1
|
Carra S, Fabian B, Taghavi H, Milanetti E, Giliberti V, Ruocco G, Shepherd J, Vendruscolo M, Fuxreiter M. Virus-like particles of retroviral origin in protein aggregation and neurodegenerative diseases. Mol Aspects Med 2025; 103:101369. [PMID: 40398193 DOI: 10.1016/j.mam.2025.101369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 05/06/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
A wide range of human diseases are associated with protein misfolding and amyloid aggregates. Recent studies suggest that in certain neurological disorders, including Amyotrophic Lateral Sclerosis (ALS), Frontotemporal Dementia (FTD) and various tauopathies, protein aggregation may be promoted by virus-like particles (VLPs) formed by endogenous retroviruses (ERVs). The molecular mechanisms by which these VLPs contribute to protein aggregation, however, remain enigmatic. Here, we discuss possible molecular mechanisms of ERV-derived VLPs in the formation and spread of protein aggregates. An intriguing possibility is that liquid-like condensates may facilitate the formation of both protein aggregates and ERV-derived VLPs. We also describe how RNA chaperoning, and the encapsulation and trafficking of misfolded proteins, may contribute to protein homeostasis through the elimination of protein aggregates from cells. Based on these insights, we discuss future potential therapeutic opportunities.
Collapse
Affiliation(s)
- Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Balazs Fabian
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Hamed Taghavi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Edoardo Milanetti
- Department of Physics, Sapienza University, Rome, Italy; Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Valeria Giliberti
- Department of Physics, Sapienza University, Rome, Italy; Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Giancarlo Ruocco
- Department of Physics, Sapienza University, Rome, Italy; Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | | | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Monika Fuxreiter
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Physics and Astronomy, University of Padova, Padova, Italy.
| |
Collapse
|
2
|
Tyagi M, Chadha R, de Hoog E, Sullivan KR, Walker AC, Northrop A, Fabian B, Fuxreiter M, Hyman BT, Shepherd JD. Arc mediates intercellular tau transmission via extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619703. [PMID: 39484489 PMCID: PMC11526995 DOI: 10.1101/2024.10.22.619703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Intracellular neurofibrillary tangles that consist of misfolded tau protein1 cause neurodegeneration in Alzheimer's disease (AD) and frontotemporal dementia (FTD). Tau pathology spreads cell-to-cell2 but the exact mechanisms of tau release and intercellular transmission remain poorly defined. Tau is released from neurons as free protein or in extracellular vesicles (EVs)3-5 but the role of these different release mechanisms in intercellular tau transmission is unclear. Here, we show that the neuronal gene Arc is critical for packaging tau into EVs. Brain EVs purified from human tau (hTau) transgenic rTg4510 mice (rTgWT) contain high levels of hTau that are capable of seeding tau pathology. In contrast, EVs purified from rTgWT crossed with Arc knock-out mice (rTgArc KO) have significantly less hTau and cannot seed tau aggregation. Arc facilitates the release of hTau in EVs produced via the I-BAR protein IRSp53, but not free tau. Arc protein directly binds hTau to form a fuzzy complex that we identified in both mouse and human brain tissue. We find that pathological intracellular hTau accumulates in neurons in rTgArc KO mice, which correlates with accelerated neuron loss in the hippocampus. Finally, we find that intercellular tau transmission is significantly abrogated in Arc KO mice. We conclude that Arc-dependent release of tau in EVs plays a significant role in intracellular tau elimination and intercellular tau transmission.
Collapse
Affiliation(s)
- Mitali Tyagi
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Radhika Chadha
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Eric de Hoog
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | | | - Alicia C. Walker
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Ava Northrop
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Balazs Fabian
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Germany
| | - Monika Fuxreiter
- Department of Biomedical Sciences University of Padova, Padova, Italy
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | |
Collapse
|
3
|
Da Silva SJ, Cabral-Castro MJ, Gonçalves CCA, Mariani D, Ferreira O, Tanuri A, Puccioni-Sohler M. Challenges in the Diagnosis of SARS-CoV-2 Infection in the Nervous System. Viruses 2024; 16:1247. [PMID: 39205221 PMCID: PMC11359543 DOI: 10.3390/v16081247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Neurological involvement has been widely reported in SARS-CoV-2 infection. However, viral identification in the cerebrospinal fluid (CSF) is rarely found. The aim of this study is to evaluate the accuracy of virological and immunological biomarkers in CSF for the diagnosis of neuroCOVID-19. We analyzed 69 CSF samples from patients with neurological manifestations: 14 with suspected/confirmed COVID-19, with 5 additional serial CSF samples (group A), and as a control, 50 non-COVID-19 cases (group B-26 with other neuroinflammatory diseases; group C-24 with non-inflammatory diseases). Real-time reverse-transcription polymerase chain reaction (real-time RT-PCR) was used to determine SARS-CoV-2, and specific IgG, IgM, neopterin, and protein 10 induced by gamma interferon (CXCL-10) were evaluated in the CSF samples. No samples were amplified for SARS-CoV-2 by real-time RT-PCR. The sensitivity levels of anti-SARS-CoV-2 IgG and IgM were 50% and 14.28%, respectively, with 100% specificity for both tests. CXCL-10 showed high sensitivity (95.83%) and specificity (95.83%) for detection of neuroinflammation. Serial CSF analysis showed an association between the neuroinflammatory biomarkers and outcome (death and hospital discharge) in two cases (meningoencephalitis and rhombencephalitis). The detection of SARS-CoV-2 RNA and specific immunoglobulins in the CSF can be used for neuroCOVID-19 confirmation. Additionally, CXCL-10 in the CSF may contribute to the diagnosis and monitoring of neuroCOVID-19.
Collapse
Affiliation(s)
- Samya Jezine Da Silva
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
| | - Mauro Jorge Cabral-Castro
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Niterói 24220-900, Brazil
- Laboratório de Líquido Cefalorraquidiano, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
| | - Cássia Cristina Alves Gonçalves
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Diana Mariani
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Orlando Ferreira
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Amílcar Tanuri
- Laboratório de Virologia Molecular, Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Marzia Puccioni-Sohler
- Programa de Pós-Graduação em Doenças Infecciosas e Parasitárias, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-853, Brazil
- Laboratório de Líquido Cefalorraquidiano, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, Brazil
- Departamento de Medicina Geral, Escola de Medicina e Cirurgia, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro 22290-250, Brazil
| |
Collapse
|
4
|
Godoy Muñoz JM, Neset L, Markússon S, Weber S, Krokengen OC, Sutinen A, Christakou E, Lopez AJ, Bramham CR, Kursula P. Structural characterization of two nanobodies targeting the ligand-binding pocket of human Arc. PLoS One 2024; 19:e0300453. [PMID: 38683783 PMCID: PMC11057775 DOI: 10.1371/journal.pone.0300453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/27/2024] [Indexed: 05/02/2024] Open
Abstract
The activity-regulated cytoskeleton-associated protein (Arc) is a complex regulator of synaptic plasticity in glutamatergic neurons. Understanding its molecular function is key to elucidate the neurobiology of memory and learning, stress regulation, and multiple neurological and psychiatric diseases. The recent development of anti-Arc nanobodies has promoted the characterization of the molecular structure and function of Arc. This study aimed to validate two anti-Arc nanobodies, E5 and H11, as selective modulators of the human Arc N-lobe (Arc-NL), a domain that mediates several molecular functions of Arc through its peptide ligand binding site. The structural characteristics of recombinant Arc-NL-nanobody complexes were solved at atomic resolution using X-ray crystallography. Both anti-Arc nanobodies bind specifically to the multi-peptide binding site of Arc-NL. Isothermal titration calorimetry showed that the Arc-NL-nanobody interactions occur at nanomolar affinity, and that the nanobodies can displace a TARPγ2-derived peptide from the binding site. Thus, both anti-Arc-NL nanobodies could be used as competitive inhibitors of endogenous Arc ligands. Differences in the CDR3 loops between the two nanobodies indicate that the spectrum of short linear motifs recognized by the Arc-NL should be expanded. We provide a robust biochemical background to support the use of anti-Arc nanobodies in attempts to target Arc-dependent synaptic plasticity. Function-blocking anti-Arc nanobodies could eventually help unravel the complex neurobiology of synaptic plasticity and allow to develop diagnostic and treatment tools.
Collapse
Affiliation(s)
| | - Lasse Neset
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Sarah Weber
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Aleksi Sutinen
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| | | | - Andrea J. Lopez
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Petri Kursula
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
5
|
Schulz L, Ramirez P, Lemieux A, Gonzalez E, Thomson T, Frost B. Tau-Induced Elevation of the Activity-Regulated Cytoskeleton Associated Protein Arc1 Causally Mediates Neurodegeneration in the Adult Drosophila Brain. Neuroscience 2023; 518:101-111. [PMID: 35487302 PMCID: PMC9606145 DOI: 10.1016/j.neuroscience.2022.04.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease and other tauopathies are neurodegenerative disorders pathologically defined by aggregated forms of tau protein in the brain. While synaptic degradation is a well-established feature of tau-induced neurotoxicity, the underlying mechanisms of how pathogenic forms of tau drive synaptic dysfunction are incompletely understood. Synaptic function and subsequent memory consolidation are dependent upon synaptic plasticity, the ability of synapses to adjust their structure and strength in response to changes in activity. The activity regulated cytoskeleton associated protein ARC acts in the nucleus and at postsynaptic densities to regulate various forms of synaptic plasticity. ARC harbors a retrovirus-like Gag domain that facilitates ARC multimerization and capsid formation. Trans-synaptic transfer of RNA-containing ARC capsids is required for synaptic plasticity. While ARC is elevated in brains of patients with Alzheimer's disease and genetic variants in ARC increase susceptibility to Alzheimer's disease, mechanistic insight into the role of ARC in Alzheimer's disease is lacking. Using a Drosophila model of tauopathy, we find that pathogenic tau significantly increases multimeric species of the protein encoded by the Drosophila homolog of ARC, Arc1, in the adult fly brain. We find that Arc1 is elevated within nuclei and the neuropil of tau transgenic Drosophila, but does not localize to synaptic vesicles or presynaptic terminals. Lastly, we find that genetic manipulation of Arc1 modifies tau-induced neurotoxicity, suggesting that tau-induced Arc1 elevation mediates neurodegeneration. Taken together, our results suggest that ARC elevation in human Alzheimer's disease is a consequence of tau pathology and is a causal factor contributing to neuronal death.
Collapse
Affiliation(s)
- Lulu Schulz
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, United States; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, United States; Department of Cell Systems and Anatomy, San Antonio, TX, United States; University of Texas Health San Antonio, San Antonio, TX, United States
| | - Paulino Ramirez
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, United States; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, United States; Department of Cell Systems and Anatomy, San Antonio, TX, United States; University of Texas Health San Antonio, San Antonio, TX, United States
| | - Adrienne Lemieux
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Elias Gonzalez
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, United States; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, United States; Department of Cell Systems and Anatomy, San Antonio, TX, United States; University of Texas Health San Antonio, San Antonio, TX, United States
| | - Travis Thomson
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Bess Frost
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, United States; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, United States; Department of Cell Systems and Anatomy, San Antonio, TX, United States; University of Texas Health San Antonio, San Antonio, TX, United States.
| |
Collapse
|
6
|
Dysregulated Signaling at Postsynaptic Density: A Systematic Review and Translational Appraisal for the Pathophysiology, Clinics, and Antipsychotics' Treatment of Schizophrenia. Cells 2023; 12:cells12040574. [PMID: 36831241 PMCID: PMC9954794 DOI: 10.3390/cells12040574] [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: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Emerging evidence from genomics, post-mortem, and preclinical studies point to a potential dysregulation of molecular signaling at postsynaptic density (PSD) in schizophrenia pathophysiology. The PSD that identifies the archetypal asymmetric synapse is a structure of approximately 300 nm in diameter, localized behind the neuronal membrane in the glutamatergic synapse, and constituted by more than 1000 proteins, including receptors, adaptors, kinases, and scaffold proteins. Furthermore, using FASS (fluorescence-activated synaptosome sorting) techniques, glutamatergic synaptosomes were isolated at around 70 nm, where the receptors anchored to the PSD proteins can diffuse laterally along the PSD and were stabilized by scaffold proteins in nanodomains of 50-80 nm at a distance of 20-40 nm creating "nanocolumns" within the synaptic button. In this context, PSD was envisioned as a multimodal hub integrating multiple signaling-related intracellular functions. Dysfunctions of glutamate signaling have been postulated in schizophrenia, starting from the glutamate receptor's interaction with scaffolding proteins involved in the N-methyl-D-aspartate receptor (NMDAR). Despite the emerging role of PSD proteins in behavioral disorders, there is currently no systematic review that integrates preclinical and clinical findings addressing dysregulated PSD signaling and translational implications for antipsychotic treatment in the aberrant postsynaptic function context. Here we reviewed a critical appraisal of the role of dysregulated PSD proteins signaling in the pathophysiology of schizophrenia, discussing how antipsychotics may affect PSD structures and synaptic plasticity in brain regions relevant to psychosis.
Collapse
|
7
|
Kim YK, Jung YS, Song J. Transcriptome Profile in the Mouse Brain of Hepatic Encephalopathy and Alzheimer's Disease. Int J Mol Sci 2022; 24:ijms24010675. [PMID: 36614117 PMCID: PMC9821016 DOI: 10.3390/ijms24010675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 01/03/2023] Open
Abstract
Hepatic encephalopathy (HE) is a chronic metabolic disease accompanied by neuropathological and neuropsychiatric features, including memory deficits, psychomotor dysfunction, depression, and anxiety. Alzheimer's disease (AD), the most common neurodegenerative disease, is characterized by tau hyperphosphorylation, excessive amyloid beta (Aβ) accumulation, the formation of fibrillary tangles, hippocampus atrophy, and neuroinflammation. Recent studies have suggested a positive correlation between HE and AD. Some studies reported that an impaired cholesterol pathway, abnormal bile acid secretion, excessive ammonia level, impaired Aβ clearance, astrocytic dysfunction, and abnormal γ-aminobutyric acid GABAergic neuronal signaling in HE may also be involved in AD pathology. However, the mechanisms and related genes involved in AD-like pathology in the HE brain are unclear. Thus, we compared the cortical transcriptome profile between an HE mouse model, bile duct ligation (BDL), and an AD mouse model, the 5×FAD. Our study showed that the expression of many genes implicated in HE is associated with neuronal dysfunction in AD mice. We found changes in various protein-coding RNAs, implicated in synapses, neurogenesis, neuron projection, neuron differentiation, and neurite outgrowth, and non-coding RNAs possibly associated with neuropathology. Our data provide an important resource for further studies to elucidate AD-like pathophysiology in HE patients.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Yoon Seok Jung
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea
- Correspondence: ; Tel.: +82-61-379-2706; Fax: +82-61-375-5834
| |
Collapse
|
8
|
Myrum C, Moreno-Castilla P, Rapp PR. 'Arc'-hitecture of normal cognitive aging. Ageing Res Rev 2022; 80:101678. [PMID: 35781092 PMCID: PMC9378697 DOI: 10.1016/j.arr.2022.101678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 06/10/2022] [Accepted: 06/24/2022] [Indexed: 12/17/2022]
Abstract
Arc is an effector immediate-early gene that is critical for forming long-term memories. Since its discovery 25 years ago, it has repeatedly surprised us with a number of intriguing properties, including the transport of its mRNA to recently-activated synapses, its master role in bidirectionally regulating synaptic strength, its evolutionary retroviral origins, its ability to mediate intercellular transfer between neurons via extracellular vesicles (EVs), and its exceptional regulation-both temporally and spatially. The current review discusses how Arc has been used as a tool to identify the neural networks involved in cognitive aging and how Arc itself may contribute to cognitive outcome in aging. In addition, we raise several outstanding questions, including whether Arc-containing EVs in peripheral blood might provide a noninvasive biomarker for memory-related synaptic failure in aging, and whether rectifying Arc dysregulation is likely to be an effective strategy for bending the arc of aging toward successful cognitive outcomes.
Collapse
Affiliation(s)
- Craig Myrum
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Perla Moreno-Castilla
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Peter R Rapp
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
9
|
Leung HW, Foo G, VanDongen A. Arc Regulates Transcription of Genes for Plasticity, Excitability and Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081946. [PMID: 36009494 PMCID: PMC9405677 DOI: 10.3390/biomedicines10081946] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
The immediate early gene Arc is a master regulator of synaptic function and a critical determinant of memory consolidation. Here, we show that Arc interacts with dynamic chromatin and closely associates with histone markers for active enhancers and transcription in cultured rat hippocampal neurons. Both these histone modifications, H3K27Ac and H3K9Ac, have recently been shown to be upregulated in late-onset Alzheimer’s disease (AD). When Arc induction by pharmacological network activation was prevented using a short hairpin RNA, the expression profile was altered for over 1900 genes, which included genes associated with synaptic function, neuronal plasticity, intrinsic excitability, and signalling pathways. Interestingly, about 100 Arc-dependent genes are associated with the pathophysiology of AD. When endogenous Arc expression was induced in HEK293T cells, the transcription of many neuronal genes was increased, suggesting that Arc can control expression in the absence of activated signalling pathways. Taken together, these data establish Arc as a master regulator of neuronal activity-dependent gene expression and suggest that it plays a significant role in the pathophysiology of AD.
Collapse
Affiliation(s)
| | - Gabriel Foo
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Antonius VanDongen
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Correspondence:
| |
Collapse
|
10
|
Transcriptomic and Proteomic Analysis of CRISPR/Cas9-Mediated ARC-Knockout HEK293 Cells. Int J Mol Sci 2022; 23:ijms23094498. [PMID: 35562887 PMCID: PMC9101110 DOI: 10.3390/ijms23094498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/15/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
Arc/Arg3.1 (activity-regulated cytoskeletal-associated protein (ARC)) is a critical regulator of long-term synaptic plasticity and is involved in the pathophysiology of schizophrenia. The functions and mechanisms of human ARC action are poorly understood and worthy of further investigation. To investigate the function of the ARC gene in vitro, we generated an ARC-knockout (KO) HEK293 cell line via CRISPR/Cas9-mediated gene editing and conducted RNA sequencing and label-free LC-MS/MS analysis to identify the differentially expressed genes and proteins in isogenic ARC-KO HEK293 cells. Furthermore, we used bioluminescence resonance energy transfer (BRET) assays to detect interactions between the ARC protein and differentially expressed proteins. Genetic deletion of ARC disturbed multiple genes involved in the extracellular matrix and synaptic membrane. Seven proteins (HSPA1A, ENO1, VCP, HMGCS1, ALDH1B1, FSCN1, and HINT2) were found to be differentially expressed between ARC-KO cells and ARC wild-type cells. BRET assay results showed that ARC interacted with PSD95 and HSPA1A. Overall, we found that ARC regulates the differential expression of genes involved in the extracellular matrix, synaptic membrane, and heat shock protein family. The transcriptomic and proteomic profiles of ARC-KO HEK293 cells presented here provide new evidence for the mechanisms underlying the effects of ARC and molecular pathways involved in schizophrenia pathophysiology.
Collapse
|
11
|
mRNA Trafficking in the Nervous System: A Key Mechanism of the Involvement of Activity-Regulated Cytoskeleton-Associated Protein (Arc) in Synaptic Plasticity. Neural Plast 2021; 2021:3468795. [PMID: 34603440 PMCID: PMC8486535 DOI: 10.1155/2021/3468795] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/15/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
Synaptic activity mediates information storage and memory consolidation in the brain and requires a fast de novo synthesis of mRNAs in the nucleus and proteins in synapses. Intracellular localization of a protein can be achieved by mRNA trafficking and localized translation. Activity-regulated cytoskeleton-associated protein (Arc) is a master regulator of synaptic plasticity and plays an important role in controlling large signaling networks implicated in learning, memory consolidation, and behavior. Transcription of the Arc gene may be induced by a short behavioral event, resulting in synaptic activation. Arc mRNA is exported into the cytoplasm and can be trafficked into the dendrite of an activated synapse where it is docked and translated. The structure of Arc is similar to the viral GAG (group-specific antigen) protein, and phylogenic analysis suggests that Arc may originate from the family of Ty3/Gypsy retrotransposons. Therefore, Arc might evolve through “domestication” of retroviruses. Arc can form a capsid-like structure that encapsulates a retrovirus-like sentence in the 3′-UTR (untranslated region) of Arc mRNA. Such complex can be loaded into extracellular vesicles and transported to other neurons or muscle cells carrying not only genetic information but also regulatory signals within neuronal networks. Therefore, Arc mRNA inter- and intramolecular trafficking is essential for the modulation of synaptic activity required for memory consolidation and cognitive functions. Recent studies with single-molecule imaging in live neurons confirmed and extended the role of Arc mRNA trafficking in synaptic plasticity.
Collapse
|
12
|
Sun GC, Lee YJ, Lee YC, Yu HF, Wang DC. Exercise prevents the impairment of learning and memory in prenatally phthalate-exposed male rats by improving the expression of plasticity-related proteins. Behav Brain Res 2021; 413:113444. [PMID: 34245761 DOI: 10.1016/j.bbr.2021.113444] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022]
Abstract
Regular exercise has been identified to facilitate neuroplasticity that maximize functional outcome after brain injuries. Brain-derived neurotrophic factor (BDNF) has emerged as a key facilitator of neuroplasticity after exercise. The activity-regulated cytoskeleton associated protein (Arc) is induced by BDNF and N-methyl-d-aspartic acid receptor (NMDAR), contributing to functional modification of neuroplasticity in the hippocampus. Meanwhile, early-life exposure to neuroendocrine disruptor di-(2-ethylhexyl)-phthalate (DEHP) is a risk factor for behavioral deficits, but the mechanisms responsible for DEHP-induced neurotoxicity are not well understood. The purpose of this study is to investigate whether hippocampal Arc expression is impaired by DEHP exposure and to examine the protective role of exercise in the prenatally DEHP-exposed male rats. Sprague Dawley dams were fed with vehicle or DEHP during gestation. The male offspring were trained to treadmill running for 5 weeks followed by examination of behavioral and biochemical outcomes. The results showed that DEHP-exposed rats exhibited impairment of spatial learning and memory as well as down-regulations of BDNF, NMDAR, Arc, and synaptophysin. Importantly, aerobic exercise during childhood-adolescence prevented the impairment of learning and memory by recovering the expressions of BDNF, NMDAR, Arc, and synaptophysin. These findings suggest that exercise may provide beneficial effects on ameliorating the impairment of neuroplasticity in the prenatally DEHP-exposed male rats at late adolescence.
Collapse
Affiliation(s)
- Gwo-Ching Sun
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Yi-Ju Lee
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Yi-Ching Lee
- Department of Food and Beverage Services, Tainan University of Technology, Tainan, Taiwan.
| | - Hsien-Fu Yu
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Dean-Chuan Wang
- Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
13
|
Przybyla M, van Eersel J, van Hummel A, van der Hoven J, Sabale M, Harasta A, Müller J, Gajwani M, Prikas E, Mueller T, Stevens CH, Power J, Housley GD, Karl T, Kassiou M, Ke YD, Ittner A, Ittner LM. Onset of hippocampal network aberration and memory deficits in P301S tau mice are associated with an early gene signature. Brain 2021; 143:1889-1904. [PMID: 32375177 DOI: 10.1093/brain/awaa133] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/10/2020] [Accepted: 03/04/2020] [Indexed: 01/30/2023] Open
Abstract
Hyperphosphorylation and deposition of tau in the brain characterizes frontotemporal dementia and Alzheimer's disease. Disease-associated mutations in the tau-encoding MAPT gene have enabled the generation of transgenic mouse models that recapitulate aspects of human neurodegenerative diseases, including tau hyperphosphorylation and neurofibrillary tangle formation. Here, we characterized the effects of transgenic P301S mutant human tau expression on neuronal network function in the murine hippocampus. Onset of progressive spatial learning deficits in P301S tau transgenic TAU58/2 mice were paralleled by long-term potentiation deficits and neuronal network aberrations during electrophysiological and EEG recordings. Gene-expression profiling just prior to onset of apparent deficits in TAU58/2 mice revealed a signature of immediate early genes that is consistent with neuronal network hypersynchronicity. We found that the increased immediate early gene activity was confined to neurons harbouring tau pathology, providing a cellular link between aberrant tau and network dysfunction. Taken together, our data suggest that tau pathology drives neuronal network dysfunction through hyperexcitation of individual, pathology-harbouring neurons, thereby contributing to memory deficits.
Collapse
Affiliation(s)
- Magdalena Przybyla
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Janet van Eersel
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Annika van Hummel
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Julia van der Hoven
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Miheer Sabale
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Anne Harasta
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Julius Müller
- Genome Informatics at Molecular Health GmbH, Heidelberg, Germany
| | - Mehul Gajwani
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia.,Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Emmanuel Prikas
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Thomas Mueller
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Claire H Stevens
- School of Chemistry and Molecular Bioscience, University of Wollongong and the Illawarra Health and Medical Research Institute, Wollongong, NSW, 2522, Australia
| | - John Power
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Gary D Housley
- Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Tim Karl
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Michael Kassiou
- School of Chemistry, University of Sydney, Sydney, NSW, Australia
| | - Yazi D Ke
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Arne Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| | - Lars M Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, 2109, NSW, Australia
| |
Collapse
|
14
|
Unger MS, Li E, Scharnagl L, Poupardin R, Altendorfer B, Mrowetz H, Hutter-Paier B, Weiger TM, Heneka MT, Attems J, Aigner L. CD8 + T-cells infiltrate Alzheimer's disease brains and regulate neuronal- and synapse-related gene expression in APP-PS1 transgenic mice. Brain Behav Immun 2020; 89:67-86. [PMID: 32479993 DOI: 10.1016/j.bbi.2020.05.070] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
Neuroinflammation is a major contributor to disease progression in Alzheimer's disease (AD) and is characterized by the activity of brain resident glial cells, in particular microglia cells. However, there is increasing evidence that peripheral immune cells infiltrate the brain at certain stages of AD progression and shape disease pathology. We recently identified CD8+ T-cells in the brain parenchyma of APP-PS1 transgenic mice being tightly associated with microglia as well as with neuronal structures. The functional role of CD8+ T-cells in the AD brain is however completely unexplored. Here, we demonstrate increased numbers of intra-parenchymal CD8+ T-cells in human AD post-mortem hippocampus, which was replicated in APP-PS1 mice. Also, aged WT mice show a remarkable infiltration of CD8+ T-cells, which was more pronounced and had an earlier onset in APP-PS1 mice. To address their functional relevance in AD, we successfully ablated the pool of CD8+ T-cells in the blood, spleen and brain from 12 months-old APP-PS1 and WT mice for a total of 4 weeks using an anti-CD8 antibody treatment. While the treatment at this time of disease stage did neither affect the cognitive outcome nor plaque pathology, RNAseq analysis of the hippocampal transcriptome from APP-PS1 mice lacking CD8+ T-cells revealed highly altered neuronal- and synapse-related gene expression including an up-regulation for neuronal immediate early genes (IEGs) such as the Activity Regulated Cytoskeleton Associated Protein (Arc) and the Neuronal PAS Domain Protein 4 (Npas4). Gene ontology enrichment analysis illustrated that the biological processes "regulation of neuronal synaptic plasticity" and the cellular components "postsynapses" were over-represented upon CD8+ T-cell ablation. Additionally, Kegg pathway analysis showed up-regulated pathways for "calcium signaling", "long-term potentiation", "glutamatergic synapse" and "axon guidance". Therefore, we conclude that CD8+ T-cells infiltrate the aged and AD brain and that brain CD8+ T-cells might directly contribute to neuronal dysfunction in modulating synaptic plasticity. Further analysis will be essential to uncover the exact mechanism of how CD8+ T-cells modulate the neuronal landscape and thereby contribute to AD pathology.
Collapse
Affiliation(s)
- M S Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - E Li
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - L Scharnagl
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - R Poupardin
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria; Experimental and Clinical Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - B Altendorfer
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - H Mrowetz
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | | | - T M Weiger
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - M T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany
| | - J Attems
- Translational and Clinical Institute, Newcastle University, Newcastle upon Tyne, UK
| | - L Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria; Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Austria.
| |
Collapse
|
15
|
Arbo B, Cechinel L, Palazzo R, Siqueira I. Endosomal dysfunction impacts extracellular vesicle release: Central role in Aβ pathology. Ageing Res Rev 2020; 58:101006. [PMID: 31891813 DOI: 10.1016/j.arr.2019.101006] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 12/19/2019] [Accepted: 12/26/2019] [Indexed: 01/04/2023]
Abstract
Alzheimer's Disease (AD) is characterized by progressive loss of cognitive abilities; senile plaques represent the major histopathological findings. Amyloid precursor protein (APP) processing machinery, and its product amyloid-beta (Aβ) peptide, have been found in extracellular vesicles (EVs), specifically exosomes, which allows for Aβ peptide aggregation and subsequent senile plaques deposition. We review the APP processing imbalance in EVs, autophagic and endosomal pathways in AD. Increased intraluminal vesicle (ILV) production and exosome release appear to counteract the endosomal dysfunction of APP processing; however, this process results in elevated amyloidogenic processing of APP and augmented senile plaque deposition. Several players related to APP processing and dysfunctional endosomal-lysosomal-exosomal (and other EVs) pathway are described, and the interconnected systems are discussed. The components Arc, p75, Rab11 and retromer complex emerge as candidates for key convergent mechanisms that lead to increased EVs loaded with APP machinery and Aβ levels, in atrophy and damage of basal forebrain cholinergic neurons in AD.
Collapse
|
16
|
Penrod RD, Thomsen M, Taniguchi M, Guo Y, Cowan CW, Smith LN. The activity-regulated cytoskeleton-associated protein, Arc/Arg3.1, influences mouse cocaine self-administration. Pharmacol Biochem Behav 2020; 188:172818. [PMID: 31682894 PMCID: PMC7202920 DOI: 10.1016/j.pbb.2019.172818] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/01/2019] [Accepted: 10/30/2019] [Indexed: 01/04/2023]
Abstract
The activity-regulated cytoskeleton-associated protein (Arc, also known as Arg3.1), an immediate early gene and synaptic regulator, is upregulated following a single cocaine exposure. However, there is not much known regarding Arc/Arg3.1's potential contribution to addiction-relevant behaviors. Despite known learning and memory deficits in contextual fear and water-maze reversal learning tasks, we find that mice lacking Arc/Arg3.1 perform conditioned place preference and operant conditioning involving positive reinforcers (food and cocaine) with little-to-no impairment. However, following normal saline-extinction, wild type (WT) mice show a classic inverted-U dose-response function, while Arc/Arg3.1 knockout (KO) mice fail to adjust their intake across multiple doses. Importantly, Arc/Arg3.1 KO and WT mice behave comparably on an increasing cost task (FR1-FR3; acquisition dose), providing evidence that both groups find cocaine reinforcing. Differences in individuals that drive variations in use patterns and particularly, drug intake levels, are critical as they influence the likelihood of developing dependence. Our data suggest that Arc/Arg3.1 may contribute to addiction as a regulator of drug-taking vulnerability under different drug availability conditions.
Collapse
Affiliation(s)
- Rachel D Penrod
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Morgane Thomsen
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Makoto Taniguchi
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Yuhong Guo
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, United States of America; Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Christopher W Cowan
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America
| | - Laura N Smith
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, United States of America; Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA 02478, United States of America.
| |
Collapse
|
17
|
Wilkerson JR, Albanesi JP, Huber KM. Roles for Arc in metabotropic glutamate receptor-dependent LTD and synapse elimination: Implications in health and disease. Semin Cell Dev Biol 2017; 77:51-62. [PMID: 28969983 DOI: 10.1016/j.semcdb.2017.09.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 10/18/2022]
Abstract
The Arc gene is robustly transcribed in specific neural ensembles in response to experience-driven activity. Upon induction, Arc mRNA is transported to dendrites, where it can be rapidly and locally translated by activation of metabotropic glutamate receptors (mGluR1/5). mGluR-induced dendritic synthesis of Arc is implicated in weakening or elimination of excitatory synapses by triggering endocytosis of postsynaptic AMPARs in both hippocampal CA1 and cerebellar Purkinje neurons. Importantly, CA1 neurons with experience-induced Arc mRNA are susceptible, or primed for mGluR-induced long-term synaptic depression (mGluR-LTD). Here we review mechanisms and function of Arc in mGluR-LTD and synapse elimination and propose roles for these forms of plasticity in Arc-dependent formation of sparse neural representations of learned experience. We also discuss accumulating evidence linking dysregulation of Arc and mGluR-LTD in human cognitive disorders such as intellectual disability, autism and Alzheimer's disease.
Collapse
Affiliation(s)
- Julia R Wilkerson
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Joseph P Albanesi
- Departments of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Kimberly M Huber
- Departments of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
18
|
Perusini JN, Cajigas SA, Cohensedgh O, Lim SC, Pavlova IP, Donaldson ZR, Denny CA. Optogenetic stimulation of dentate gyrus engrams restores memory in Alzheimer's disease mice. Hippocampus 2017; 27:1110-1122. [PMID: 28667669 PMCID: PMC5610644 DOI: 10.1002/hipo.22756] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/24/2017] [Accepted: 06/19/2017] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by amyloid-beta (Aβ) plaques and tau neurofibrillary tangles. APPswe/PS1dE9 (APP/PS1) mice have been developed as an AD model and are characterized by plaque formation at 4-6 months of age. Here, we sought to better understand AD-related cognitive decline by characterizing various types of memory. In order to better understand how memory declines with AD, APP/PS1 mice were bred with ArcCreERT2 mice. In this line, neural ensembles activated during memory encoding can be indelibly tagged and directly compared with neural ensembles activated during memory retrieval (i.e., memory traces/engrams). We first administered a battery of tests examining depressive- and anxiety-like behaviors, as well as spatial, social, and cognitive memory to APP/PS1 × ArcCreERT2 × channelrhodopsin (ChR2)-enhanced yellow fluorescent protein (EYFP) mice. Dentate gyrus (DG) neural ensembles were then optogenetically stimulated in these mice to improve memory impairment. AD mice had the most extensive differences in fear memory, as assessed by contextual fear conditioning (CFC), which was accompanied by impaired DG memory traces. Optogenetic stimulation of DG neural ensembles representing a CFC memory increased memory retrieval in the appropriate context in AD mice when compared with control (Ctrl) mice. Moreover, optogenetic stimulation facilitated reactivation of the neural ensembles that were previously activated during memory encoding. These data suggest that activating previously learned DG memory traces can rescue cognitive impairments and point to DG manipulation as a potential target to treat memory loss commonly seen in AD.
Collapse
Affiliation(s)
- Jennifer N. Perusini
- Department of Psychiatry, Columbia University, New York, NY, U.S.A
- Division of Integrative Neuroscience, New York State Psychiatric Institute (NYSPI) / Research Foundation for Mental Hygiene, Inc. (RFMH), New York, NY, U.S.A
| | | | - Omid Cohensedgh
- Department of Psychiatry, Columbia University, New York, NY, U.S.A
| | - Sean C. Lim
- Division of Integrative Neuroscience, New York State Psychiatric Institute (NYSPI) / Research Foundation for Mental Hygiene, Inc. (RFMH), New York, NY, U.S.A
- Department of Neuroscience, MD-PhD Program, College of Physicians & Surgeons, Columbia University Medical Center, New York, NY, U.S.A
| | - Ina P. Pavlova
- Division of Integrative Neuroscience, New York State Psychiatric Institute (NYSPI) / Research Foundation for Mental Hygiene, Inc. (RFMH), New York, NY, U.S.A
| | - Zoe R. Donaldson
- Department of Psychiatry, Columbia University, New York, NY, U.S.A
- Division of Integrative Neuroscience, New York State Psychiatric Institute (NYSPI) / Research Foundation for Mental Hygiene, Inc. (RFMH), New York, NY, U.S.A
- Department of Molecular, Cellular, and Developmental Biology; Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, U.S.A
| | - Christine A. Denny
- Department of Psychiatry, Columbia University, New York, NY, U.S.A
- Division of Integrative Neuroscience, New York State Psychiatric Institute (NYSPI) / Research Foundation for Mental Hygiene, Inc. (RFMH), New York, NY, U.S.A
| |
Collapse
|
19
|
Nikolaienko O, Patil S, Eriksen MS, Bramham CR. Arc protein: a flexible hub for synaptic plasticity and cognition. Semin Cell Dev Biol 2017; 77:33-42. [PMID: 28890419 DOI: 10.1016/j.semcdb.2017.09.006] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/06/2017] [Accepted: 09/06/2017] [Indexed: 02/08/2023]
Abstract
Mammalian excitatory synapses express diverse types of synaptic plasticity. A major challenge in neuroscience is to understand how a neuron utilizes different types of plasticity to sculpt brain development, function, and behavior. Neuronal activity-induced expression of the immediate early protein, Arc, is critical for long-term potentiation and depression of synaptic transmission, homeostatic synaptic scaling, and adaptive functions such as long-term memory formation. However, the molecular basis of Arc protein function as a regulator of synaptic plasticity and cognition remains a puzzle. Recent work on the biophysical and structural properties of Arc, its protein-protein interactions and post-translational modifications have shed light on the issue. Here, we present Arc protein as a flexible, multifunctional and interactive hub. Arc interacts with specific effector proteins in neuronal compartments (dendritic spines, nuclear domains) to bidirectionally regulate synaptic strength by distinct molecular mechanisms. Arc stability, subcellular localization, and interactions are dictated by synaptic activity and post-translational modification of Arc. This functional versatility and context-dependent signaling supports a view of Arc as a highly specialized master organizer of long-term synaptic plasticity, critical for information storage and cognition.
Collapse
Affiliation(s)
- Oleksii Nikolaienko
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Sudarshan Patil
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Maria Steene Eriksen
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine and KG Jebsen Center for Neuropsychiatric Disorders, University of Bergen, Jonas Lies vei 91, N-5009, Bergen, Norway.
| |
Collapse
|
20
|
Bi R, Kong LL, Xu M, Li GD, Zhang DF, Li T, Fang Y, Zhang C, Zhang B, Yao YG. The Arc Gene Confers Genetic Susceptibility to Alzheimer’s Disease in Han Chinese. Mol Neurobiol 2017; 55:1217-1226. [DOI: 10.1007/s12035-017-0397-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/10/2017] [Indexed: 01/13/2023]
|
21
|
Chuang YA, Hu TM, Chen CH, Hsu SH, Tsai HY, Cheng MC. Rare mutations and hypermethylation of the ARC gene associated with schizophrenia. Schizophr Res 2016; 176:106-113. [PMID: 27464451 DOI: 10.1016/j.schres.2016.07.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/18/2016] [Accepted: 07/20/2016] [Indexed: 11/19/2022]
Abstract
Activity-regulated cytoskeleton-associated protein (ARC), which interacts with the N-methyl-d-aspartate receptor (NMDAR) complex, is a critical effector molecule downstream of multiple neuronal signaling pathways. Dysregulation of the ARC/NMDAR complex can disrupt learning, memory, and normal brain functions. This study examined the role of ARC in susceptibility to schizophrenia. We used a resequencing strategy to identify the variants of ARC in 1078 subjects, including patients with schizophrenia and normal controls. We identified 16 known SNPs and 27 rare mutations. SNP-based analysis showed no association of ARC with schizophrenia. In addition, the rare mutations did not increase the burden in patients compared with controls. However, one patient-specific allele in the putative ARC promoter region and seven patient-specific mutants in ARC exon regions significantly reduced the reporter gene activity compared with ARC wild-type. Methylation of a putative ARC promoter attenuated reporter activity in vitro, suggesting that ARC expression is regulated by DNA methylation. Pyrosequencing revealed eight hypermethylated CpG sites in the putative ARC promoter region in 64 schizophrenic patients compared with 63 controls. Taken together, our results suggest that both rare variants and epigenetic regulation of ARC contribute to the pathogenesis of schizophrenia in some patients.
Collapse
Affiliation(s)
- Yang-An Chuang
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Tsung-Ming Hu
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Chia-Hsiang Chen
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou and Department and Graduate School of Biomedical Sciences Chang Gung University, Taoyuan, Taiwan
| | - Shih-Hsin Hsu
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Hsin-Yao Tsai
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan
| | - Min-Chih Cheng
- Department of Psychiatry, Yuli Mental Health Research Center, Yuli Branch, Taipei Veterans General Hospital, Hualien, Taiwan; Center for General Education, St. Mary's Junior College of Medicine, Nursing and Management, Yilan County, Taiwan.
| |
Collapse
|
22
|
Tang AY. RNA processing-associated molecular mechanisms of neurodegenerative diseases. J Appl Genet 2015; 57:323-33. [DOI: 10.1007/s13353-015-0330-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/22/2015] [Accepted: 11/26/2015] [Indexed: 12/13/2022]
|
23
|
Stargardt A, Swaab DF, Bossers K. The storm before the quiet: neuronal hyperactivity and Aβ in the presymptomatic stages of Alzheimer's disease. Neurobiol Aging 2015; 36:1-11. [DOI: 10.1016/j.neurobiolaging.2014.08.014] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/29/2014] [Accepted: 08/12/2014] [Indexed: 12/27/2022]
|
24
|
Hsu WCJ, Nilsson CL, Laezza F. Role of the axonal initial segment in psychiatric disorders: function, dysfunction, and intervention. Front Psychiatry 2014; 5:109. [PMID: 25191280 PMCID: PMC4139700 DOI: 10.3389/fpsyt.2014.00109] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 08/06/2014] [Indexed: 12/22/2022] Open
Abstract
The progress of developing effective interventions against psychiatric disorders has been limited due to a lack of understanding of the underlying cellular and functional mechanisms. Recent research findings focused on exploring novel causes of psychiatric disorders have highlighted the importance of the axonal initial segment (AIS), a highly specialized neuronal structure critical for spike initiation of the action potential. In particular, the role of voltage-gated sodium channels, and their interactions with other protein partners in a tightly regulated macromolecular complex has been emphasized as a key component in the regulation of neuronal excitability. Deficits and excesses of excitability have been linked to the pathogenesis of brain disorders. Identification of the factors and regulatory pathways involved in proper AIS function, or its disruption, can lead to the development of novel interventions that target these mechanistic interactions, increasing treatment efficacy while reducing deleterious off-target effects for psychiatric disorders.
Collapse
Affiliation(s)
- Wei-Chun Jim Hsu
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Graduate Program in Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- M.D.–Ph.D. Combined Degree Program, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Carol Lynn Nilsson
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Sealy Center for Molecular Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Center for Addiction Research, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Center for Biomedical Engineering, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| |
Collapse
|
25
|
Mapping memory function in the medial temporal lobe with the immediate-early gene Arc. Behav Brain Res 2013; 254:22-33. [DOI: 10.1016/j.bbr.2013.04.048] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 04/27/2013] [Indexed: 12/29/2022]
|
26
|
Christensen DZ, Thomsen MS, Mikkelsen JD. Reduced basal and novelty-induced levels of activity-regulated cytoskeleton associated protein (Arc) and c-Fos mRNA in the cerebral cortex and hippocampus of APPswe/PS1ΔE9 transgenic mice. Neurochem Int 2013; 63:54-60. [PMID: 23598246 DOI: 10.1016/j.neuint.2013.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/26/2013] [Accepted: 04/06/2013] [Indexed: 02/04/2023]
Abstract
Activity-regulated cytoskeletal-associated protein (Arc) and c-Fos are immediate early gene (IEG) products induced by novelty in the hippocampus and involved in the consolidation of synaptic plasticity and long-term memory. We investigated whether induction of arc and c-fos after exposure to a novel open field environment was compromised in different neocortical areas and the hippocampal formation in APP/PS1ΔE9 transgenic mice characterized by pronounced accumulation and deposition of beta amyloid (Aβ). Notably, the basal level of Arc and c-fos mRNA in the neocortex was significantly lower in APP/PS1ΔE9 compared to wild-type mice. Novelty exposure induced an increase in Arc and c-Fos mRNA in the medial prefrontal cortex (mPFC), parietal cortex, and hippocampal formation in both APP/PS1ΔE9 transgenic and wild-type mice. However, novelty-induced IEG expression did not reach the same levels in APP/PS1ΔE9 as in the wild-type mice. In contrast, synaptophysin levels did not differ between mutant and wild type mice, suggesting that the observed effect was not due to a general decrease in the number of presynapses. These data suggest a reduction in basal and novelty-induced neuronal activity in a transgenic mouse model of Alzheimer's disease, which is most pronounced in cortical regions, indicating that a decreased functional response in IEG expression could be partly responsible for the cognitive deficits observed in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Ditte Z Christensen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Denmark
| | | | | |
Collapse
|