cognitive Impairments in Huntington's Disease? J Huntingtons Dis 2021; 10:227-238. [PMID: 33780373 PMCID: PMC8293641 DOI: 10.3233/jhd-200466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although Huntington's disease (HD) is classically considered from the perspective of the motor syndrome, the cognitive changes in HD are prominent and often an early manifestation of disease. As such, investigating the underlying pathophysiology of cognitive changes may give insight into important and early neurodegenerative events. In this review, we first discuss evidence from both HD patients and animal models that cognitive changes correlate with early pathological changes at the synapse, an observation that is similarly made in other neurodegenerative conditions that primarily affect cognition. We then describe how autophagy plays a critical role supporting synaptic maintenance in the healthy brain, and how autophagy dysfunction in HD may thereby lead to impaired synaptic maintenance and thus early manifestations of disease.
Collapse
Affiliation(s)
| | - Ai Yamamoto
- Department of Neurology, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|