1
|
Sullivan KR, Ravens A, Walker AC, Shepherd JD. "Arc - A viral vector of memory and synaptic plasticity". Curr Opin Neurobiol 2025; 91:102979. [PMID: 39956025 PMCID: PMC11938376 DOI: 10.1016/j.conb.2025.102979] [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: 06/21/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 02/18/2025]
Abstract
Learning induces gene expression and memory consolidation requires new protein synthesis. Many of these activity-induced genes are transcription factors. One of the exceptions is a key immediate early gene, Arc, which has been implicated in several forms of synaptic plasticity and is critical for long-term memory formation. Recently, Arc was discovered to have retroviral properties, such as the ability to form virus-like capsids, that were repurposed from an ancient retrotransposon. Arc capsids are released in extracellular vesicles that mediate intercellular communication. Here, we review Arc's role in synaptic plasticity and propose a model for how Arc mediates memory consolidation via a novel intercellular non-cell autonomous form of long-term depression.
Collapse
Affiliation(s)
| | - Alicia Ravens
- Department of Neurobiology, University of Utah, United States
| | - Alicia C Walker
- Department of Neurobiology, University of Utah, United States
| | | |
Collapse
|
2
|
Baldinotti R, Pauzin FP, Fevang H, Ishizuka Y, Bramham CR. A Nanobody-Based Proximity Ligation Assay Detects Constitutive and Stimulus-Regulated Native Arc/Arg3.1 Oligomers in Hippocampal Neuronal Dendrites. Mol Neurobiol 2025; 62:3973-3990. [PMID: 39367947 PMCID: PMC11880080 DOI: 10.1007/s12035-024-04508-7] [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: 05/22/2024] [Accepted: 09/16/2024] [Indexed: 10/07/2024]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc), the product of an immediate early gene, plays critical roles in synaptic plasticity and memory. Evidence suggests that Arc function is determined by its oligomeric state; however, methods for localization of native Arc oligomers are lacking. Here, we developed a nanobody-based proximity ligation assay (PLA) for detection, localization, and quantification of Arc-Arc complexes in primary rat hippocampal neuronal cultures. We used nanobodies with single, structurally defined epitopes in the bilobar Arc capsid domain. Nanobody H11 binds inside the N-lobe ligand pocket, while nanobody C11 binds to the C-lobe surface. For each nanobody, ALFA- and FLAG-epitope tags created a platform for antibody binding and PLA. Surprisingly, PLA puncta in neuronal dendrites revealed widespread constitutive Arc-Arc complexes. Treatment of cultures with tetrodotoxin or cycloheximide had no effect, suggesting stable complexes that are independent of recent neuronal activity and protein synthesis. To assess detection of oligomers, cultures were exposed to a cell-penetrating peptide inhibitor of the Arc oligomerization motif (OligoOFF). Arc-Arc complexes detected by H11 PLA were inhibited by OligoOff but not by control peptide. Notably, Arc complexes detected by C11 were unaffected by OligoOFF. Furthermore, we evaluated Arc complex formation after chemical stimuli that increase Arc synthesis. Brain-derived neurotrophic factor increased Arc-Arc signal detected by C11, but not H11. Conversely, dihydroxyphenylglycine (DHPG) treatment selectively enhanced H11 PLA signals. In sum, nanobody-based PLA reveals constitutive and stimulus-regulated Arc oligomers in hippocampal neuronal dendrites. A model is proposed based on detection of Arc dimer by C11 and higher-order oligomer by H11 nanobody.
Collapse
Affiliation(s)
- Rodolfo Baldinotti
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Francois P Pauzin
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Hauk Fevang
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Yuta Ishizuka
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway.
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway.
| |
Collapse
|
3
|
Al Othman A, Polyanskaya A, Durymanov M. Human Virus-Like Proteins: Implications for Gene Therapy. Curr Gene Ther 2025; 25:227-236. [PMID: 38798208 DOI: 10.2174/0115665232303436240515071754] [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: 01/09/2024] [Revised: 03/28/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024]
Abstract
An analysis of mammalian genomes has revealed a significant number of DNA sequences with transposon or viral origin. Some of these elements encode functional proteins, repurposed during evolution to play significant physiological roles in certain tissues. Some human virus-like proteins, such as Peg10 and Arc/Arg3.1, structurally demonstrate significant similarity with Gag retroviral proteins, while others, like syncytins-1 and -2, resemble envelope viral proteins. In recent years, it has become clear that these proteins can be exploited for bioengineering 'humanized' capsid particles aimed at targeted mRNA delivery. Realizing this idea could provide efficient virus-like particles for gene therapy and address the problem of viral vector immunogenicity. This review provides an overview of the most-studied human proteins of viral or transposon origin and highlights their biological functions. Additionally, recent advances in exploiting these proteins for targeted mRNA delivery and prospects for their clinical application are discussed.
Collapse
Affiliation(s)
- Aya Al Othman
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky per. 9, Dolgoprudny, Moscow Region, 141701, Russia
| | - Anna Polyanskaya
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky per. 9, Dolgoprudny, Moscow Region, 141701, Russia
| | - Mikhail Durymanov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky per. 9, Dolgoprudny, Moscow Region, 141701, Russia
- Department of Radiochemistry, Faculty of Chemistry, M.V. Lomonosov Moscow State University, 1-3 Leninskie Gory, Moscow, 119991, Russia
| |
Collapse
|
4
|
Upadhayay V, Gu W, Yu Q. Enhancing mRNA Interactions by Engineering the Arc Protein with Nucleocapsid Domain. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:23473-23482. [PMID: 39433292 DOI: 10.1021/acs.langmuir.4c03151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) forms virus-like capsids for mRNA transport between neurons. Unlike HIV-1 Group-specific Antigen (Gag), which uses its Nucleocapsid (NC) domain to bind HIV-1 genomic mRNA, mammalian Arc lacks the NC domain, and their direct mRNA binding interactions remain underexplored. This study examined rat Arc's binding to rat Arc 5' UTR (A5U), HIV-1 5' UTR (H5U), and GFP mRNAs, revealing weak binding with no significant preference. Adding the HIV-1 NC domain to rArc's C-terminus significantly improved binding to H5U, while also showing substantial binding to A5U at about 60% of its H5U level and exhibiting twice the affinity for A5U over GFP mRNA. Importantly, rArc-NC binds 3.4 times more A5U and H5U than GST-NC, indicating that rArc NTD-CA aids mRNA binding by HIV-1 NC. These findings suggest a conserved Gag protein-mRNA interaction mechanism, highlighting the potential for developing mRNA delivery systems that combine endogenous Gag NTD-CA with retroviral NC and UTRs.
Collapse
Affiliation(s)
- Vaibhav Upadhayay
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Wenchao Gu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Qiuming Yu
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
5
|
Mehta K, Yentsch H, Lee J, Yook Y, Lee KY, Gao TT, Tsai NP, Zhang K. Phosphatidylinositol-3-phosphate mediates Arc capsid secretion through the multivesicular body pathway. Proc Natl Acad Sci U S A 2024; 121:e2322422121. [PMID: 39178227 PMCID: PMC11363301 DOI: 10.1073/pnas.2322422121] [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: 01/03/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024] Open
Abstract
Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) is an immediate early gene that plays a vital role in learning and memory. Arc protein has structural and functional properties similar to viral Group-specific antigen (Gag) protein and mediates the intercellular RNA transfer through virus-like capsids. However, the regulators and secretion pathway through which Arc capsids maneuver cargos are unclear. Here, we identified that phosphatidylinositol-3-phosphate (PI3P) mediates Arc capsid assembly and secretion through the endosomal-multivesicular body (MVB) pathway. Indeed, reconstituted Arc protein preferably binds to PI3P. In HEK293T cells, Arc forms puncta that colocalize with FYVE, an endosomal PI3P marker, as well as Rab5 and CD63, early endosomal and MVB markers, respectively. Superresolution imaging resolves Arc accumulates within the intraluminal vesicles of MVB. CRISPR double knockout of RalA and RalB, crucial GTPases for MVB biogenesis and exocytosis, severely reduces the Arc-mediated RNA transfer efficiency. RalA/B double knockdown in cultured rat cortical neurons increases the percentage of mature dendritic spines. Intake of extracellular vesicles purified from Arc-expressing wild-type, but not RalA/B double knockdown, cells in mouse cortical neurons reduces their surface GlutA1 levels. These results suggest that unlike the HIV Gag, whose membrane targeting requires interaction with plasma-membrane-specific phosphatidyl inositol (4,5) bisphosphate (PI(4,5)P2), the assembly of Arc capsids is mediated by PI3P at endocytic membranes. Understanding Arc's secretion pathway helps gain insights into its role in intercellular cargo transfer and highlights the commonality and distinction of trafficking mechanisms between structurally resembled capsid proteins.
Collapse
Affiliation(s)
- Kritika Mehta
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
- NSF Science and Technology Center for Quantitative Cell Biology (STC-QCB) Center, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Henry Yentsch
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Jungbin Lee
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Yeeun Yook
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Tianyu Terry Gao
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Kai Zhang
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL61801
- NSF Science and Technology Center for Quantitative Cell Biology (STC-QCB) Center, University of Illinois Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, IL61801
- Neuroscience Program, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL61801
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL61801
| |
Collapse
|
6
|
Li Y, Zhao X, Tang J, Yi M, Zai X, Zhang J, Cheng G, Yang Y, Xu J. Endogenous capsid-forming protein ARC for self-assembling nanoparticle vaccines. J Nanobiotechnology 2024; 22:513. [PMID: 39192264 PMCID: PMC11348728 DOI: 10.1186/s12951-024-02767-z] [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: 05/13/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
The application of nanoscale scaffolds has become a promising strategy in vaccine design, with protein-based nanoparticles offering desirable avenues for the biocompatible and efficient delivery of antigens. Here, we presented a novel endogenous capsid-forming protein, activated-regulated cytoskeleton-associated protein (ARC), which could be engineered through the plug-and-play strategy (SpyCatcher3/SpyTag3) for multivalent display of antigens. Combined with the self-assembly capacity and flexible modularity of ARC, ARC-based vaccines elicited robust immune responses against Mpox or SARS-CoV-2, comparable to those induced by ferritin-based vaccines. Additionally, ARC-based nanoparticles functioned as immunostimulants, efficiently stimulating dendritic cells and facilitating germinal center responses. Even without adjuvants, ARC-based vaccines generated protective immune responses in a lethal challenge model. Hence, this study showed the feasibility of ARC as a novel protein-based nanocarrier for multivalent surface display of pathogenic antigens and demonstrated the potential of exploiting recombinant mammalian retrovirus-like protein as a delivery vehicle for bioactive molecules.
Collapse
Affiliation(s)
- Yu Li
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaofan Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jiaqi Tang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Mengran Yi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaodong Zai
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Junjie Xu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
7
|
Barylko B, Taylor CA, Wang J, Hedde PN, Chen Y, Hur KH, Binns DD, Brautigam CA, DeMartino GN, Mueller JD, Jameson DM, Albanesi JP. Analysis of Arc/Arg3.1 Oligomerization In Vitro and in Living Cells. Int J Mol Sci 2024; 25:6454. [PMID: 38928159 PMCID: PMC11203824 DOI: 10.3390/ijms25126454] [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: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Arc (also known as Arg3.1) is an activity-dependent immediate early gene product enriched in neuronal dendrites. Arc plays essential roles in long-term potentiation, long-term depression, and synaptic scaling. Although its mechanisms of action in these forms of synaptic plasticity are not completely well established, the activities of Arc include the remodeling of the actin cytoskeleton, the facilitation of AMPA receptor (AMPAR) endocytosis, and the regulation of the transcription of AMPAR subunits. In addition, Arc has sequence and structural similarity to retroviral Gag proteins and self-associates into virus-like particles that encapsulate mRNA and perhaps other cargo for intercellular transport. Each of these activities is likely to be influenced by Arc's reversible self-association into multiple oligomeric species. Here, we used mass photometry to show that Arc exists predominantly as monomers, dimers, and trimers at approximately 20 nM concentration in vitro. Fluorescence fluctuation spectroscopy revealed that Arc is almost exclusively present as low-order (monomer to tetramer) oligomers in the cytoplasm of living cells, over a 200 nM to 5 μM concentration range. We also confirmed that an α-helical segment in the N-terminal domain contains essential determinants of Arc's self-association.
Collapse
Affiliation(s)
- Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.)
| | - Clinton A. Taylor
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.)
| | - Jason Wang
- Department of Physiology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (J.W.); (G.N.D.)
| | - Per Niklas Hedde
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St., BSB 222, Honolulu, HI 96813, USA;
- Laboratory for Fluorescence Dynamics, University of California, Irvine, CA 92697, USA
| | - Yan Chen
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; (Y.C.); (K.-H.H.); (J.D.M.)
| | - Kwang-Ho Hur
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; (Y.C.); (K.-H.H.); (J.D.M.)
| | - Derk D. Binns
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.)
| | - Chad A. Brautigam
- Department of Biophysics, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA;
| | - George N. DeMartino
- Department of Physiology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (J.W.); (G.N.D.)
| | - Joachim D. Mueller
- School of Physics and Astronomy, University of Minnesota, Minneapolis, MN 55455, USA; (Y.C.); (K.-H.H.); (J.D.M.)
| | - David M. Jameson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St., BSB 222, Honolulu, HI 96813, USA;
| | - Joseph P. Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.)
| |
Collapse
|
8
|
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
|
9
|
Barylko B, Taylor CA, Wang J, Earnest S, Stippec S, Binns DD, Brautigam CA, Jameson DM, DeMartino GN, Cobb MH, Albanesi JP. Mimicking Protein Kinase C Phosphorylation Inhibits Arc/Arg3.1 Palmitoylation and Its Interaction with Nucleic Acids. Int J Mol Sci 2024; 25:780. [PMID: 38255853 PMCID: PMC10815921 DOI: 10.3390/ijms25020780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) plays essential roles in diverse forms of synaptic plasticity, including long-term potentiation (LTP), long-term depression (LTD), and homeostatic plasticity. In addition, it assembles into virus-like particles that may deliver mRNAs and/or other cargo between neurons and neighboring cells. Considering this broad range of activities, it is not surprising that Arc is subject to regulation by multiple types of post-translational modification, including phosphorylation, palmitoylation, SUMOylation, ubiquitylation, and acetylation. Here we explore the potential regulatory role of Arc phosphorylation by protein kinase C (PKC), which occurs on serines 84 and 90 within an α-helical segment in the N-terminal domain. To mimic the effect of PKC phosphorylation, we mutated the two serines to negatively charged glutamic acid. A consequence of introducing these phosphomimetic mutations is the almost complete inhibition of Arc palmitoylation, which occurs on nearby cysteines and contributes to synaptic weakening. The mutations also inhibit the binding of nucleic acids and destabilize high-order Arc oligomers. Thus, PKC phosphorylation of Arc may limit the full expression of LTD and may suppress the interneuronal transport of mRNAs.
Collapse
Affiliation(s)
- Barbara Barylko
- Department of Pharmacology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.); (M.H.C.)
| | - Clinton A. Taylor
- Department of Pharmacology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.); (M.H.C.)
| | - Jason Wang
- Department of Physiology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (J.W.); (G.N.D.)
| | - Svetlana Earnest
- Department of Pharmacology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.); (M.H.C.)
| | - Steve Stippec
- Department of Pharmacology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.); (M.H.C.)
| | - Derk D. Binns
- Department of Pharmacology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.); (M.H.C.)
| | - Chad A. Brautigam
- Department of Biophysics, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA;
| | - David M. Jameson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96844, USA;
| | - George N. DeMartino
- Department of Physiology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (J.W.); (G.N.D.)
| | - Melanie H. Cobb
- Department of Pharmacology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.); (M.H.C.)
| | - Joseph P. Albanesi
- Department of Pharmacology, U.T. Southwestern Medical Center, 6001 Forest Park, Dallas, TX 75390, USA; (B.B.); (C.A.T.4th); (D.D.B.); (M.H.C.)
| |
Collapse
|
10
|
Mehta K, Yentsch H, Lee J, Gao TT, Zhang K. Phosphatidylinositol 3-phosphate mediates Arc capsids secretion through the multivesicular body pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572392. [PMID: 38187623 PMCID: PMC10769229 DOI: 10.1101/2023.12.19.572392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc/Arg3.1) is an immediate early gene that plays a vital role in learning and memory. The recent discovery that Arc mediates the inter-neuronal RNA transfer implies its role in regulating neuronal functions across long distances. Arc protein has structural and functional properties similar to viral Group-specific antigen (Gag). By assembling into high-order, virus-like capsids, Arc mediates the intercellular RNA transfer. However, the exact secretion pathway through which Arc capsids maneuver cargos is unclear. Here, we identified that Arc capsids assemble and secrete through the endosomal-multivesicular body (MVB) pathway. Arc's endosomal entry is likely mediated by phosphatidylinositol-3-phosphate (PI3P). Indeed, reconstituted Arc protein preferably binds to PI3P. In mammalian cells, Arc forms puncta that colocalizes with FYVE, an endosomal PI3P marker, and competitive binding to PI3P via prolonged FYVE expression reduces the average number of Arc puncta per cell. Overexpression of MTMR1, a PI3P phosphatase, significantly reduces Arc capsid secretion. Arc capsids secrete through the endosomal-MVB axis as extracellular vesicles. Live-cell imaging shows that fluorescently labeled Arc primarily colocalizes Rab5 and CD63, early endosomal and MVB markers, respectively. Superresolution imaging resolves Arc accumulates within the intraluminal vesicles of MVB. CRISPR double knockout of RalA and RalB, crucial GTPases for MVB biogenesis and exocytosis, severely reduces Arc-mediated RNA transfer efficiency. These results suggest that, unlike the Human Immunodeficiency Virus Gag, which assembles on and bud off from the plasma membrane, Arc capsids assemble at the endocytic membranes of the endosomal-MVB pathway mediated by PI3P. Understanding Arc's secretion pathway helps gain insights into its role in intercellular cargo transfer and highlights the commonality and distinction of trafficking mechanisms between structurally resembled capsid proteins.
Collapse
|
11
|
Xiao C, M’Angale PG, Wang S, Lemieux A, Thomson T. Identifying new players in structural synaptic plasticity through dArc1 interrogation. iScience 2023; 26:108048. [PMID: 37876812 PMCID: PMC10590816 DOI: 10.1016/j.isci.2023.108048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/28/2023] [Accepted: 09/22/2023] [Indexed: 10/26/2023] Open
Abstract
The formation, expansion, and pruning of synapses, known as structural synaptic plasticity, is needed for learning and memory, and perturbation of plasticity is associated with many neurological disorders and diseases. Previously, we observed that the Drosophila homolog of Activity-regulated cytoskeleton-associated protein (dArc1), forms a capsid-like structure, associates with its own mRNA, and is transported across synapses. We demonstrated that this transfer is needed for structural synaptic plasticity. To identify mRNAs that are modified by dArc1 in presynaptic neuron and postsynaptic muscle, we disrupted the expression of dArc1 and performed genomic analysis with deep sequencing. We found that dArc1 affects the expression of genes involved in metabolism, phagocytosis, and RNA-splicing. Through immunoprecipitation we also identified potential mRNA cargos of dArc1 capsids. This study suggests that dArc1 acts as a master regulator of plasticity by affecting several distinct and highly conserved cellular processes.
Collapse
Affiliation(s)
- Cong Xiao
- Department of Neurobiology, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - P. Githure M’Angale
- Department of Neurobiology, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Shuhao Wang
- Department of Neurobiology, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Adrienne Lemieux
- Department of Neurobiology, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | - Travis Thomson
- Department of Neurobiology, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| |
Collapse
|
12
|
Mergiya TF, Gundersen JET, Kanhema T, Brighter G, Ishizuka Y, Bramham CR. Detection of Arc/Arg3.1 oligomers in rat brain: constitutive and synaptic activity-evoked dimer expression in vivo. Front Mol Neurosci 2023; 16:1142361. [PMID: 37363319 PMCID: PMC10289200 DOI: 10.3389/fnmol.2023.1142361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/12/2023] [Indexed: 06/28/2023] Open
Abstract
The immediate early gene product activity-regulated cytoskeleton-associated protein (Arc or Arg3.1) is a major regulator of long-term synaptic plasticity with critical roles in postnatal cortical development and memory formation. However, the molecular basis of Arc function is undefined. Arc is a hub protein with interaction partners in the postsynaptic neuronal compartment and nucleus. Previous in vitro biochemical and biophysical analysis of purified recombinant Arc showed formation of low-order oligomers and larger particles including retrovirus-like capsids. Here, we provide evidence for naturally occurring Arc oligomers in the mammalian brain. Using in situ protein crosslinking to trap weak Arc-Arc interactions, we identified in various preparations a prominent Arc immunoreactive band on SDS-PAGE of molecular mass corresponding to a dimer. While putative trimers, tetramers and heavier Arc species were detected, they were of lower abundance. Stimulus-evoked induction of Arc expression and dimer formation was first demonstrated in SH-SY5Y neuroblastoma cells treated with the muscarinic cholinergic agonist, carbachol, and in primary cortical neuronal cultures treated with brain-derived neurotrophic factor (BDNF). In the dentate gyrus (DG) of adult anesthetized rats, induction of long-term potentiation (LTP) by high-frequency stimulation (HFS) of medial perforant synapses or by brief intrahippocampal infusion of BDNF led to a massive increase in Arc dimer expression. Arc immunoprecipitation of crosslinked DG tissue showed enhanced dimer expression during 4 h of LTP maintenance. Mass spectrometric proteomic analysis of immunoprecipitated, gel-excised bands corroborated detection of Arc dimer. Furthermore, Arc dimer was constitutively expressed in naïve cortical, hippocampal and DG tissue, with the lowest levels in the DG. Taken together the results implicate Arc dimer as the predominant low-oligomeric form in mammalian brain, exhibiting regional differences in its constitutive expression and enhanced synaptic activity-evoked expression in LTP.
Collapse
Affiliation(s)
- Tadiwos F. Mergiya
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Jens Edvard Trygstad Gundersen
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Tambudzai Kanhema
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Grant Brighter
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Yuta Ishizuka
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Clive R. Bramham
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| |
Collapse
|
13
|
Cruz P, Paredes N, Asela I, Kolimi N, Molina JA, Ramírez-Sarmiento CA, Goutam R, Huang G, Medina E, Sanabria H. Domain tethering impacts dimerization and DNA-mediated allostery in the human transcription factor FoxP1. J Chem Phys 2023; 158:2890482. [PMID: 37184020 DOI: 10.1063/5.0138782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
Transcription factors are multidomain proteins with specific DNA binding and regulatory domains. In the human FoxP subfamily (FoxP1, FoxP2, FoxP3, and FoxP4) of transcription factors, a 90 residue-long disordered region links a Leucine Zipper (ZIP)-known to form coiled-coil dimers-and a Forkhead (FKH) domain-known to form domain swapping dimers. We used replica exchange discrete molecular dynamics simulations, single-molecule fluorescence experiments, and other biophysical tools to understand how domain tethering in FoxP1 impacts dimerization at ZIP and FKH domains and how DNA binding allosterically regulates their dimerization. We found that domain tethering promotes FoxP1 dimerization but inhibits a FKH domain-swapped structure. Furthermore, our findings indicate that the linker mediates the mutual organization and dynamics of ZIP and FKH domains, forming closed and open states with and without interdomain contacts, thus highlighting the role of the linkers in multidomain proteins. Finally, we found that DNA allosterically promotes structural changes that decrease the dimerization propensity of FoxP1. We postulate that, upon DNA binding, the interdomain linker plays a crucial role in the gene regulatory function of FoxP1.
Collapse
Affiliation(s)
- Perla Cruz
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Nicolás Paredes
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Isabel Asela
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Narendar Kolimi
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| | - José Alejandro Molina
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago 7820436, Chile
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Santiago 7820436, Chile
| | - César A Ramírez-Sarmiento
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Av. Vicuña Mackenna 4860, Santiago 7820436, Chile
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Santiago 7820436, Chile
| | - Rajen Goutam
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| | - Gangton Huang
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| | - Exequiel Medina
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Casilla 653, Santiago 7800003, Chile
| | - Hugo Sanabria
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina 29634, USA
| |
Collapse
|
14
|
Reinhardt L, Musacchio F, Bichmann M, Behrendt A, Ercan-Herbst E, Stein J, Becher I, Haberkant P, Mader J, Schöndorf DC, Schmitt M, Korffmann J, Reinhardt P, Pohl C, Savitski M, Klein C, Gasparini L, Fuhrmann M, Ehrnhoefer DE. Dual truncation of tau by caspase-2 accelerates its CHIP-mediated degradation. Neurobiol Dis 2023; 182:106126. [PMID: 37086756 DOI: 10.1016/j.nbd.2023.106126] [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: 12/09/2022] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 04/24/2023] Open
Abstract
Intraneuronal aggregates of the microtubule binding protein Tau are a hallmark of different neurodegenerative diseases including Alzheimer's disease (AD). In these aggregates, Tau is modified by posttranslational modifications such as phosphorylation as well as by proteolytic cleavage. Here we identify a novel Tau cleavage site at aspartate 65 (D65) that is specific for caspase-2. In addition, we show that the previously described cleavage site at D421 is also efficiently processed by caspase-2, and both sites are cleaved in human brain samples. Caspase-2-generated Tau fragments show increased aggregation potential in vitro, but do not accumulate in vivo after AAV-mediated overexpression in mouse hippocampus. Interestingly, we observe that steady-state protein levels of caspase-2 generated Tau fragments are low in our in vivo model despite strong RNA expression, suggesting efficient clearance. Consistent with this hypothesis, we find that caspase-2 cleavage significantly improves the recognition of Tau by the ubiquitin E3 ligase CHIP, leading to increased ubiquitination and faster degradation of Tau fragments. Taken together our data thus suggest that CHIP-induced ubiquitination is of particular importance for the clearance of caspase-2 generated Tau fragments in vitro and in vivo.
Collapse
Affiliation(s)
- Lydia Reinhardt
- BioMed X Institute, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany; AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Fabrizio Musacchio
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Maria Bichmann
- BioMed X Institute, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Annika Behrendt
- BioMed X Institute, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Ebru Ercan-Herbst
- BioMed X Institute, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany
| | - Juliane Stein
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Isabelle Becher
- European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Per Haberkant
- European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Julia Mader
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - David C Schöndorf
- BioMed X Institute, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany; AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Melanie Schmitt
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Jürgen Korffmann
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Peter Reinhardt
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Christian Pohl
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Mikhail Savitski
- European Molecular Biology Laboratory (EMBL), Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Corinna Klein
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Laura Gasparini
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany
| | - Martin Fuhrmann
- Neuroimmunology and Imaging Group, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127 Bonn, Germany
| | - Dagmar E Ehrnhoefer
- BioMed X Institute, Im Neuenheimer Feld 515, 69120 Heidelberg, Germany; AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery, Knollstrasse, 67061 Ludwigshafen am Rhein, Germany.
| |
Collapse
|
15
|
Volpato A, Ollech D, Alvelid J, Damenti M, Müller B, York AG, Ingaramo M, Testa I. Extending fluorescence anisotropy to large complexes using reversibly switchable proteins. Nat Biotechnol 2023; 41:552-559. [PMID: 36217028 PMCID: PMC10110461 DOI: 10.1038/s41587-022-01489-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 08/26/2022] [Indexed: 11/08/2022]
Abstract
The formation of macromolecular complexes can be measured by detection of changes in rotational mobility using time-resolved fluorescence anisotropy. However, this method is limited to relatively small molecules (~0.1-30 kDa), excluding the majority of the human proteome and its complexes. We describe selective time-resolved anisotropy with reversibly switchable states (STARSS), which overcomes this limitation and extends the observable mass range by more than three orders of magnitude. STARSS is based on long-lived reversible molecular transitions of switchable fluorescent proteins to resolve the relatively slow rotational diffusivity of large complexes. We used STARSS to probe the rotational mobility of several molecular complexes in cells, including chromatin, the retroviral Gag lattice and activity-regulated cytoskeleton-associated protein oligomers. Because STARSS can probe arbitrarily large structures, it is generally applicable to the entire human proteome.
Collapse
Affiliation(s)
- Andrea Volpato
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Dirk Ollech
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jonatan Alvelid
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martina Damenti
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Barbara Müller
- Department of Infectious Diseases, Virology, Centre for Integrative Infectious Disease Research, University Hospital Heidelberg, Heidelberg, Germany
| | - Andrew G York
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | | | - Ilaria Testa
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
16
|
Fan H, Wang Y, Zou Y, Song W, Xie J, Tang X, Chen S. ARC/Arg3.1 expression in the lateral geniculate body of monocular form deprivation amblyopic kittens. BMC Ophthalmol 2023; 23:3. [PMID: 36597053 PMCID: PMC9809052 DOI: 10.1186/s12886-022-02757-5] [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: 09/21/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
PURPOSE The present study compared the expression of activity-regulated cytoskeleton-associated protein (ARC/Arg3.1) in the lateral geniculate body between form deprivation amblyopia kittens and normal kittens to examine the significance of ARC/Arg3.1 in the lateral geniculate body in the pathogenesis of amblyopia. METHODS Twenty kittens were randomly divided into an experimental group (n = 10) and a control group (n = 10). Black opaque covering cloth was used to cover the right eye of kittens in the experimental group. Pattern visual evoked potentials (PVEP) were detected weekly in all kittens. The expression of the ARC/Arg3.1 gene was detected by immunohistochemistry and in situ hybridization, and apoptosis of lateral geniculate body cells was detected by TUNEL. RESULTS PVEP detection showed that at the age of 5 and 7 weeks, the latency of P100 in the right eye of the experimental group was higher than that of the other three groups (P < 0.05), and the amplitude of P100 was lower than that of the other three groups (P < 0.05). The expression of ARC/Arg3.1 protein (P < 0.05) and mRNA (P < 0.05) in the lateral geniculate body of the experimental group was significantly lower than that of the control group. The level of neuronal apoptosis in the experimental group was higher than that in the control group (P < 0.05). The expression of the ARC/Arg3.1 gene was negatively correlated with the apoptosis level of lateral geniculate body neurons. CONCLUSIONS The expression of ARC/Arg3.1 is associated with monocular form deprivation amblyopia and apoptosis of lateral geniculate body cells.
Collapse
Affiliation(s)
- Haobo Fan
- Department of Optometry, North Sichuan Medical College, No.234 FuJiang Road, Nanchong, 637000, China
- Department of Optometry and Pediatric Ophthalmology, Ineye Hospital of Chengdu University of TCM, Chengdu, China
| | - Ying Wang
- Department of Optometry, North Sichuan Medical College, No.234 FuJiang Road, Nanchong, 637000, China
| | - Yunchun Zou
- Department of Optometry, North Sichuan Medical College, No.234 FuJiang Road, Nanchong, 637000, China.
- Department of Ophthalmology, the Second Clinical College of North Sichuan Medical College (Nanchong Central Hospital), Nanchong, China.
| | - Weiqi Song
- Department of Optometry, North Sichuan Medical College, No.234 FuJiang Road, Nanchong, 637000, China
| | - Juan Xie
- Department of Optometry, North Sichuan Medical College, No.234 FuJiang Road, Nanchong, 637000, China
| | - Xiuping Tang
- Department of Optometry, North Sichuan Medical College, No.234 FuJiang Road, Nanchong, 637000, China
| | - Siyu Chen
- Department of Optometry, North Sichuan Medical College, No.234 FuJiang Road, Nanchong, 637000, China
| |
Collapse
|
17
|
Eriksen MS, Bramham CR. Molecular physiology of Arc/Arg3.1: The oligomeric state hypothesis of synaptic plasticity. Acta Physiol (Oxf) 2022; 236:e13886. [PMID: 36073248 PMCID: PMC9787330 DOI: 10.1111/apha.13886] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 01/29/2023]
Abstract
The immediate early gene, Arc, is a pivotal regulator of synaptic plasticity, memory, and cognitive flexibility. But what is Arc protein? How does it work? Inside the neuron, Arc is a protein interaction hub and dynamic regulator of intra-cellular signaling in synaptic plasticity. In remarkable contrast, Arc can also self-assemble into retrovirus-like capsids that are released in extracellular vesicles and capable of intercellular transfer of RNA. Elucidation of the molecular basis of Arc hub and capsid functions, and the relationship between them, is vital for progress. Here, we discuss recent findings on Arc structure-function and regulation of oligomerization that are giving insight into the molecular physiology of Arc. The unique features of mammalian Arc are emphasized, while drawing comparisons with Drosophila Arc and retroviral Gag. The Arc N-terminal domain, found only in mammals, is proposed to play a key role in regulating Arc hub signaling, oligomerization, and formation of capsids. Bringing together several lines of evidence, we hypothesize that Arc function in synaptic plasticity-long-term potentiation (LTP) and long-term depression (LTD)-are dictated by different oligomeric forms of Arc. Specifically, monomer/dimer function in LTP, tetramer function in basic LTD, and 32-unit oligomer function in enhanced LTD. The role of mammalian Arc capsids is unclear but likely depends on the cross-section of captured neuronal activity-induced RNAs. As the functional states of Arc are revealed, it may be possible to selectively manipulate specific forms of Arc-dependent plasticity and intercellular communication involved in brain function and dysfunction.
Collapse
Affiliation(s)
| | - Clive R. Bramham
- Department of BiomedicineUniversity of BergenBergenNorway,Mohn Research Center for the BrainUniversity of BergenBergenNorway
| |
Collapse
|
18
|
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
|
19
|
Ishizuka Y, Mergiya TF, Baldinotti R, Xu J, Hallin EI, Markússon S, Kursula P, Bramham CR. Development and Validation of Arc Nanobodies: New Tools for Probing Arc Dynamics and Function. Neurochem Res 2022; 47:2656-2666. [PMID: 35307777 PMCID: PMC9463278 DOI: 10.1007/s11064-022-03573-5] [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: 01/21/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 11/06/2022]
Abstract
Activity-regulated cytoskeleton-associated (Arc) protein plays key roles in long-term synaptic plasticity, memory, and cognitive flexibility. However, an integral understanding of Arc mechanisms is lacking. Arc is proposed to function as an interaction hub in neuronal dendrites and the nucleus, yet Arc can also form retrovirus-like capsids with proposed roles in intercellular communication. Here, we sought to develop anti-Arc nanobodies (ArcNbs) as new tools for probing Arc dynamics and function. Six ArcNbs representing different clonal lines were selected from immunized alpaca. Immunoblotting with recombinant ArcNbs fused to a small ALFA-epitope tag demonstrated binding to recombinant Arc as well as endogenous Arc from rat cortical tissue. ALFA-tagged ArcNb also provided efficient immunoprecipitation of stimulus-induced Arc after carbachol-treatment of SH-SY5Y neuroblastoma cells and induction of long-term potentiation in the rat dentate gyrus in vivo. Epitope mapping showed that all Nbs recognize the Arc C-terminal region containing the retroviral Gag capsid homology domain, comprised of tandem N- and C-lobes. ArcNbs E5 and H11 selectively bound the N-lobe, which harbors a peptide ligand binding pocket specific to mammals. Four additional ArcNbs bound the region containing the C-lobe and C-terminal tail. For use as genetically encoded fluorescent intrabodies, we show that ArcNbs fused to mScarlet-I are uniformly expressed, without aggregation, in the cytoplasm and nucleus of HEK293FT cells. Finally, mScarlet-I-ArcNb H11 expressed as intrabody selectively bound the N-lobe and enabled co-immunoprecipitation of full-length intracellular Arc. ArcNbs are versatile tools for live-cell labeling and purification of Arc, and interrogation of Arc capsid domain specific functions.
Collapse
Affiliation(s)
- Yuta Ishizuka
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway.
- Department of Pathophysiology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Tadiwos F Mergiya
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Mohn Center for Research on the Brain, University of Bergen, Bergen, Norway
| | - Rodolfo Baldinotti
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Mohn Center for Research on the Brain, University of Bergen, Bergen, Norway
| | - Ju Xu
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
| | - Erik I Hallin
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
| | - Sigurbjörn Markússon
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
| | - Petri Kursula
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway.
- Mohn Center for Research on the Brain, University of Bergen, Bergen, Norway.
| |
Collapse
|
20
|
Markússon S, Hallin EI, Bustad HJ, Raasakka A, Xu J, Muruganandam G, Loris R, Martinez A, Bramham CR, Kursula P. High-affinity anti-Arc nanobodies provide tools for structural and functional studies. PLoS One 2022; 17:e0269281. [PMID: 35671319 PMCID: PMC9173642 DOI: 10.1371/journal.pone.0269281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/17/2022] [Indexed: 11/19/2022] Open
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is a multidomain protein of retroviral origin with a vital role in the regulation of synaptic plasticity and memory formation in mammals. However, the mechanistic and structural basis of Arc function is poorly understood. Arc has an N-terminal domain (NTD) involved in membrane binding and a C-terminal domain (CTD) that binds postsynaptic protein ligands. In addition, the NTD and CTD both function in Arc oligomerisation, including assembly of retrovirus-like capsids involved in intercellular signalling. To obtain new tools for studies on Arc structure and function, we produced and characterised six high-affinity anti-Arc nanobodies (Nb). The CTD of rat and human Arc were both crystallised in ternary complexes with two Nbs. One Nb bound deep into the stargazin-binding pocket of Arc CTD and suggested competitive binding with Arc ligand peptides. The crystallisation of the human Arc CTD in two different conformations, accompanied by SAXS data and molecular dynamics simulations, paints a dynamic picture of the mammalian Arc CTD. The collapsed conformation closely resembles Drosophila Arc in capsids, suggesting that we have trapped a capsid-like conformation of the human Arc CTD. Our data obtained with the help of anti-Arc Nbs suggest that structural dynamics of the CTD and dimerisation of the NTD may promote the formation of capsids. Taken together, the recombinant high-affinity anti-Arc Nbs are versatile tools that can be further developed for studying mammalian Arc structure and function, as well as mechanisms of Arc capsid formation, both in vitro and in vivo. For example, the Nbs could serve as a genetically encoded tools for inhibition of endogenous Arc interactions in the study of neuronal function and plasticity.
Collapse
Affiliation(s)
| | - Erik I. Hallin
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Arne Raasakka
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ju Xu
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Gopinath Muruganandam
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium
- Department of Bioengineering Sciences, Structural Biology Brussels, Vrije Universiteit Brussel, Brussel, Belgium
| | - Remy Loris
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium
- Department of Bioengineering Sciences, Structural Biology Brussels, Vrije Universiteit Brussel, Brussel, Belgium
| | - Aurora Martinez
- 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
- * E-mail:
| |
Collapse
|
21
|
Hedde PN, Barylko B, Binns DD, Jameson DM, Albanesi JP. Differential Mobility and Self-Association of Arc/Arg3.1 in the Cytoplasm and Nucleus of Living Cells. ACS Chem Neurosci 2022; 13:876-882. [PMID: 35319179 DOI: 10.1021/acschemneuro.1c00744] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Arc, also known as Arg3.1, is an activity-dependent immediate-early gene product that plays essential roles in memory consolidation. A pool of Arc is located in the postsynaptic cytoplasm, where it promotes AMPA receptor endocytosis and cytoskeletal remodeling. However, Arc is also found in the nucleus, with a major portion being associated with promyelocytic leukemia nuclear bodies (PML-NBs). Nuclear Arc has been implicated in epigenetic control of gene transcription associated with learning and memory. In this study, we use a battery of fluorescence nanoimaging approaches to characterize the behavior of Arc ectopically expressed in heterologous cells. Our results indicate that in the cytoplasm, Arc exists predominantly as monomers and dimers associated with slowly diffusing particles. In contrast, nuclear Arc is almost exclusively monomeric and displays a higher diffusivity than cytoplasmic Arc. We further show that Arc moves freely and rapidly between PML-NBs and the nucleoplasm and that its movement within PML-NBs is relatively unobstructed.
Collapse
Affiliation(s)
- Per Niklas Hedde
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, 651 Ilalo Street, BSB 222, University of Hawaii, Honolulu, Hawaii 96813, United States
| | - Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, Texas 75390, United States
| | - Derk D. Binns
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, Texas 75390, United States
| | - David M. Jameson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, 651 Ilalo Street, BSB 222, University of Hawaii, Honolulu, Hawaii 96813, United States
| | - Joseph P. Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, 6001 Forest Park, Dallas, Texas 75390, United States
| |
Collapse
|
22
|
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
|
23
|
Segel M, Lash B, Song J, Ladha A, Liu CC, Jin X, Mekhedov SL, Macrae RK, Koonin EV, Zhang F. Mammalian retrovirus-like protein PEG10 packages its own mRNA and can be pseudotyped for mRNA delivery. Science 2021; 373:882-889. [PMID: 34413232 DOI: 10.1126/science.abg6155] [Citation(s) in RCA: 259] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/26/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022]
Abstract
Eukaryotic genomes contain domesticated genes from integrating viruses and mobile genetic elements. Among these are homologs of the capsid protein (known as Gag) of long terminal repeat (LTR) retrotransposons and retroviruses. We identified several mammalian Gag homologs that form virus-like particles and one LTR retrotransposon homolog, PEG10, that preferentially binds and facilitates vesicular secretion of its own messenger RNA (mRNA). We showed that the mRNA cargo of PEG10 can be reprogrammed by flanking genes of interest with Peg10's untranslated regions. Taking advantage of this reprogrammability, we developed selective endogenous encapsidation for cellular delivery (SEND) by engineering both mouse and human PEG10 to package, secrete, and deliver specific RNAs. Together, these results demonstrate that SEND is a modular platform suited for development as an efficient therapeutic delivery modality.
Collapse
Affiliation(s)
- Michael Segel
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Blake Lash
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jingwei Song
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alim Ladha
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Catherine C Liu
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xin Jin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Society of Fellows, Harvard University, Cambridge, MA 02138 USA
| | - Sergei L Mekhedov
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Rhiannon K Macrae
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Feng Zhang
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
24
|
Hallin EI, Bramham CR, Kursula P. Structural properties and peptide ligand binding of the capsid homology domains of human Arc. Biochem Biophys Rep 2021; 26:100975. [PMID: 33732907 PMCID: PMC7941041 DOI: 10.1016/j.bbrep.2021.100975] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
The activity-regulated cytoskeleton-associated protein (Arc) is important for synaptic plasticity and the normal function of the brain. Arc interacts with neuronal postsynaptic proteins, but the mechanistic details of its function have not been fully established. The C-terminal domain of Arc consists of tandem domains, termed the N- and C-lobe. The N-lobe harbours a peptide binding site, able to bind multiple targets. By measuring the affinity of human Arc towards various peptides from stargazin and guanylate kinase-associated protein (GKAP), we have refined its specificity determinants. We found two sites in the GKAP repeat region that bind to Arc and confirmed these interactions by X-ray crystallography. Phosphorylation of the stargazin peptide did not affect binding affinity but caused changes in thermodynamic parameters. Comparison of the crystal structures of three high-resolution human Arc-peptide complexes identifies three conserved C-H…π interactions at the binding cavity, explaining the sequence specificity of short linear motif binding by Arc. We further characterise central residues of the Arc lobe fold, show the effects of peptide binding on protein dynamics, and identify acyl carrier proteins as structures similar to the Arc lobes. We hypothesise that Arc may affect protein-protein interactions and phase separation at the postsynaptic density, affecting protein turnover and re-modelling of the synapse. The present data on Arc structure and ligand binding will help in further deciphering these processes.
Collapse
Affiliation(s)
| | | | - Petri Kursula
- Department of Biomedicine, University of Bergen, Norway
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland
- Biocenter Oulu, University of Oulu, Finland
| |
Collapse
|
25
|
Walczyk-Mooradally A, Holborn J, Singh K, Tyler M, Patnaik D, Wesseling H, Brandon NJ, Steen J, Graether SP, Haggarty SJ, Lalonde J. Phosphorylation-dependent control of Activity-regulated cytoskeleton-associated protein (Arc) protein by TNIK. J Neurochem 2021. [PMID: 34077555 DOI: 10.1111/jnc.15077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene product that support neuroplastic changes important for cognitive function and memory formation. As a protein with homology to the retroviral Gag protein, a particular characteristic of Arc is its capacity to self-assemble into virus-like capsids that can package mRNAs and transfer those transcripts to other cells. Although a lot has been uncovered about the contributions of Arc to neuron biology and behavior, very little is known about how different functions of Arc are coordinately regulated both temporally and spatially in neurons. The answer to this question we hypothesized must involve the occurrence of different protein post-translational modifications acting to confer specificity. In this study, we used mass spectrometry and sequence prediction strategies to map novel Arc phosphorylation sites. Our approach led us to recognize serine 67 (S67) and threonine 278 (T278) as residues that can be modified by TNIK, which is a kinase abundantly expressed in neurons that shares many functional overlaps with Arc and has, along with its interacting proteins such as the NMDA receptor, and been implicated as a risk factor for psychiatric disorders. Furthermore, characterization of each residue using site-directed mutagenesis to create S67 and T278 mutant variants revealed that TNIK action at those amino acids can strongly influence Arc's subcellular distribution and self-assembly as capsids. Together, our findings reveal an unsuspected connection between Arc and TNIK. Better understanding of the interplay between these two proteins in neuronal cells could lead to new insights about apparition and progression of psychiatric disorders. Cover Image for this issue: https://doi.org/10.1111/jnc.15077.
Collapse
Affiliation(s)
| | - Jennifer Holborn
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Karamjeet Singh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Marshall Tyler
- Massachusetts General Hospital, Centre for Genomic Medicine, Boston, MA, USA
| | - Debasis Patnaik
- Massachusetts General Hospital, Centre for Genomic Medicine, Boston, MA, USA
| | - Hendrik Wesseling
- Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Nicholas J Brandon
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca Boston, Waltham, MA, USA
| | - Judith Steen
- Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Steffen P Graether
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Stephen J Haggarty
- Massachusetts General Hospital, Centre for Genomic Medicine, Boston, MA, USA
| | - Jasmin Lalonde
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
26
|
Walczyk-Mooradally A, Holborn J, Singh K, Tyler M, Patnaik D, Wesseling H, Brandon NJ, Steen J, Graether SP, Haggarty SJ, Lalonde J. Phosphorylation-dependent control of Activity-regulated cytoskeleton-associated protein (Arc) protein by TNIK. J Neurochem 2021; 158:1058-1073. [PMID: 34077555 DOI: 10.1111/jnc.15440] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 12/21/2022]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene product that support neuroplastic changes important for cognitive function and memory formation. As a protein with homology to the retroviral Gag protein, a particular characteristic of Arc is its capacity to self-assemble into virus-like capsids that can package mRNAs and transfer those transcripts to other cells. Although a lot has been uncovered about the contributions of Arc to neuron biology and behavior, very little is known about how different functions of Arc are coordinately regulated both temporally and spatially in neurons. The answer to this question we hypothesized must involve the occurrence of different protein post-translational modifications acting to confer specificity. In this study, we used mass spectrometry and sequence prediction strategies to map novel Arc phosphorylation sites. Our approach led us to recognize serine 67 (S67) and threonine 278 (T278) as residues that can be modified by TNIK, which is a kinase abundantly expressed in neurons that shares many functional overlaps with Arc and has, along with its interacting proteins such as the NMDA receptor, and been implicated as a risk factor for psychiatric disorders. Furthermore, characterization of each residue using site-directed mutagenesis to create S67 and T278 mutant variants revealed that TNIK action at those amino acids can strongly influence Arc's subcellular distribution and self-assembly as capsids. Together, our findings reveal an unsuspected connection between Arc and TNIK. Better understanding of the interplay between these two proteins in neuronal cells could lead to new insights about apparition and progression of psychiatric disorders.
Collapse
Affiliation(s)
| | - Jennifer Holborn
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Karamjeet Singh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Marshall Tyler
- Massachusetts General Hospital, Centre for Genomic Medicine, Boston, MA, USA
| | - Debasis Patnaik
- Massachusetts General Hospital, Centre for Genomic Medicine, Boston, MA, USA
| | - Hendrik Wesseling
- Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Nicholas J Brandon
- Neuroscience, BioPharmaceuticals R&D, AstraZeneca Boston, Waltham, MA, USA
| | - Judith Steen
- Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Steffen P Graether
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Stephen J Haggarty
- Massachusetts General Hospital, Centre for Genomic Medicine, Boston, MA, USA
| | - Jasmin Lalonde
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
27
|
Acuña-Hinrichsen F, Covarrubias-Pinto A, Ishizuka Y, Stolzenbach MF, Martin C, Salazar P, Castro MA, Bramham CR, Otth C. Herpes Simplex Virus Type 1 Neuronal Infection Triggers the Disassembly of Key Structural Components of Dendritic Spines. Front Cell Neurosci 2021; 15:580717. [PMID: 33708072 PMCID: PMC7940845 DOI: 10.3389/fncel.2021.580717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/01/2021] [Indexed: 11/13/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a widespread neurotropic virus. Primary infection of HSV-1 in facial epithelium leads to retrograde axonal transport to the central nervous system (CNS) where it establishes latency. Under stressful conditions, the virus reactivates, and new progeny are transported anterogradely to the primary site of infection. During the late stages of neuronal infection, axonal damage can occur, however, the impact of HSV-1 infection on the morphology and functional integrity of neuronal dendrites during the early stages of infection is unknown. We previously demonstrated that acute HSV-1 infection in neuronal cell lines selectively enhances Arc protein expression - a major regulator of long-term synaptic plasticity and memory consolidation, known for being a protein-interaction hub in the postsynaptic dendritic compartment. Thus, HSV-1 induced Arc expression may alter the functionality of infected neurons and negatively impact dendritic spine dynamics. In this study we demonstrated that HSV-1 infection induces structural disassembly and functional deregulation in cultured cortical neurons, an altered glutamate response, Arc accumulation within the somata, and decreased expression of spine scaffolding-like proteins such as PSD-95, Drebrin and CaMKIIβ. However, whether these alterations are specific to the HSV-1 infection mechanism or reflect a secondary neurodegenerative process remains to be determined.
Collapse
Affiliation(s)
- Francisca Acuña-Hinrichsen
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Post-graduate Program, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Adriana Covarrubias-Pinto
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Post-graduate Program, Science Faculty, Universidad Austral de Chile, Valdivia, Chile
- Institute of Biochemistry II, Goethe University School of Medicine, Frankfurt am Main, Germany
- Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
| | - Yuta Ishizuka
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - Carolina Martin
- School of Medical Technology, Austral University of Chile, Puerto Montt, Chile
| | - Paula Salazar
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Maite A. Castro
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
- Janelia Research Campus, HHMI, VA, United States
| | | | - Carola Otth
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
28
|
Yakout DW, Shree N, Mabb AM. Effect of pharmacological manipulations on Arc function. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2020; 2:100013. [PMID: 34909648 PMCID: PMC8663979 DOI: 10.1016/j.crphar.2020.100013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022] Open
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is a brain-enriched immediate early gene that regulates important mechanisms implicated in learning and memory. Arc levels are controlled through a balance of induction and degradation in an activity-dependent manner. Arc further undergoes multiple post-translational modifications that regulate its stability, localization and function. Recent studies demonstrate that these features of Arc can be pharmacologically manipulated. In this review, we discuss some of these compounds, with an emphasis on drugs of abuse and psychotropic drugs. We also discuss inflammatory states that regulate Arc.
Collapse
Affiliation(s)
- Dina W. Yakout
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Nitheyaa Shree
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
29
|
Eriksen MS, Nikolaienko O, Hallin EI, Grødem S, Bustad HJ, Flydal MI, Merski I, Hosokawa T, Lascu D, Akerkar S, Cuéllar J, Chambers JJ, O'Connell R, Muruganandam G, Loris R, Touma C, Kanhema T, Hayashi Y, Stratton MM, Valpuesta JM, Kursula P, Martinez A, Bramham CR. Arc self-association and formation of virus-like capsids are mediated by an N-terminal helical coil motif. FEBS J 2020; 288:2930-2955. [PMID: 33175445 DOI: 10.1111/febs.15618] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/13/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
Activity-regulated cytoskeleton-associated protein (Arc) is a protein interaction hub with diverse roles in intracellular neuronal signaling, and important functions in neuronal synaptic plasticity, memory, and postnatal cortical development. Arc has homology to retroviral Gag protein and is capable of self-assembly into virus-like capsids implicated in the intercellular transfer of RNA. However, the molecular basis of Arc self-association and capsid formation is largely unknown. Here, we identified a 28-amino-acid stretch in the mammalian Arc N-terminal (NT) domain that is necessary and sufficient for self-association. Within this region, we identified a 7-residue oligomerization motif, critical for the formation of virus-like capsids. Purified wild-type Arc formed capsids as shown by transmission and cryo-electron microscopy, whereas mutant Arc with disruption of the oligomerization motif formed homogenous dimers. An atomic-resolution crystal structure of the oligomerization region peptide demonstrated an antiparallel coiled-coil interface, strongly supporting NT-NT domain interactions in Arc oligomerization. The NT coil-coil interaction was also validated in live neurons using fluorescence lifetime FRET imaging, and mutation of the oligomerization motif disrupted Arc-facilitated endocytosis. Furthermore, using single-molecule photobleaching, we show that Arc mRNA greatly enhances higher-order oligomerization in a manner dependent on the oligomerization motif. In conclusion, a helical coil in the Arc NT domain supports self-association above the dimer stage, mRNA-induced oligomerization, and formation of virus-like capsids. DATABASE: The coordinates and structure factors for crystallographic analysis of the oligomerization region were deposited at the Protein Data Bank with the entry code 6YTU.
Collapse
Affiliation(s)
- Maria S Eriksen
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Oleksii Nikolaienko
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Erik I Hallin
- Department of Biomedicine, University of Bergen, Norway
| | - Sverre Grødem
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Helene J Bustad
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Marte I Flydal
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Ian Merski
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, MA, USA
| | - Tomohisa Hosokawa
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daniela Lascu
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Shreeram Akerkar
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Jorge Cuéllar
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - James J Chambers
- Institute for Applied Life Sciences, University of Massachusetts Amherst, MA, USA
| | - Rory O'Connell
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, MA, USA
| | - Gopinath Muruganandam
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium.,Structural Biology Brussels, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Belgium
| | - Remy Loris
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium.,Structural Biology Brussels, Department of Bioengineering Sciences, Vrije Universiteit Brussel, Belgium
| | - Christine Touma
- Faculty of Biochemistry and Molecular Biology & Biocenter Oulu, University of Oulu, Finland
| | - Tambudzai Kanhema
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Margaret M Stratton
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, MA, USA
| | | | - Petri Kursula
- Department of Biomedicine, University of Bergen, Norway.,Faculty of Biochemistry and Molecular Biology & Biocenter Oulu, University of Oulu, Finland
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Norway.,KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, Norway
| |
Collapse
|
30
|
Boldridge M, Shimabukuro J, Nakamatsu K, Won C, Jansen C, Turner H, Wang L. Characterization of the C-terminal tail of the Arc protein. PLoS One 2020; 15:e0239870. [PMID: 32991626 PMCID: PMC7523963 DOI: 10.1371/journal.pone.0239870] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022] Open
Abstract
The activity-regulated cytoskeleton-associate protein Arc (or Arg3.1) is specifically linked to memory formation and a number of cognitive disorders, including Alzheimer's disease and schizophrenia. Since the discovery of Arc in 1995, extensive research has been conducted on the protein to identify its function and mechanisms of action, with solving the structure of Arc as a major goal. However, the Arc protein tends to self-oligomerize in vitro, and is difficult to crystallize. These properties have hindered efforts to obtain the structure of the full-length, whole protein Arc. As an alternative approach, we and others, have sought to solve the structures of various subdomain proteins of Arc, including the N-lobe, C-lobe, and capsid domain (N-lobe + C-lobe). In this study, we characterized the C-terminal tail of Arc using integrated bioinformatic and structural biology techniques. We compared the sequences of Arc proteins in different mammal species and found that the amino-acid composition in the C-terminal tail region has a significantly higher degree of variation rate than the rest of the protein. Structural prediction programs suggested that the C-terminal tail is structurally disordered. Chemical shift analysis based on solution NMR spectra confirmed that the C-terminal tail has a random coil (disordered) structure, and the tail starts from the residue D357. Furthermore, the NMR spectra showed that the C-terminal tail has minimum (if any) interaction with its neighboring capsid domain in Arc. This study fills gaps in our specific understanding of the structural nature and functional contributions of the Arc C-terminus.
Collapse
Affiliation(s)
- Melissa Boldridge
- Department of Natural Sciences, Hawaii Pacific University, Honolulu, Hawaii, United States of America
| | - Jody Shimabukuro
- Department of Natural Sciences, Hawaii Pacific University, Honolulu, Hawaii, United States of America
| | - Keith Nakamatsu
- Department of Natural Sciences, Windward Community College, Kaneohe, Hawaii, United States of America
| | - Christian Won
- Department of Natural Sciences, Windward Community College, Kaneohe, Hawaii, United States of America
| | - Chad Jansen
- Laboratory of Immunology and Signal Transduction, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, Hawaii, United States of America
| | - Helen Turner
- Laboratory of Immunology and Signal Transduction, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, Hawaii, United States of America
| | - Lei Wang
- Department of Natural Sciences, Hawaii Pacific University, Honolulu, Hawaii, United States of America
| |
Collapse
|
31
|
Zhang H, Bramham CR. Arc/Arg3.1 function in long-term synaptic plasticity: Emerging mechanisms and unresolved issues. Eur J Neurosci 2020; 54:6696-6712. [PMID: 32888346 DOI: 10.1111/ejn.14958] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/18/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Arc (activity-regulated cytoskeleton-associated protein) is posited as a critical regulator of long-term synaptic plasticity at excitatory synapses, including long-term potentiation, long-term depression, inverse synaptic tagging and homoeostatic scaling, with pivotal roles in memory and postnatal cortical development. However, the mechanisms underlying the bidirectional regulation of synaptic strength are poorly understood. Here we review evidence from different plasticity paradigms, highlight outstanding issues and discuss stimulus-specific mechanisms that dictate Arc function. We propose a model in which Arc bidirectionally controls synaptic strength by coordinate regulation of AMPA-type glutamate receptor (AMPAR) trafficking and actin cytoskeletal dynamics in dendritic spines. Key to this model, Arc is proposed to function as an activity-dependent regulator of AMPAR lateral membrane diffusion and trapping at synapses.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
32
|
Albanesi JP, Barylko B, DeMartino GN, Jameson DM. Palmitoylated Proteins in Dendritic Spine Remodeling. Front Synaptic Neurosci 2020; 12:22. [PMID: 32655390 PMCID: PMC7325885 DOI: 10.3389/fnsyn.2020.00022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022] Open
Abstract
Activity-responsive changes in the actin cytoskeleton are required for the biogenesis, motility, and remodeling of dendritic spines. These changes are governed by proteins that regulate the polymerization, depolymerization, bundling, and branching of actin filaments. Thus, processes that have been extensively characterized in the context of non-neuronal cell shape change and migration are also critical for learning and memory. In this review article, we highlight actin regulatory proteins that associate, at least transiently, with the dendritic plasma membrane. All of these proteins have been shown, either in directed studies or in high-throughput screens, to undergo palmitoylation, a potentially reversible, and stimulus-dependent cysteine modification. Palmitoylation increases the affinity of peripheral proteins for the membrane bilayer and contributes to their subcellular localization and recruitment to cholesterol-rich membrane microdomains.
Collapse
Affiliation(s)
- Joseph P. Albanesi
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Barbara Barylko
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - George N. DeMartino
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David M. Jameson
- Department of Cell and Molecular Biology, University of Hawaii, Honolulu, HI, United States
| |
Collapse
|
33
|
Affiliation(s)
- Vivian Budnik
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Travis Thomson
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
34
|
Kiss-Tóth A, Dobson L, Péterfia B, Ángyán AF, Ligeti B, Lukács G, Gáspári Z. Occurrence of Ordered and Disordered Structural Elements in Postsynaptic Proteins Supports Optimization for Interaction Diversity. ENTROPY (BASEL, SWITZERLAND) 2019; 21:E761. [PMID: 33267475 PMCID: PMC7515291 DOI: 10.3390/e21080761] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/30/2019] [Accepted: 08/02/2019] [Indexed: 12/15/2022]
Abstract
The human postsynaptic density is an elaborate network comprising thousands of proteins, playing a vital role in the molecular events of learning and the formation of memory. Despite our growing knowledge of specific proteins and their interactions, atomic-level details of their full three-dimensional structure and their rearrangements are mostly elusive. Advancements in structural bioinformatics enabled us to depict the characteristic features of proteins involved in different processes aiding neurotransmission. We show that postsynaptic protein-protein interactions are mediated through the delicate balance of intrinsically disordered regions and folded domains, and this duality is also imprinted in the amino acid sequence. We introduce Diversity of Potential Interactions (DPI), a structure and regulation based descriptor to assess the diversity of interactions. Our approach reveals that the postsynaptic proteome has its own characteristic features and these properties reliably discriminate them from other proteins of the human proteome. Our results suggest that postsynaptic proteins are especially susceptible to forming diverse interactions with each other, which might be key in the reorganization of the postsynaptic density (PSD) in molecular processes related to learning and memory.
Collapse
Affiliation(s)
- Annamária Kiss-Tóth
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50A, 1083 Budapest, Hungary
- 3in-PPCU Research Group, 2500 Esztergom, Hungary
| | - Laszlo Dobson
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50A, 1083 Budapest, Hungary
| | - Bálint Péterfia
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50A, 1083 Budapest, Hungary
| | - Annamária F. Ángyán
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50A, 1083 Budapest, Hungary
| | - Balázs Ligeti
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50A, 1083 Budapest, Hungary
| | - Gergely Lukács
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50A, 1083 Budapest, Hungary
| | - Zoltán Gáspári
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50A, 1083 Budapest, Hungary
| |
Collapse
|
35
|
Nielsen LD, Pedersen CP, Erlendsson S, Teilum K. The Capsid Domain of Arc Changes Its Oligomerization Propensity through Direct Interaction with the NMDA Receptor. Structure 2019; 27:1071-1081.e5. [DOI: 10.1016/j.str.2019.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/28/2019] [Accepted: 03/31/2019] [Indexed: 12/20/2022]
|
36
|
Zhang W, Chuang YA, Na Y, Ye Z, Yang L, Lin R, Zhou J, Wu J, Qiu J, Savonenko A, Leahy DJ, Huganir R, Linden DJ, Worley PF. Arc Oligomerization Is Regulated by CaMKII Phosphorylation of the GAG Domain: An Essential Mechanism for Plasticity and Memory Formation. Mol Cell 2019; 75:13-25.e5. [PMID: 31151856 DOI: 10.1016/j.molcel.2019.05.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/27/2019] [Accepted: 05/01/2019] [Indexed: 12/17/2022]
Abstract
Arc is a synaptic protein essential for memory consolidation. Recent studies indicate that Arc originates in evolution from a Ty3-Gypsy retrotransposon GAG domain. The N-lobe of Arc GAG domain acquired a hydrophobic binding pocket in higher vertebrates that is essential for Arc's canonical function to weaken excitatory synapses. Here, we report that Arc GAG also acquired phosphorylation sites that can acutely regulate its synaptic function. CaMKII phosphorylates the N-lobe of the Arc GAG domain and disrupts an interaction surface essential for high-order oligomerization. In Purkinje neurons, CaMKII phosphorylation acutely reverses Arc's synaptic action. Mutant Arc that cannot be phosphorylated by CaMKII enhances metabotropic receptor-dependent depression in the hippocampus but does not alter baseline synaptic transmission or long-term potentiation. Behavioral studies indicate that hippocampus- and amygdala-dependent learning requires Arc GAG domain phosphorylation. These studies provide an atomic model for dynamic and local control of Arc function underlying synaptic plasticity and memory.
Collapse
Affiliation(s)
- Wenchi Zhang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yang-An Chuang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Youn Na
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zengyou Ye
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Liuqing Yang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Raozhou Lin
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiechao Zhou
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jing Wu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jessica Qiu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alena Savonenko
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel J Leahy
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Richard Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David J Linden
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul F Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
37
|
The Arc gene: Retroviral heritage in cognitive functions. Neurosci Biobehav Rev 2019; 99:275-281. [PMID: 30772431 DOI: 10.1016/j.neubiorev.2019.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/29/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022]
Abstract
Stabilization of neuronal plastic changes is mediated by transient gene expression, including transcription of the activity-regulated cytoskeleton-associated gene (Arc), also known as Arg 3.1. Arc is implicated in several types of synaptic plasticity, including synaptic scaling, long-term potentiation, and long-term depression. However, the precise mechanisms by which Arc mediates these forms of long-term plasticity are unclear. It was recently found that Arc protein is capable of forming capsid-like structures and of transferring its own mRNA to neighboring cells. Moreover, Arc mRNA undergoes activity-dependent translation in these "transfected" cells. These new data raise unexpected possibilities for the mechanisms of the Arc action, and many intriguing questions concerning the role of Arc transcellular traffic in neuronal plasticity. In this mini-review, we discuss a possible link between the role of Arc in learning and memory and the virus-like properties of this protein. Additionally, we highlight some of the emerging questions for future neurobiological studies and translational applications of Arc transsynaptic effects.
Collapse
|
38
|
Acuña-Hinrichsen F, Muñoz M, Hott M, Martin C, Mancilla E, Salazar P, Leyton L, Zambrano A, Concha MI, Burgos PV, Otth C. Herpes Simplex Virus Type 1 Enhances Expression of the Synaptic Protein Arc for Its Own Benefit. Front Cell Neurosci 2019; 12:505. [PMID: 30692913 PMCID: PMC6340317 DOI: 10.3389/fncel.2018.00505] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/06/2018] [Indexed: 12/30/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a neurotropic virus able to reach the central nervous system (CNS) after primary infection in oronasal mucosa. HSV-1 establishes latency inside neurons due the repression of its gene expression process, which is related to periodic reactivations in response to cellular stress conditions, constituting a risk factor for neurodegenerative diseases such as Alzheimer’s disease (AD). The immediate-early gene Arc plays an essential role in neuronal morphology, synaptic plasticity and memory formation. Arc acts as a hub protein, interacting with components of the endocytic machinery required for AMPA receptor (AMPAR) recycling as well as with proteins of the post-synaptic density and actin cytoskeleton. However, to date, no studies have evaluated whether persistent neurotropic HSV-1 infection modulates the expression or function of Arc protein in brain tissue. Here, we report that neuronal in vivo and in vitro infection of HSV-1 significantly increases Arc protein levels, showing a robust perinuclear distribution in neuronal cell lines, a process that is dependent on an active HSV-1 replication cycle. Finally, we found that silencing Arc protein caused a decrease in HSV-1 proteins and viral progeny, suggesting that Arc is involved in the lifecycle of HSV-1. Our studies strongly suggest that pathogenicity of HSV-1 neuronal reactivations in humans could be mediated in part by Arc neuronal upregulation and its potential role in endocytic trafficking and AMPA-neuronal function impairment. Further studies are necessary to define whether this phenomenon could have repercussions in cognition and learning processes in infected individuals.
Collapse
Affiliation(s)
| | - Mariela Muñoz
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Excelencia en Estudios Morfológicos y Quirúrgicos (CEMyQ), Universidad de La Frontera, Temuco, Chile
| | - Melissa Hott
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Martin
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Evelyn Mancilla
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Paula Salazar
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Luis Leyton
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Angara Zambrano
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.,Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
| | - Margarita I Concha
- Institute of Biochemistry and Microbiology, Faculty of Science, Universidad Austral de Chile, Valdivia, Chile
| | - Patricia V Burgos
- Institute of Physiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Centro de Biología Celular y Biomedicina, Facultad de Ciencia y Facultad de Medicina, Universidad San Sebastián, Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carola Otth
- Institute of Clinical Microbiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
39
|
Bitard‐Feildel T, Lamiable A, Mornon J, Callebaut I. Order in Disorder as Observed by the "Hydrophobic Cluster Analysis" of Protein Sequences. Proteomics 2018; 18:e1800054. [PMID: 30299594 PMCID: PMC7168002 DOI: 10.1002/pmic.201800054] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/29/2018] [Indexed: 12/17/2022]
Abstract
Hydrophobic cluster analysis (HCA) is an original approach for protein sequence analysis, which provides access to the foldable repertoire of the protein universe, including yet unannotated protein segments ("dark proteome"). Foldable segments correspond to ordered regions, as well as to intrinsically disordered regions (IDRs) undergoing disorder to order transitions. In this review, how HCA can be used to give insight into this last category of foldable segments is illustrated, with examples matching known 3D structures. After reviewing the HCA principles, examples of short foldable segments are given, which often contain short linear motifs, typically matching hydrophobic clusters. These segments become ordered upon contact with partners, with secondary structure preferences generally corresponding to those observed in the 3D structures within the complexes. Such small foldable segments are sometimes larger than the segments of known 3D structures, including flanking hydrophobic clusters that may be critical for interaction specificity or regulation, as well as intervening sequences allowing fuzziness. Cases of larger conditionally disordered domains are also presented, with lower density in hydrophobic clusters than well-folded globular domains or with exposed hydrophobic patches, which are stabilized by interaction with partners.
Collapse
Affiliation(s)
- Tristan Bitard‐Feildel
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
- Laboratoire de Biologie Computationnelle et Quantitative (LCQB)Institute of Biology Paris‐Seine (IBPS)Centre national de la recherche scientifique (CNRS)Sorbonne Université75005ParisFrance
| | - Alexis Lamiable
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
| | - Jean‐Paul Mornon
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
| | - Isabelle Callebaut
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC)Institut de recherche pour le développement (IRD)UMR CNRS 7590Muséum National d'Histoire NaturelleSorbonne Université75005ParisFrance
| |
Collapse
|
40
|
Goo BMSS, Sanstrum BJ, Holden DZY, Yu Y, James NG. Arc/Arg3.1 has an activity-regulated interaction with PICK1 that results in altered spatial dynamics. Sci Rep 2018; 8:14675. [PMID: 30279480 PMCID: PMC6168463 DOI: 10.1038/s41598-018-32821-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/25/2018] [Indexed: 01/28/2023] Open
Abstract
Activity-regulated cytoskeleton-associated protein (Arc; also known as Arg3.1) is an immediate early gene product that is transcribed in dendritic spines and, to date, has been best characterized as a positive regulator of AMPAR endocytosis during long-term depression (LTD) through interaction with endocytic proteins. Here, we show that protein interacting with C terminal kinase 1 (PICK1), a protein known to bind to the GluA2 subunit of AMPARs and associated with AMPAR trafficking, was pulled-down from brain homogenates and synaptosomes when using Arc as immobilized bait. Fluctuation and FLIM-FRET-Phasor analysis revealed direct interaction between these proteins when co-expressed that was increased under depolarizing conditions in live cells. At the plasma membrane, Arc-mCherry oligomerization was found to be concentration dependent. Additionally, co-expression of Arc-mCherry and EGFP-PICK1 followed by depolarizing conditions resulted in significant increases in the number and size of puncta containing both proteins. Furthermore, we identified the Arc binding region to be the first 126 amino acids of the PICK1 BAR domain. Overall, our data support a novel interaction and model where PICK1 mediates Arc regulation of AMPARs particularly under depolarizing conditions.
Collapse
Affiliation(s)
- Brandee M S S Goo
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, 651 Ilalo St., BSB 222, University of Hawaii, Honolulu, HI, 96813, USA
| | - Bethany J Sanstrum
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, 651 Ilalo St., BSB 222, University of Hawaii, Honolulu, HI, 96813, USA
| | - Diana Z Y Holden
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, 651 Ilalo St., BSB 222, University of Hawaii, Honolulu, HI, 96813, USA
| | | | - Nicholas G James
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, 651 Ilalo St., BSB 222, University of Hawaii, Honolulu, HI, 96813, USA.
| |
Collapse
|
41
|
Hallin EI, Eriksen MS, Baryshnikov S, Nikolaienko O, Grødem S, Hosokawa T, Hayashi Y, Bramham CR, Kursula P. Structure of monomeric full-length ARC sheds light on molecular flexibility, protein interactions, and functional modalities. J Neurochem 2018; 147:323-343. [DOI: 10.1111/jnc.14556] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/29/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Erik I. Hallin
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - Maria S. Eriksen
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Sergei Baryshnikov
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Oleksii Nikolaienko
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Sverre Grødem
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Tomohisa Hosokawa
- Department of Pharmacology; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Yasunori Hayashi
- Department of Pharmacology; Kyoto University Graduate School of Medicine; Kyoto Japan
| | - Clive R. Bramham
- Department of Biomedicine; University of Bergen; Bergen Norway
- K.G. Jebsen Centre for Neuropsychiatric Disorders; University of Bergen; Bergen Norway
| | - Petri Kursula
- Department of Biomedicine; University of Bergen; Bergen Norway
- Faculty of Biochemistry and Molecular Medicine; University of Oulu; Oulu Finland
| |
Collapse
|
42
|
Nakahata Y, Yasuda R. Plasticity of Spine Structure: Local Signaling, Translation and Cytoskeletal Reorganization. Front Synaptic Neurosci 2018; 10:29. [PMID: 30210329 PMCID: PMC6123351 DOI: 10.3389/fnsyn.2018.00029] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/07/2018] [Indexed: 12/31/2022] Open
Abstract
Dendritic spines are small protrusive structures on dendritic surfaces, and function as postsynaptic compartments for excitatory synapses. Plasticity of spine structure is associated with many forms of long-term neuronal plasticity, learning and memory. Inside these small dendritic compartments, biochemical states and protein-protein interactions are dynamically modulated by synaptic activity, leading to the regulation of protein synthesis and reorganization of cytoskeletal architecture. This in turn causes plasticity of structure and function of the spine. Technical advances in monitoring molecular behaviors in single dendritic spines have revealed that each signaling pathway is differently regulated across multiple spatiotemporal domains. The spatial pattern of signaling activity expands from a single spine to the nearby dendritic area, dendritic branch and the nucleus, regulating different cellular events at each spatial scale. Temporally, biochemical events are typically triggered by short Ca2+ pulses (~10–100 ms). However, these signals can then trigger activation of downstream protein cascades that can last from milliseconds to hours. Recent imaging studies provide many insights into the biochemical processes governing signaling events of molecular assemblies at different spatial localizations. Here, we highlight recent findings of signaling dynamics during synaptic plasticity and discuss their roles in long-term structural plasticity of dendritic spines.
Collapse
Affiliation(s)
- Yoshihisa Nakahata
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience (MPFI), Jupiter, FL, United States
| | - Ryohei Yasuda
- Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience (MPFI), Jupiter, FL, United States
| |
Collapse
|
43
|
Myrum C, Nikolaienko O, Bramham CR, Haavik J, Zayats T. Implication of the APP Gene in Intellectual Abilities. J Alzheimers Dis 2018; 59:723-735. [PMID: 28671113 PMCID: PMC5523840 DOI: 10.3233/jad-170049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Cognitive functions are highly heritable and polygenic, though the source of this genetic influence is unclear. On the neurobiological level, these functions rely on effective neuroplasticity, in which the activity-regulated cytoskeleton associated protein (ARC) plays an essential role. Objectives: To examine whether the ARC gene complex may contribute to the genetic components of intellectual function given the crucial role of ARC in brain plasticity and memory formation. Methods: The ARC complex was tested for association with intelligence (IQ) in children from the Avon Longitudinal Study of Parents and Children (ALSPAC, N = 5,165). As Alzheimer’s disease (AD) shares genetics with cognitive functioning, the association was followed up in an AD sample (17,008 cases, 37,154 controls). Results: The ARC complex revealed association with verbal and total IQ (empirical p = 0.027 and 0.041, respectively) in the ALSPAC. The strongest single variant signal (rs2830077; empirical p = 0.018), within the APP gene, was confirmed in the AD sample (p = 2.76E-03). Functional analyses of this variant showed its preferential binding to the transcription factor CP2. Discussion: This study implicates APP in childhood IQ. While follow-up studies are needed, this observation could help elucidate the etiology of disorders associated with cognitive dysfunction, such as AD.
Collapse
Affiliation(s)
- Craig Myrum
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Oleksii Nikolaienko
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Clive R Bramham
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jan Haavik
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Tetyana Zayats
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
44
|
Ashley J, Cordy B, Lucia D, Fradkin LG, Budnik V, Thomson T. Retrovirus-like Gag Protein Arc1 Binds RNA and Traffics across Synaptic Boutons. Cell 2018; 172:262-274.e11. [PMID: 29328915 PMCID: PMC5793882 DOI: 10.1016/j.cell.2017.12.022] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 08/15/2017] [Accepted: 12/18/2017] [Indexed: 12/31/2022]
Abstract
Arc/Arg3.1 is required for synaptic plasticity and cognition, and mutations in this gene are linked to autism and schizophrenia. Arc bears a domain resembling retroviral/retrotransposon Gag-like proteins, which multimerize into a capsid that packages viral RNA. The significance of such a domain in a plasticity molecule is uncertain. Here, we report that the Drosophila Arc1 protein forms capsid-like structures that bind darc1 mRNA in neurons and is loaded into extracellular vesicles that are transferred from motorneurons to muscles. This loading and transfer depends on the darc1-mRNA 3' untranslated region, which contains retrotransposon-like sequences. Disrupting transfer blocks synaptic plasticity, suggesting that transfer of dArc1 complexed with its mRNA is required for this function. Notably, cultured cells also release extracellular vesicles containing the Gag region of the Copia retrotransposon complexed with its own mRNA. Taken together, our results point to a trans-synaptic mRNA transport mechanism involving retrovirus-like capsids and extracellular vesicles.
Collapse
Affiliation(s)
- James Ashley
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Benjamin Cordy
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Diandra Lucia
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lee G Fradkin
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Vivian Budnik
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Travis Thomson
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
45
|
Pastuzyn ED, Day CE, Kearns RB, Kyrke-Smith M, Taibi AV, McCormick J, Yoder N, Belnap DM, Erlendsson S, Morado DR, Briggs JAG, Feschotte C, Shepherd JD. The Neuronal Gene Arc Encodes a Repurposed Retrotransposon Gag Protein that Mediates Intercellular RNA Transfer. Cell 2018; 172:275-288.e18. [PMID: 29328916 PMCID: PMC5884693 DOI: 10.1016/j.cell.2017.12.024] [Citation(s) in RCA: 328] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/15/2017] [Accepted: 12/18/2017] [Indexed: 12/25/2022]
Abstract
The neuronal gene Arc is essential for long-lasting information storage in the mammalian brain, mediates various forms of synaptic plasticity, and has been implicated in neurodevelopmental disorders. However, little is known about Arc's molecular function and evolutionary origins. Here, we show that Arc self-assembles into virus-like capsids that encapsulate RNA. Endogenous Arc protein is released from neurons in extracellular vesicles that mediate the transfer of Arc mRNA into new target cells, where it can undergo activity-dependent translation. Purified Arc capsids are endocytosed and are able to transfer Arc mRNA into the cytoplasm of neurons. These results show that Arc exhibits similar molecular properties to retroviral Gag proteins. Evolutionary analysis indicates that Arc is derived from a vertebrate lineage of Ty3/gypsy retrotransposons, which are also ancestors to retroviruses. These findings suggest that Gag retroelements have been repurposed during evolution to mediate intercellular communication in the nervous system.
Collapse
Affiliation(s)
- Elissa D Pastuzyn
- Department of Neurobiology and Anatomy, The University of Utah, Salt Lake City, UT, USA
| | - Cameron E Day
- Department of Neurobiology and Anatomy, The University of Utah, Salt Lake City, UT, USA
| | - Rachel B Kearns
- Department of Neurobiology and Anatomy, The University of Utah, Salt Lake City, UT, USA
| | - Madeleine Kyrke-Smith
- Department of Neurobiology and Anatomy, The University of Utah, Salt Lake City, UT, USA
| | - Andrew V Taibi
- Department of Neurobiology and Anatomy, The University of Utah, Salt Lake City, UT, USA
| | - John McCormick
- Department of Human Genetics, The University of Utah, Salt Lake City, UT, USA
| | - Nathan Yoder
- Department of Neurobiology and Anatomy, The University of Utah, Salt Lake City, UT, USA
| | - David M Belnap
- Department of Biochemistry, The University of Utah, Salt Lake City, UT, USA; Department of Biology, The University of Utah, Salt Lake City, UT, USA
| | - Simon Erlendsson
- Department of Biology, University of Copenhagen, Copenhagen, Denmark; MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | | | - Cédric Feschotte
- Department of Human Genetics, The University of Utah, Salt Lake City, UT, USA
| | - Jason D Shepherd
- Department of Neurobiology and Anatomy, The University of Utah, Salt Lake City, UT, USA; Department of Biochemistry, The University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
46
|
Lalonde J, Reis SA, Sivakumaran S, Holland CS, Wesseling H, Sauld JF, Alural B, Zhao WN, Steen JA, Haggarty SJ. Chemogenomic analysis reveals key role for lysine acetylation in regulating Arc stability. Nat Commun 2017; 8:1659. [PMID: 29162813 PMCID: PMC5698418 DOI: 10.1038/s41467-017-01750-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 10/11/2017] [Indexed: 01/11/2023] Open
Abstract
The role of Arc in synaptic plasticity and memory consolidation has been investigated for many years with recent evidence that defects in the expression or activity of this immediate-early gene may also contribute to the pathophysiology of brain disorders including schizophrenia and fragile X syndrome. These results bring forward the concept that reversing Arc abnormalities could provide an avenue to improve cognitive or neurological impairments in different disease contexts, but how to achieve this therapeutic objective has remained elusive. Here, we present results from a chemogenomic screen that probed a mechanistically diverse library of small molecules for modulators of BDNF-induced Arc expression in primary cortical neurons. This effort identified compounds with a range of influences on Arc, including promoting its acetylation-a previously uncharacterized post-translational modification of this protein. Together, our data provide insights into the control of Arc that could be targeted to harness neuroplasticity for clinical applications.
Collapse
Affiliation(s)
- Jasmin Lalonde
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA.
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, ON, Canada, N1G 2W1.
| | - Surya A Reis
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Sudhir Sivakumaran
- Department of Anatomy and Neurobiology, Boston University School of Medicine, 72 East Concord, Boston, MA, 02118, USA
| | - Carl S Holland
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Hendrik Wesseling
- Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - John F Sauld
- Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Begum Alural
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, 35210, Turkey
- Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, 35210, Turkey
| | - Wen-Ning Zhao
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA
| | - Judith A Steen
- Boston Children's Hospital, F.M. Kirby Center for Neurobiology, Harvard Medical School, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Massachusetts General Hospital, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Harvard Medical School, 185 Cambridge Street, Boston, MA, 02114, USA.
| |
Collapse
|
47
|
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: 42] [Impact Index Per Article: 5.3] [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
|
48
|
Shepherd JD. Arc - An endogenous neuronal retrovirus? Semin Cell Dev Biol 2017; 77:73-78. [PMID: 28941877 DOI: 10.1016/j.semcdb.2017.09.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 12/29/2022]
Abstract
The neuronal gene Arc is essential for long-lasting information storage in the mammalian brain and has been implicated in various neurological disorders. However, little is known about Arc's evolutionary origins. Recent studies suggest that mammalian Arc originated from a vertebrate lineage of Ty3/gypsy retrotransposons, which are also ancestral to retroviruses. In particular, Arc contains homology to the Gag polyprotein that forms the viral capsid and is essential for viral infectivity. This surprising connection raises the intriguing possibility that Arc may share molecular characteristics of retroviruses.
Collapse
Affiliation(s)
- Jason D Shepherd
- Department of Neurobiology and Anatomy, The University of Utah School of Medicine, 4539 SMBB, 36 S. Wasatch Dr., Salt Lake City, UT, 84112, United States.
| |
Collapse
|
49
|
Okuno H, Minatohara K, Bito H. Inverse synaptic tagging: An inactive synapse-specific mechanism to capture activity-induced Arc/arg3.1 and to locally regulate spatial distribution of synaptic weights. Semin Cell Dev Biol 2017; 77:43-50. [PMID: 28939038 DOI: 10.1016/j.semcdb.2017.09.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/15/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022]
Abstract
Long-lasting forms of synaptic plasticity such as long-term potentiation (LTP) and long-term depression (LTD) are fundamental cellular mechanisms underlying learning and memory. The synaptic tagging and capture (STC) hypothesis has provided a theoretical framework on how products of activity-dependent genes may interact with potentiated synapses to facilitate and maintain such long-lasting synaptic plasticity. Although Arc/arg3.1 was initially assumed to participate in STC processes during LTP, accumulating evidence indicated that Arc/arg3.1 might rather contribute in weakening of synaptic weights than in their strengthening. In particular, analyses of Arc/Arg3.1 protein dynamics and function in the dendrites after plasticity-inducing stimuli have revealed a new type of inactivity-dependent redistribution of synaptic weights, termed "inverse synaptic tagging". The original synaptic tagging and inverse synaptic tagging likely co-exist and are mutually non-exclusive mechanisms, which together may help orchestrate the redistribution of synaptic weights and promote the enhancement and maintenance of their contrast between potentiated and non-potentiated synapses during the late phase of long-term synaptic plasticity. In this review, we describe the inverse synaptic tagging mechanism that controls synaptic dynamics of Arc/Arg3.1, an immediate early gene product which is captured and preferentially targeted to non-potentiated synapses, and discuss its impact on neuronal circuit refinement and cognitive function.
Collapse
Affiliation(s)
- Hiroyuki Okuno
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Keiichiro Minatohara
- SK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
50
|
Managò F, Papaleo F. Schizophrenia: What's Arc Got to Do with It? Front Behav Neurosci 2017; 11:181. [PMID: 28979198 PMCID: PMC5611489 DOI: 10.3389/fnbeh.2017.00181] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/11/2017] [Indexed: 01/08/2023] Open
Abstract
Human studies of schizophrenia are now reporting a previously unidentified genetic convergence on postsynaptic signaling complexes such as the activity-regulated cytoskeletal-associated (Arc) gene. However, because this evidence is still very recent, the neurobiological implication of Arc in schizophrenia is still scattered and unrecognized. Here, we first review current and developing findings connecting Arc in schizophrenia. We then highlight recent and previous findings from preclinical mouse models that elucidate how Arc genetic modifications might recapitulate schizophrenia-relevant behavioral phenotypes following the novel Research Domain Criteria (RDoC) framework. Building on this, we finally compare and evaluate several lines of evidence demonstrating that Arc genetics can alter both glutamatergic and dopaminergic systems in a very selective way, again consistent with molecular alterations characteristic of schizophrenia. Despite being only initial, accumulating and compelling data are showing that Arc might be one of the primary biological players in schizophrenia. Synaptic plasticity alterations in the genetic architecture of psychiatric disorders might be a rule, not an exception. Thus, we anticipate that additional evidence will soon emerge to clarify the Arc-dependent mechanisms involved in the psychiatric-related dysfunctional behavior.
Collapse
Affiliation(s)
- Francesca Managò
- Department of Neuroscience and Brain Technologies, Istituto Italiano di TecnologiaGenova, Italy
| | - Francesco Papaleo
- Department of Neuroscience and Brain Technologies, Istituto Italiano di TecnologiaGenova, Italy
| |
Collapse
|