1
|
Liu S, Feng A, Li Z. Neuron-Derived Extracellular Vesicles: Emerging Regulators in Central Nervous System Disease Progression. Mol Neurobiol 2025:10.1007/s12035-025-05010-4. [PMID: 40325332 DOI: 10.1007/s12035-025-05010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
The diagnosis and exploration of central nervous system (CNS) diseases remain challenging due to the blood-brain barrier (BBB), complex signaling pathways, and heterogeneous clinical manifestations. Neurons, as the core functional units of the CNS, play a pivotal role in CNS disease progression. Extracellular vesicles (EVs), capable of crossing the BBB, facilitate intercellular and cell-extracellular matrix (ECM) communication, making neuron-derived extracellular vesicles (NDEVs) a focal point of research. Recent studies reveal that NDEVs, carrying various bioactive substances, can exert either pathogenic or protective effects in numerous CNS diseases. Additionally, NDEVs show significant potential as biomarkers for CNS diseases. This review summarizes the emerging roles of NDEVs in CNS diseases, including Alzheimer's disease, depression, traumatic brain injury, schizophrenia, ischemic stroke, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. It aims to provide a novel perspective on developing therapeutic and diagnostic strategies for CNS diseases through the study of NDEVs.
Collapse
Affiliation(s)
- Sitong Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Aitong Feng
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zhigang Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen, 518107, China.
| |
Collapse
|
2
|
Chang NC, Wells JN, Wang AY, Schofield P, Huang YC, Truong VH, Simoes-Costa M, Feschotte C. Gag proteins encoded by endogenous retroviruses are required for zebrafish development. Proc Natl Acad Sci U S A 2025; 122:e2411446122. [PMID: 40294259 DOI: 10.1073/pnas.2411446122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/21/2025] [Indexed: 04/30/2025] Open
Abstract
Transposable elements (TEs) make up the bulk of eukaryotic genomes and examples abound of TE-derived sequences repurposed for organismal function. The process by which TEs become coopted remains obscure because most cases involve ancient, transpositionally inactive elements. Reports of active TEs serving beneficial functions are scarce and often contentious due to difficulties in manipulating repetitive sequences. Here, we show that recently active TEs in zebrafish encode products critical for embryonic development. Knockdown and rescue experiments demonstrate that the endogenous retrovirus family BHIKHARI-1 (Bik-1) encodes a Gag protein essential for mesoderm development. Mechanistically, Bik-1 Gag associates with the cell membrane, and its ectopic expression in chicken embryos alters cell migration. Similarly, depletion of BHIKHARI-2 Gag, a relative of Bik-1, causes defects in neural crest development in zebrafish. We propose an "addiction" model to explain how active TEs can be integrated into conserved developmental processes.
Collapse
Affiliation(s)
- Ni-Chen Chang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850
| | - Jonathan N Wells
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850
| | - Andrew Y Wang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850
| | - Phillip Schofield
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850
| | - Yi-Chia Huang
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115
| | - Vinh H Truong
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850
| | - Marcos Simoes-Costa
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115
| | - Cédric Feschotte
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14850
| |
Collapse
|
3
|
Beamish CR, Becker J, Tam LM, Love T, Rand MD. Transcriptomic analysis identifies muscle-specific mitochondrial and vesicular transport genes as methylmercury toxicity targets in a Drosophila model of congenital Minamata disease. Toxicol Sci 2025; 205:106-123. [PMID: 39951334 PMCID: PMC12038250 DOI: 10.1093/toxsci/kfaf018] [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] [Indexed: 04/30/2025] Open
Abstract
Prenatal methylmercury (MeHg) exposure presents a heightened concern in early human development, as has been exemplified in historic cases of congenital minimata disease (CMD). Children who experience CMD characteristically present with various degrees of cognitive and motor symptoms and signs, much like cerebral palsy. MeHg has thus been characterized as a neurotoxicant, where motor deficits are ascribed to central nervous system targets. Skeletal muscle as a post-synaptic MeHg target and contributor to the etiology of CMD has garnered far less attention. Prior studies using Drosophila to model CMD revealed that developmental exposure of MeHg in the larval/pupal stages can elicit graded and latent dose responses affecting adult flight behavior at lower doses (0.4-2.5 ppm in food) and eclosion (emergence from the pupa case) at higher doses (>2.5 ppm in food). The latter phenotype is accompanied by dysmorphogenesis of skeletal muscles. Here, we investigate respective roles for muscle and neural targets in MeHg toxicity. Using RNA-seq analysis, we find that developmental MeHg exposure produces 10 times as many differentially expressed transcripts in indirect flight muscle compared to the ventral nerve cord. Among known MeHg response genes, Nrf2 antioxidant response pathway genes showed muscle-specific MeHg-induced expression changes. Within the muscle transcriptome, the most enriched and significant Gene Ontology terms identified genes required for mitochondrial ribosomal translation at the pupa stage and mitochondrial function (respiratory chain complex I) and vesicle trafficking (ESCRT III) pathways in adults, all showing decreased expression with MeHg exposure. By using an intact, whole-animal developmental model, we identify preferential candidates to evaluate a novel role for muscle-specific mitochondria and intercellular vesicular communication mechanisms as targets in MeHg toxicity and the etiology of CMD.
Collapse
Affiliation(s)
- Catherine R Beamish
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, United States
| | - Jennifer Becker
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, United States
| | - Lok Ming Tam
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, United States
| | - Tanzy Love
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, United States
| |
Collapse
|
4
|
Li M, Cheng L. Mapping Fear-Related Neural Activity and Circuitry Changes Following Prophylactic Administration of (R,S)-Ketamine and (2S,6S)-Hydroxynorketamine. Neurosci Bull 2025; 41:925-928. [PMID: 39923210 PMCID: PMC12014964 DOI: 10.1007/s12264-025-01362-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/08/2025] [Indexed: 02/10/2025] Open
Affiliation(s)
- Minzhu Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Li Cheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
5
|
Rousseau C, Morand T, Haas G, Lauret E, Kuhn L, Chicher J, Hammann P, Meignin C. In vivo Dicer-2 interactome during viral infection reveals novel pro and antiviral factors in Drosophila melanogaster. PLoS Pathog 2025; 21:e1013093. [PMID: 40334246 PMCID: PMC12058146 DOI: 10.1371/journal.ppat.1013093] [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: 03/25/2024] [Accepted: 04/01/2025] [Indexed: 05/09/2025] Open
Abstract
RNA interference has a major role in the control of viral infection in insects. It is initialized by the sensing of double stranded RNA (dsRNA) by the RNAse III enzyme Dicer-2. Many in vitro studies have helped understand how Dicer-2 discriminates between different dsRNA substrate termini, however it is unclear whether the same mechanisms are at work in vivo, and notably during recognition of viral dsRNA. Indeed, although Dicer-2 associates with several dsRNA-binding proteins (dsRBPs) that can modify its specificity for a substrate, it remains unknown how Dicer-2 is able to recognize the protected termini of viral dsRNAs. In order to study how the ribonucleoprotein network of Dicer-2 impacts antiviral immunity, we used an IP-MS approach to identify in vivo interactants of different versions of GFP::Dicer-2 in transgenic lines. We provide a global overview of the partners of Dicer-2 in vivo, and reveal how this interactome is modulated by different factors such as viral infection and/or different point mutations inactivating the helicase or RNase III domains of GFP::Dicer-2. Our analysis uncovers several previously unknown Dicer-2 interactants associated with RNA granules, i.e., Me31B, Rump, eIF4E1, eIF4G1, Rin and Syncrip. Functional characterization of the candidates, both in cells and in vivo, reveals pro- and antiviral factors in the context of an infection by the picorna-like DCV virus. This work highlights protein complexes assembled around Dicer-2 in vivo, and provides a resource to investigate their contribution to antiviral RNAi and related pathways.
Collapse
Affiliation(s)
- Claire Rousseau
- Université de Strasbourg, M3i CNRS UPR9022, Strasbourg, France
| | - Thomas Morand
- Université de Strasbourg, M3i CNRS UPR9022, Strasbourg, France
| | - Gabrielle Haas
- Université de Strasbourg, M3i CNRS UPR9022, Strasbourg, France
| | - Emilie Lauret
- Université de Strasbourg, M3i CNRS UPR9022, Strasbourg, France
| | - Lauriane Kuhn
- Plateforme Protéomique Strasbourg-Esplanade, Université de Strasbourg, CNRS UAR1589, Strasbourg, France
| | - Johana Chicher
- Plateforme Protéomique Strasbourg-Esplanade, Université de Strasbourg, CNRS UAR1589, Strasbourg, France
| | - Philippe Hammann
- Plateforme Protéomique Strasbourg-Esplanade, Université de Strasbourg, CNRS UAR1589, Strasbourg, France
| | - Carine Meignin
- Université de Strasbourg, M3i CNRS UPR9022, Strasbourg, France
| |
Collapse
|
6
|
Mastrodonato A, Jin M, Kee N, Lanio M, Tapia J, Quintana L, Muñoz Zamora A, Deng SX, Xu X, Landry DW, Denny CA. Prophylactic (R,S)-Ketamine and (2S,6S)-Hydroxynorketamine Decrease Fear Expression by Differentially Modulating Fear Neural Ensembles. Biol Psychiatry 2025; 97:887-899. [PMID: 39389408 PMCID: PMC11978926 DOI: 10.1016/j.biopsych.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND We previously reported that a single injection of (R,S)-ketamine or its metabolite (2S,6S)-hydroxynorketamine (HNK) prior to stress attenuated learned fear. However, whether these drugs attenuate learned fear through divergent or convergent effects on neural activity remains to be determined. METHODS 129S6/SvEv male mice were injected with saline, (R,S)-ketamine, or (2S,6S)-HNK 1 week before a 3-shock contextual fear conditioning paradigm. Five days later, mice were re-exposed to the aversive context and euthanized 1 hour later to quantify active cells. Brains were processed for c-fos immunoreactivity, and neural networks were built with a novel, wide-scale imaging pipeline. RESULTS We found that (R,S)-ketamine and (2S,6S)-HNK attenuated learned fear. Fear-related neural activity was altered in dorsal CA3 following (2S,6S)-HNK; ventral CA3 and CA1, infralimbic and prelimbic regions, insular cortex, retrosplenial cortex, piriform cortex, nucleus reuniens, and periaqueductal gray following both (R,S)-ketamine and (2S,6S)-HNK; and in the paraventricular nucleus of the thalamus (PVT) following (R,S)-ketamine. Dorsal CA3 and ventral hippocampus activation correlated with freezing in the (R,S)-ketamine group, and retrosplenial cortex activation correlated with freezing in both (R,S)-ketamine and (2S,6S)-HNK groups. (R,S)-ketamine increased connectivity between cortical and subcortical regions while (2S,6S)-HNK increased connectivity within these regions. CONCLUSIONS This work identifies novel nodes in fear networks that involve the nucleus reuniens, piriform cortex, insular cortex, periaqueductal gray, and retrosplenial cortex that can be targeted with neuromodulatory strategies or pharmaceutical compounds to treat fear-induced disorders. This approach could be used to optimize target engagement and dosing strategies of existing medications.
Collapse
Affiliation(s)
- Alessia Mastrodonato
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York; MIND Area, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York.
| | - Michelle Jin
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, New York
| | - Noelle Kee
- Department of Neuroscience and Behavior, Barnard College, New York, New York
| | - Marcos Lanio
- Neurobiology and Behavior Graduate Program, Columbia University, New York, New York; Medical Scientist Training Program, Columbia University Irving Medical Center, New York, New York
| | - Juliana Tapia
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York
| | - Liliette Quintana
- Department of Neuroscience and Behavior, Barnard College, New York, New York
| | - Andrea Muñoz Zamora
- Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York
| | - Shi-Xian Deng
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Xiaoming Xu
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Donald W Landry
- Department of Medicine, Columbia University Irving Medical Center, New York, New York; Organic Chemistry Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Christine A Denny
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York; Division of Systems Neuroscience, Area Neuroscience, Research Foundation for Mental Hygiene, Inc./New York State Psychiatric Institute, New York, New York.
| |
Collapse
|
7
|
Wood TWP, Henriques WS, Cullen HB, Romero M, Blengini CS, Sarathy S, Sorkin J, Bekele H, Jin C, Kim S, Wang X, Laureau R, Chemiakine A, Khondker RC, Isola JVV, Stout MB, Gennarino VA, Mogessie B, Jain D, Schindler K, Suh Y, Wiedenheft B, Berchowitz LE. The retrotransposon-derived capsid genes PNMA1 and PNMA4 maintain reproductive capacity. NATURE AGING 2025:10.1038/s43587-025-00852-y. [PMID: 40263616 DOI: 10.1038/s43587-025-00852-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 03/05/2025] [Indexed: 04/24/2025]
Abstract
Almost half of the human genome consists of retrotransposons-'parasitic' sequences that insert themselves into the host genome via an RNA intermediate. Although most of these sequences are silenced or mutationally deactivated, they can present opportunities for evolutionary innovation: mutation of a deteriorating retrotransposon can result in a gene that provides a selective advantage to the host in a process termed 'domestication'1-3. The PNMA family of gag-like capsid genes was domesticated from an ancient vertebrate retrotransposon of the Metaviridae clade at least 100 million years ago4,5. PNMA1 and PNMA4 are positively regulated by the master germ cell transcription factors MYBL1 and STRA8, and their transcripts are bound by the translational regulator DAZL during gametogenesis6. This developmental regulation of PNMA1 and PNMA4 expression in gonadal tissue suggested to us that they might serve a reproductive function. Through the analysis of donated human ovaries, genome-wide association studies (GWASs) and mouse models, we found that PNMA1 and PNMA4 are necessary for the maintenance of a normal reproductive lifespan. These proteins self-assemble into capsid-like structures that exit human cells, and we observed large PNMA4 particles in mouse male gonadal tissue that contain RNA and are consistent with capsid formation.
Collapse
Affiliation(s)
- Thomas W P Wood
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - William S Henriques
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Harrison B Cullen
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Mayra Romero
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Cecilia S Blengini
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Shreya Sarathy
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Julia Sorkin
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Hilina Bekele
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Chen Jin
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Seungsoo Kim
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Raphaelle Laureau
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Alexei Chemiakine
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Rishad C Khondker
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Vincenzo A Gennarino
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, New York, NY, USA
- Initiative for Columbia Ataxia and Tremor, New York, NY, USA
| | - Binyam Mogessie
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Devanshi Jain
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Karen Schindler
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Yousin Suh
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Blake Wiedenheft
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Luke E Berchowitz
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
- Taub Institute for Research on Alzheimer's and the Aging Brain, New York, NY, USA.
| |
Collapse
|
8
|
Kanhema T, Parobczak K, Patil S, Holm-Kaczmarek D, Hallin EI, Ludwiczak J, Szczepankiewicz AA, Pauzin FP, Mahboob A, Szum A, Ishizuka Y, Dunin-Horkawicz S, Kursula P, Wilczynski G, Magalska A, Bramham CR. ARC/ARG3.1 binds the nuclear polyadenylate-binding protein RRM and regulates neuronal activity-dependent formation of nuclear speckles. Cell Rep 2025; 44:115525. [PMID: 40208793 DOI: 10.1016/j.celrep.2025.115525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 01/30/2025] [Accepted: 03/14/2025] [Indexed: 04/12/2025] Open
Abstract
ARC is a neuronal activity-induced protein interaction hub with critical roles in synaptic plasticity and memory. ARC localizes to synapses and the nucleus, but its nuclear functions are little known. Following in vivo long-term potentiation (LTP) induction in the dentate gyrus, we show that ARC accumulates in the nucleosol fraction and interchromatin space of granule cells. Proteomic analysis of immunoprecipitated ARC complexes identifies proteins involved in pre-mRNA processing. We demonstrate endogenous ARC protein-protein interaction with polyadenylate-binding nuclear protein 1 (PABPN1) and the paraspeckles protein polypyrimidine tract-binding protein (PTB)-associated splicing factor (PSF). In vitro peptide binding arrays show direct binding of purified ARC to the PABPN1 poly(A)-RNA recognition motif. 3D morphometric imaging reveals structural changes in PABPN1 foci corresponding to classical nuclear speckles following in vivo and in vitro LTP. Depletion of ARC disrupts the maintenance and activity-dependent formation of PABPN1 speckles, thus implicating ARC in regulation of nuclear speckle dynamics and pre-mRNA processing.
Collapse
Affiliation(s)
- Tambudzai Kanhema
- Department of Biomedicine, University of Bergen, Bergen, Norway; Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Kamil Parobczak
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Sudarshan Patil
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Dagmara Holm-Kaczmarek
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Erik I Hallin
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jan Ludwiczak
- Institute of Evolutionary Biology, Faculty of Biology, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland; Prescient Design, Genentech Research & Early Development, Roche Group, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Andrzej Antoni Szczepankiewicz
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Francois Philippe Pauzin
- Department of Biomedicine, University of Bergen, Bergen, Norway; Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Aamra Mahboob
- Department of Biomedicine, University of Bergen, Bergen, Norway; Mohn Research Center for the Brain, University of Bergen, Bergen, Norway
| | - Adrian Szum
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Yuta Ishizuka
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Stanisław Dunin-Horkawicz
- Institute of Evolutionary Biology, Faculty of Biology, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland; Department of Protein Evolution, Max Planck Institute for Biology Tübingen, Tübingen, Germany
| | - Petri Kursula
- Department of Biomedicine, University of Bergen, Bergen, Norway; Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; LINXS Institute of Advanced Neutron and X-ray Science, Lund, Sweden
| | - Grzegorz Wilczynski
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Adriana Magalska
- Laboratory of Molecular and Systemic Neuromorphology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland; Laboratory of Spatial Epigenetics, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland.
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Bergen, Norway; Mohn Research Center for the Brain, University of Bergen, Bergen, Norway.
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Mozafari S, Peruzzotti-Jametti L, Pluchino S. Mitochondria transfer for myelin repair. J Cereb Blood Flow Metab 2025:271678X251325805. [PMID: 40079508 PMCID: PMC11907575 DOI: 10.1177/0271678x251325805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/15/2025]
Abstract
Demyelination is a common feature of neuroinflammatory and degenerative diseases of the central nervous system (CNS), such as multiple sclerosis (MS). It is often linked to disruptions in intercellular communication, bioenergetics and metabolic balance accompanied by mitochondrial dysfunction in cells such as oligodendrocytes, neurons, astrocytes, and microglia. Although current MS treatments focus on immunomodulation, they fail to stop or reverse demyelination's progression. Recent advancements highlight intercellular mitochondrial exchange as a promising therapeutic target, with potential to restore metabolic homeostasis, enhance immunomodulation, and promote myelin repair. With this review we will provide insights into the CNS intercellular metabolic decoupling, focusing on the role of mitochondrial dysfunction in neuroinflammatory demyelinating conditions. We will then discuss emerging cell-free biotherapies exploring the therapeutic potential of transferring mitochondria via biogenic carriers like extracellular vesicles (EVs) or synthetic liposomes, aimed at enhancing mitochondrial function and metabolic support for CNS and myelin repair. Lastly, we address the key challenges for the clinical application of these strategies and discuss future directions to optimize mitochondrial biotherapies. The advancements in this field hold promise for restoring metabolic homeostasis, and enhancing myelin repair, potentially transforming the therapeutic landscape for neuroinflammatory and demyelinating diseases.
Collapse
Affiliation(s)
- Sabah Mozafari
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Hermann DM, Wang C, Mohamud Yusuf A, Herz J, Doeppner TR, Giebel B. Extracellular vesicles lay the ground for neuronal plasticity by restoring mitochondrial function, cell metabolism and immune balance. J Cereb Blood Flow Metab 2025:271678X251325039. [PMID: 40072028 PMCID: PMC11904928 DOI: 10.1177/0271678x251325039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/15/2025] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs) convey complex signals between cells that can be used to promote neuronal plasticity and neurological recovery in brain disease models. These EV signals are multimodal and context-dependent, making them unique therapeutic principles. This review analyzes how EVs released from various cell sources control neuronal metabolic function, neuronal survival and plasticity. Preferential sites of EV communication in the brain are interfaces between pre- and postsynaptic neurons at synapses, between astrocytes and neurons at plasma membranes or tripartite synapses, between oligodendrocytes and neurons at axons, between microglial cells/macrophages and neurons, and between cerebral microvascular cells and neurons. At each of these interfaces, EVs support mitochondrial function and cell metabolism under physiological conditions and orchestrate neuronal survival and plasticity in response to brain injury. In the injured brain, the promotion of neuronal survival and plasticity by EVs is tightly linked with EV actions on mitochondrial function, cell metabolism, oxidative stress and immune responses. Via the stabilization of cell metabolism and immune balance, neuronal plasticity responses are activated and functional neurological recovery is induced. As such, EV lay the ground for neuronal plasticity.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Pediatrics I, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thorsten R Doeppner
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Department of Neurology, University Hospital Gießen and Marburg, Justus-Liebig-University Gießen, Gießen, Germany
| |
Collapse
|
13
|
Xie ZF, Wang SY, Gao Y, Zhang YD, Han YN, Huang J, Gao MN, Wang CG. Vagus nerve stimulation (VNS) preventing postoperative cognitive dysfunction (POCD): two potential mechanisms in cognitive function. Mol Cell Biochem 2025; 480:1343-1357. [PMID: 39138750 DOI: 10.1007/s11010-024-05091-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Postoperative cognitive dysfunction (POCD) impacts a significant number of patients annually, frequently impairing their cognitive abilities and resulting in unfavorable clinical outcomes. Aimed at addressing cognitive impairment, vagus nerve stimulation (VNS) is a therapeutic approach, which was used in many mental disordered diseases, through the modulation of vagus nerve activity. In POCD model, the enhancement of cognition function provided by VNS was shown, demonstrating VNS effect on cognition in POCD. In the present study, we primarily concentrates on elucidating the role of the VNS improving the cognitive function in POCD, via two potential mechanisms: the inflammatory microenvironment and epigenetics. This study provided a theoretical support for the feasibility that VNS can be a potential method to enhance cognition function in POCD.
Collapse
Affiliation(s)
- Zi-Feng Xie
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
- The First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Sheng-Yu Wang
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Graduate College, Chengde Medical College, Chengde, 067000, Hebei, China
| | - Yuan Gao
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
- The First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Yi-Dan Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
- The First Clinical Medical College, Jinzhou Medical University, Jinzhou, 121000, Liaoning, China
| | - Ya-Nan Han
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Graduate College, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Jin Huang
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
- Graduate College, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Mei-Na Gao
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China
| | - Chun-Guang Wang
- Department of Anesthesiology, The First Central Hospital of Baoding, Northern Great Wall Street 320#, Baoding, 071000, Hebei, China.
| |
Collapse
|
14
|
Garat J, Di Paolo A, Eastman G, Castillo PE, Sotelo-Silveira J. The Trail of Axonal Protein Synthesis: Origins and Current Functional Landscapes. Neuroscience 2025; 567:195-208. [PMID: 39755230 DOI: 10.1016/j.neuroscience.2024.12.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/03/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
Local protein synthesis (LPS) in axons is now recognized as a physiological process, participating both in the maintenance of axonal function and diverse plastic phenomena. In the last decades of the 20th century, the existence and function of axonal LPS were topics of significant debate. Very early, axonal LPS was thought not to occur at all and was later accepted to play roles only during development or in response to specific conditions. However, compelling evidence supports its essential and pervasive role in axonal function in the mature nervous system. Remarkably, in the last five decades, Uruguayan neuroscientists have contributed significantly to demonstrating axonal LPS by studying motor and sensory axons of the peripheral nervous system of mammals, as well as giant axons of the squid and the Mauthner cell of fish. For LPS to occur, a highly regulated transport system must deliver the necessary macromolecules, such as mRNAs and ribosomes. This review discusses key findings related to the localization and abundance of axonal mRNAs and their translation levels, both in basal states and in response to physiological processes, such as learning and memory consolidation, as well as neurodevelopmental and neurodegenerative disorders, including Alzheimer's disease, autism spectrum disorder, and axonal injury. Moreover, we discuss the current understanding of axonal ribosomes, from their localization to the potential roles of locally translated ribosomal proteins, in the context of emerging research that highlights the regulatory roles of the ribosome in translation. Lastly, we address the main challenges and open questions for future studies.
Collapse
Affiliation(s)
- Joaquin Garat
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, MEC, Av. Italia 3318, Montevideo, CP 11600, Uruguay
| | - Andres Di Paolo
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, MEC, Av. Italia 3318, Montevideo, CP 11600, Uruguay
| | - Guillermo Eastman
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, MEC, Av. Italia 3318, Montevideo, CP 11600, Uruguay; Department of Biology, University of Virginia, 485 McCormick Rd, Charlottesville, VA, 22904, USA
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Psychiatry & Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - José Sotelo-Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable, MEC, Av. Italia 3318, Montevideo, CP 11600, Uruguay; Departamento de Biología Celular y Molecular, Facultad de Ciencias, Universidad de la República, Iguá, Montevideo, 4225, CP 11400, Uruguay.
| |
Collapse
|
15
|
Shugarts Devanapally NM, Sathya A, Yi AL, Chan WM, Marre JA, Jose AM. Intergenerational transport of double-stranded RNA in C. elegans can limit heritable epigenetic changes. eLife 2025; 13:RP99149. [PMID: 39902803 PMCID: PMC11793870 DOI: 10.7554/elife.99149] [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] [Indexed: 02/06/2025] Open
Abstract
RNAs in circulation carry sequence-specific regulatory information between cells in plant, animal, and host-pathogen systems. Such RNA can cross generational boundaries, as evidenced by somatic double-stranded RNA (dsRNA) in the nematode Caenorhabditis elegans silencing genes of matching sequence in progeny. Here we dissect the intergenerational path taken by dsRNA from parental circulation and discover that cytosolic import through the dsRNA importer SID-1 in the parental germline and/or developing progeny varies with developmental time and dsRNA substrates. Loss of SID-1 enhances initiation of heritable RNA silencing within the germline and causes changes in the expression of the sid-1-dependent gene sdg-1 that last for more than 100 generations after restoration of SID-1. The SDG-1 protein is enriched in perinuclear germ granules required for heritable RNA silencing but is expressed from a retrotransposon targeted by such silencing. This auto-inhibitory loop suggests how retrotransposons could persist by hosting genes that regulate their own silencing.
Collapse
Affiliation(s)
| | - Aishwarya Sathya
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Andrew L Yi
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Winnie M Chan
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Julia A Marre
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| | - Antony M Jose
- Department of Cell Biology and Molecular Genetics, University of MarylandCollege ParkUnited States
| |
Collapse
|
16
|
Yee G, Wu R, Oshi M, Endo I, Ishikawa T, Takabe K. Activity-Regulated Cytoskeleton-Associated Protein Gene Expression Is Associated With High Infiltration of Stromal Cells and Immune Cells, but With Less Cancer Cell Proliferation and Better Overall Survival in Estrogen Receptor-Positive/Human Epidermal Growth Factor Receptor 2-Negative Breast Cancers. World J Oncol 2025; 16:16-29. [PMID: 39850523 PMCID: PMC11750752 DOI: 10.14740/wjon1936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/03/2025] [Indexed: 01/25/2025] Open
Abstract
Background Peritumoral lidocaine infiltration prior to excision is associated with better survival in breast cancer (BC), which led us to hypothesize that innervation to the tumor affects its biology and patient survival. Activity-regulated cytoskeleton-associated protein (ARC) gene expression is known to be regulated by neuronal activity. Therefore, we studied the clinical relevance of ARC gene expression as a surrogate of neuronal activity in BC. Methods Sweden Cancerome Analysis Network - Breast (SCAN-B (GSE96058), n = 3,273) cohort and The Cancer Genome Atlas (TCGA, n = 1,069) were analyzed. Results High ARC expression was significantly associated with smaller tumor size, without lymph node metastasis, and less stage IV disease in one cohort, but not validated by the other. Estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) and luminal A expressed significantly higher ARC compared to the other subtypes in both cohorts (P < 0.005). High ARC BC was significantly associated with lower Nottingham histological grade and lower Ki67 gene expression consistently in ER+/HER2- but not triple negative breast cancer (TNBC) in both cohorts (P < 0.001). Cell proliferation-related gene sets in the Hallmark collection (E2F targets, G2M checkpoint, and mitotic spindle) were significantly enriched to low ARC BC in ER+/HER2- but not TNBC in TCGA. The stromal cells (fibroblasts, vascular endothelial cells, and adipocytes) were all significantly infiltrated in high ARC ER+/HER2-, but not in TNBC, except for neurons. Homologous recombination deficiency, intratumor heterogeneity, fraction altered, silent or non-silent mutation rate were all significantly lower in high ARC ER+/HER2- but not TNBC. Although there was no difference in single nucleotide variant or indel neoantigens, tumor infiltrating lymphocytes, and cytolytic activity by ARC expression regardless of subtype, multiple immune cells were significantly infiltrated in high ARC ER+/HER2-, including CD8, CD4 memory cells, helper type II T cells, regulatory T cells, M2 macrophages, and B cells (all P < 0.03 in both cohorts), but not in TNBC. Disease-specific and overall survival were significantly improved in high ARC ER+/HER2- consistently in both cohorts (all P < 0.05), but this was not the case in TNBC. Conclusion ARC gene expression was associated with less cancer cell proliferation, high infiltration of stromal cells and immune cells, and better survival in the ER+/HER2- but not TNBC subtype.
Collapse
Affiliation(s)
- Gabrielle Yee
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
- These authors equally contributed to this manuscript
| | - Rongrong Wu
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- These authors equally contributed to this manuscript
| | - Masanori Oshi
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
17
|
M’Angale PG, Lemieux A, Liu Y, Wang S, Zinter M, Alegre G, Simkin A, Budnik V, Kelch BA, Thomson T. Capsid transfer of the retrotransposon Copia controls structural synaptic plasticity in Drosophila. PLoS Biol 2025; 23:e3002983. [PMID: 39964983 PMCID: PMC11835246 DOI: 10.1371/journal.pbio.3002983] [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: 08/21/2024] [Accepted: 12/11/2024] [Indexed: 02/20/2025] Open
Abstract
Transposons are parasitic genome elements that can also serve as raw material for the evolution of new cellular functions. However, how retrotransposons are selected and domesticated by host organisms to modulate synaptic plasticity remains largely unknown. Here, we show that the Ty1 retrotransposon Copia forms virus-like capsids in vivo and transfers between cells. Copia is enriched at the Drosophila neuromuscular junction (NMJ) and transported across synapses, and disrupting its expression promotes both synapse development and structural synaptic plasticity. We show that proper synaptic plasticity is maintained in Drosophila by the balance of Copia and the Arc1 (activity-regulated cytoskeleton-associated protein) homolog. High-resolution cryogenic-electron microscopy imaging shows that the structure of the Copia capsid has a large capacity and pores like retroviruses but is distinct from domesticated capsids such as dArc1. Our results suggest a fully functional transposon mediates synaptic plasticity, possibly representing an early stage of domestication of a retrotransposon.
Collapse
Affiliation(s)
- P. Githure M’Angale
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Adrienne Lemieux
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Yumeng Liu
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Shuhao Wang
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Max Zinter
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Gimena Alegre
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Alfred Simkin
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Vivian Budnik
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Brian A. Kelch
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Travis Thomson
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
18
|
Yoneyama Y, Zhang RR, Maezawa M, Masaki H, Kimura M, Cai Y, Adam M, Parameswaran S, Mizuno N, Bhadury J, Maezawa S, Ochiai H, Nakauchi H, Potter SS, Weirauch MT, Takebe T. Intercellular mRNA transfer alters the human pluripotent stem cell state. Proc Natl Acad Sci U S A 2025; 122:e2413351122. [PMID: 39841146 PMCID: PMC11789055 DOI: 10.1073/pnas.2413351122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/07/2024] [Indexed: 01/23/2025] Open
Abstract
Intercellular transmission of messenger RNA (mRNA) is being explored in mammalian species using immortal cell lines. Here, we uncover an intercellular mRNA transfer phenomenon that allows for the adaptation and reprogramming of human primed pluripotent stem cells (hPSCs). This process is induced by the direct cell contact-mediated coculture with mouse embryonic stem cells under the condition impermissible for primed hPSC culture. Mouse-derived mRNA contents are transmitted into adapted hPSCs only in the coculture. Transfer-specific mRNA analysis shows the enrichment for divergent biological pathways involving transcription/translational machinery and stress-coping mechanisms, wherein such transfer is diminished when direct cell contacts are lost. After 5 d of coculture with mouse embryonic stem cells, surface marker analysis and global gene profiling confirmed that mRNA transfer-prone hPSC efficiently gains a naïve-like state. Furthermore, transfer-specific knockdown experiments targeting mouse-specific transcription factor-coding mRNAs in hPSC show that mouse-derived Tfcp2l1, Tfap2c, and Klf4 are indispensable for human naïve-like conversion. Thus, interspecies mRNA transfer triggers cellular reprogramming in mammalian cells. Our results support that episodic mRNA transfer can occur in cell cooperative and competitive processes, which provides a fresh perspective on understanding the roles of mRNA mobility for intra- and interspecies cellular communications.
Collapse
Affiliation(s)
- Yosuke Yoneyama
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Ran-Ran Zhang
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Mari Maezawa
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Hideki Masaki
- Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Masaki Kimura
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Yuqi Cai
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Mike Adam
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Naoaki Mizuno
- Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Joydeep Bhadury
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - So Maezawa
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba278-8510, Japan
| | - Hiroshi Ochiai
- Division of Gene Expression Dynamics, Medical Institute of Bioregulation, Kyushu University, Fukuoka812-0054, Japan
| | - Hiromitsu Nakauchi
- Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - S. Steven Potter
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Matthew T. Weirauch
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229-3039
| | - Takanori Takebe
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229-3039
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- World Premier International Research Center Initiative Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka565-0871, Japan
| |
Collapse
|
19
|
Tregub PP, Komleva YK, Kukla MV, Averchuk AS, Vetchinova AS, Rozanova NA, Illarioshkin SN, Salmina AB. Brain Plasticity and Cell Competition: Immediate Early Genes Are the Focus. Cells 2025; 14:143. [PMID: 39851571 PMCID: PMC11763428 DOI: 10.3390/cells14020143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Brain plasticity is at the basis of many cognitive functions, including learning and memory. It includes several mechanisms of synaptic and extrasynaptic changes, neurogenesis, and the formation and elimination of synapses. The plasticity of synaptic transmission involves the expression of immediate early genes (IEGs) that regulate neuronal activity, thereby supporting learning and memory. In addition, IEGs are involved in the regulation of brain cells' metabolism, proliferation, and survival, in the establishment of multicellular ensembles, and, presumably, in cell competition in the tissue. In this review, we analyze the current understanding of the role of IEGs (c-Fos, c-Myc, Arg3.1/Arc) in controlling brain plasticity in physiological and pathological conditions, including brain aging and neurodegeneration. This work might inspire new gene therapy strategies targeting IEGs to regulate synaptic plasticity, and potentially prevent or mitigate neurodegenerative diseases.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Research Center of Neurology, 125367 Moscow, Russia
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | | | | | - Anna S. Vetchinova
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | | | | | | |
Collapse
|
20
|
Nikolopoulos N, Oda SI, Prigozhin DM, Modis Y. Structure and Methyl-lysine Binding Selectivity of the HUSH Complex Subunit MPP8. J Mol Biol 2025; 437:168890. [PMID: 39638237 DOI: 10.1016/j.jmb.2024.168890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 11/19/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
The Human Silencing Hub (HUSH) guards the genome from the pathogenic effects of retroelement expression. Composed of MPP8, TASOR, and Periphilin-1, HUSH recognizes actively transcribed retrotransposed sequences by the presence of long (>1.5-kb) nascent transcripts without introns. HUSH recruits effectors that alter chromatin structure, degrade transcripts, and deposit transcriptionally repressive epigenetic marks. Here, we report the crystal structure of the C-terminal domain (CTD) of MPP8 necessary for HUSH activity. The MPP8 CTD consists of five ankyrin repeats followed by a domain with structural homology to the PINIT domains of Siz/PIAS-family SUMO E3 ligases. AlphaFold3 modeling of the MPP8-TASOR complex predicts that a SPOC domain and a domain with a novel fold in TASOR form extended interaction interfaces with the MPP8 CTD. Point mutations at these interfaces resulted in loss of HUSH-dependent transcriptional repression in a cell-based reporter assay, validating the AlphaFold3 model. The MPP8 chromodomain, known to bind the repressive mark H3K9me3, bound with similar or higher affinity to sequences in the H3K9 methyltransferase subunits SETDB1, ATF7IP, G9a, and GLP. Hence, MPP8 promotes heterochromatinization by recruiting H3K9 methyltransferases. Our work identifies novel structural elements in MPP8 required for HUSH complex assembly and silencing, thereby fulfilling vital functions in controlling retrotransposons.
Collapse
Affiliation(s)
- Nikos Nikolopoulos
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Shun-Ichiro Oda
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK
| | - Daniil M Prigozhin
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Yorgo Modis
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge CB2 0AW, UK.
| |
Collapse
|
21
|
Mottarlini F, Caffino L, Fumagalli F, Calabrese F, Brivio P. NeuropsychopharmARCology: Shaping Neuroplasticity through Arc/ Arg3.1 Modulation. Curr Neuropharmacol 2025; 23:650-670. [PMID: 39473108 DOI: 10.2174/011570159x338335240903075655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 11/05/2024] Open
Abstract
Activity-regulated cytoskeleton-associated protein (aka activity-regulated gene Arg3.1) belongs to the effector gene family of the immediate early genes. This family encodes effector proteins, which act directly on cellular homeostasis and function. Arc/Arg3.1 is localized at dendritic processes, allowing the protein local synthesis on demand, and it is considered a reliable index of activity- dependent synaptic changes. Evidence also exists showing the critical role of Arc/Arg3.1 in memory processes. The high sensitivity to changes in neuronal activity, its specific localization as well as its involvement in long-term synaptic plasticity indeed make this effector gene a potential, critical target of the action of psychotropic drugs. In this review, we focus on antipsychotic and antidepressant drugs as well as on psychostimulants, which belong to the category of drugs of abuse but can also be used as drugs for specific disorders of the central nervous system (i.e., Attention Deficit Hyperactivity Disorder). It is demonstrated that psychotropic drugs with different mechanisms of action converge on Arc/Arg3.1, providing a means whereby Arc/Arg3.1 synaptic modulation may contribute to their therapeutic activity. The potential translational implications for different neuropsychiatric conditions are also discussed, recognizing that the treatment of these disorders is indeed complex and involves the simultaneous regulation of several dysfunctional mechanisms.
Collapse
Affiliation(s)
- Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| |
Collapse
|
22
|
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
|
23
|
Blumenstiel JP. From the cauldron of conflict: Endogenous gene regulation by piRNA and other modes of adaptation enabled by selfish transposable elements. Semin Cell Dev Biol 2025; 164:1-12. [PMID: 38823219 DOI: 10.1016/j.semcdb.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Transposable elements (TEs) provide a prime example of genetic conflict because they can proliferate in genomes and populations even if they harm the host. However, numerous studies have shown that TEs, though typically harmful, can also provide fuel for adaptation. This is because they code functional sequences that can be useful for the host in which they reside. In this review, I summarize the "how" and "why" of adaptation enabled by the genetic conflict between TEs and hosts. In addition, focusing on mechanisms of TE control by small piwi-interacting RNAs (piRNAs), I highlight an indirect form of adaptation enabled by conflict. In this case, mechanisms of host defense that regulate TEs have been redeployed for endogenous gene regulation. I propose that the genetic conflict released by meiosis in early eukaryotes may have been important because, among other reasons, it spurred evolutionary innovation on multiple interwoven trajectories - on the part of hosts and also embedded genetic parasites. This form of evolution may function as a complexity generating engine that was a critical player in eukaryotic evolution.
Collapse
Affiliation(s)
- Justin P Blumenstiel
- Department of Ecology and Evolutionary Biology, University of Kansas, 1200 Sunnyside Avenue, Lawrence, KS 66045, United States.
| |
Collapse
|
24
|
Campodonico W, Mohan HM, Huynh PT, Black HH, Lau CI, Paulson HL, Sharkey LM, Whiteley AM. The gag-like gene RTL8 antagonizes PEG10-mediated virus like particles. PLoS One 2024; 19:e0310946. [PMID: 39775359 PMCID: PMC11684626 DOI: 10.1371/journal.pone.0310946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/09/2024] [Indexed: 01/30/2025] Open
Abstract
PEG10 is a retroelement-derived Mart-family gene that is necessary for placentation and has been implicated in neurological disease. PEG10 resembles both retrotransposon and retroviral proteins and forms virus-like particles (VLPs) that can be purified using iodixanol ultracentrifugation. It is hypothesized that formation of VLPs is crucial to the biological roles of PEG10 in reproduction and neurological health. Here, we describe the regulation of PEG10 VLP formation and release in human cells with a role for the related Mart gene RTL8. RTL8 resembles a truncated form of PEG10 that shares homology with the N-terminal gag-like capsid domain. Alone, RTL8 is unable to form VLPs, but was incorporated into PEG10-derived particles. RTL8 co-expression decreased the abundance of PEG10 VLPs and increased intracellular levels of PEG10, suggesting a model where RTL8 inhibits PEG10 VLP formation or release. Consistent with this model, RTL8 bound to the N-terminal domain of PEG10 capsid, and modulation of RTL8 influenced PEG10-derived VLP abundance in naturally producing cells. RTL8 is broadly expressed in many of the same tissues as PEG10, including in human brain. Taken together, these results describe a novel antagonistic relationship between two human retroelement-derived genes and have implications for our understanding of PEG10 biology and disease.
Collapse
Affiliation(s)
- Will Campodonico
- Department of Biochemistry, University of Colorado, Boulder, CO, United States of America
| | - Harihar M. Mohan
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States of America
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Phuoc T. Huynh
- Department of Biochemistry, University of Colorado, Boulder, CO, United States of America
- Molecular, Cellular and Developmental Biology Program, University of Colorado, Boulder, CO, United States of America
| | - Holly H. Black
- Department of Biochemistry, University of Colorado, Boulder, CO, United States of America
| | - Cristina I. Lau
- Department of Biochemistry, University of Colorado, Boulder, CO, United States of America
| | - Henry L. Paulson
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Lisa M. Sharkey
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Alexandra M. Whiteley
- Department of Biochemistry, University of Colorado, Boulder, CO, United States of America
| |
Collapse
|
25
|
Orozco-Arias S, Sierra P, Durbin R, González J. MCHelper automatically curates transposable element libraries across eukaryotic species. Genome Res 2024; 34:2256-2268. [PMID: 39653419 PMCID: PMC11694758 DOI: 10.1101/gr.278821.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 09/18/2024] [Indexed: 12/25/2024]
Abstract
The number of species with high-quality genome sequences continues to increase, in part due to the scaling up of multiple large-scale biodiversity sequencing projects. While the need to annotate genic sequences in these genomes is widely acknowledged, the parallel need to annotate transposable element (TE) sequences that have been shown to alter genome architecture, rewire gene regulatory networks, and contribute to the evolution of host traits is becoming ever more evident. However, accurate genome-wide annotation of TE sequences is still technically challenging. Several de novo TE identification tools are now available, but manual curation of the libraries produced by these tools is needed to generate high-quality genome annotations. Manual curation is time-consuming, and thus impractical for large-scale genomic studies, and lacks reproducibility. In this work, we present the Manual Curator Helper tool MCHelper, which automates the TE library curation process. By leveraging MCHelper's fully automated mode with the outputs from three de novo TE identification tools, RepeatModeler2, EDTA, and REPET, in the fruit fly, rice, hooded crow, zebrafish, maize, and human, we show a substantial improvement in the quality of the TE libraries and genome annotations. MCHelper libraries are less redundant, with up to 65% reduction in the number of consensus sequences, have up to 11.4% fewer false positive sequences, and up to ∼48% fewer "unclassified/unknown" TE consensus sequences. Genome-wide TE annotations are also improved, including larger unfragmented insertions. Moreover, MCHelper is an easy-to-install and easy-to-use tool.
Collapse
Affiliation(s)
| | - Pío Sierra
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Richard Durbin
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, United Kingdom
| | - Josefa González
- Institute of Evolutionary Biology, CSIC, UPF, 08003 Barcelona, Spain;
- Institut Botànic de Barcelona (IBB), CSIC-CMCNB, 08038 Barcelona, Spain
| |
Collapse
|
26
|
Kobayashi H, Yasukochi M, Horie M, Orba Y, Sawa H, Fujino K, Taharaguchi S. Neuron-associated retroelement-derived protein Arc/Arg3.1 assists in the early stages of alphaherpesvirus infection in human neuronal cells. PLoS One 2024; 19:e0314980. [PMID: 39666775 PMCID: PMC11637343 DOI: 10.1371/journal.pone.0314980] [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: 05/30/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024] Open
Abstract
Alphaherpesviruses, including herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV), are neurotropic double-stranded DNA viruses. Alphaherpesviruses control the expression of various host factors to ensure efficient infection and propagation. Recently, HSV-1 was found to upregulate Arc/Arg3.1 (Arc) expression, which is a retroelement-derived domesticated gene. Arc is associated with learning and neuroplasticity in host neuronal cells, and its abnormal expression leads to neurological disorders. However, the detailed mechanisms underlying the upregulation of Arc and its physiological significance in viral infections remain unclear. In this study, we found that PRV infection upregulated Arc expression in vitro and identified ICP0 and EP0, the transcriptional regulatory genes of HSV-1 and PRV, as triggers for enhanced Arc expression. Mass spectrometry and co-immunoprecipitation assays identified VP5, the major capsid protein of PRV and HSV-1, as the viral factor that interacted with Arc. Arc knockdown delayed viral infection during the early stages of the viral life cycle, but did not impact the viral attachment and entry. In conclusion, we provide evidence that alphaherpesvirus ICP0 homologues control Arc expression. Additionally, we demonstrate that Arc interacts with the major capsid protein VP5 and plays an important role in the viral lifecycle after intracellular entry. This study advances our knowledge of herpesvirus and retroelement-derived Arc interactions, providing fundamental insights into the pathogenesis of retroelement-derived domesticated genes and herpesvirus-induced neurological diseases.
Collapse
Affiliation(s)
- Hiroko Kobayashi
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mitsuki Yasukochi
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Masayuki Horie
- Laboratory of Veterinary Microbiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka, Japan
- Osaka International Research Center for Infectious Diseases, Osaka Metropolitan University, Osaka, Japan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kan Fujino
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| | - Satoshi Taharaguchi
- Laboratory of Microbiology, School of Veterinary Medicine, Azabu University, Sagamihara, Kanagawa, Japan
| |
Collapse
|
27
|
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
|
28
|
Tyagi M, Chadha R, de Hoog E, Sullivan KR, Walker AC, Northrop A, Fabian B, Fuxreiter M, Hyman BT, Shepherd JD. Arc mediates intercellular tau transmission via extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619703. [PMID: 39484489 PMCID: PMC11526995 DOI: 10.1101/2024.10.22.619703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Intracellular neurofibrillary tangles that consist of misfolded tau protein1 cause neurodegeneration in Alzheimer's disease (AD) and frontotemporal dementia (FTD). Tau pathology spreads cell-to-cell2 but the exact mechanisms of tau release and intercellular transmission remain poorly defined. Tau is released from neurons as free protein or in extracellular vesicles (EVs)3-5 but the role of these different release mechanisms in intercellular tau transmission is unclear. Here, we show that the neuronal gene Arc is critical for packaging tau into EVs. Brain EVs purified from human tau (hTau) transgenic rTg4510 mice (rTgWT) contain high levels of hTau that are capable of seeding tau pathology. In contrast, EVs purified from rTgWT crossed with Arc knock-out mice (rTgArc KO) have significantly less hTau and cannot seed tau aggregation. Arc facilitates the release of hTau in EVs produced via the I-BAR protein IRSp53, but not free tau. Arc protein directly binds hTau to form a fuzzy complex that we identified in both mouse and human brain tissue. We find that pathological intracellular hTau accumulates in neurons in rTgArc KO mice, which correlates with accelerated neuron loss in the hippocampus. Finally, we find that intercellular tau transmission is significantly abrogated in Arc KO mice. We conclude that Arc-dependent release of tau in EVs plays a significant role in intracellular tau elimination and intercellular tau transmission.
Collapse
Affiliation(s)
- Mitali Tyagi
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Radhika Chadha
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Eric de Hoog
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | | | - Alicia C. Walker
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Ava Northrop
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Balazs Fabian
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Germany
| | - Monika Fuxreiter
- Department of Biomedical Sciences University of Padova, Padova, Italy
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | |
Collapse
|
29
|
D'Egidio F, Castelli V, d'Angelo M, Ammannito F, Quintiliani M, Cimini A. Brain incoming call from glia during neuroinflammation: Roles of extracellular vesicles. Neurobiol Dis 2024; 201:106663. [PMID: 39251030 DOI: 10.1016/j.nbd.2024.106663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
The functionality of the central nervous system (CNS) relies on the connection, integration, and the exchange of information among neural cells. The crosstalk among glial cells and neurons is pivotal for a series of neural functions, such as development of the nervous system, electric conduction, synaptic transmission, neural circuit establishment, and brain homeostasis. Glial cells are crucial players in the maintenance of brain functionality in physiological and disease conditions. Neuroinflammation is a common pathological process in various brain disorders, such as neurodegenerative diseases, and infections. Glial cells, including astrocytes, microglia, and oligodendrocytes, are the main mediators of neuroinflammation, as they can sense and respond to brain insults by releasing pro-inflammatory or anti-inflammatory factors. Recent evidence indicates that extracellular vesicles (EVs) are pivotal players in the intercellular communication that underlies physiological and pathological processes. In particular, glia-derived EVs play relevant roles in modulating neuroinflammation, either by promoting or inhibiting the activation of glial cells and neurons, or by facilitating the clearance or propagation of pathogenic proteins. The involvement of EVs in neurodegenerative diseases such as Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD), and Multiple Sclerosis (MS)- which share hallmarks such as neuroinflammation and oxidative stress to DNA damage, alterations in neurotrophin levels, mitochondrial impairment, and altered protein dynamics- will be dissected, showing how EVs act as pivotal cell-cell mediators of toxic stimuli, thereby propagating degeneration and cell death signaling. Thus, this review focuses on the EVs secreted by microglia, astrocytes, oligodendrocytes and in neuroinflammatory conditions, emphasizing on their effects on neurons and on central nervous system functions, considering both their beneficial and detrimental effects.
Collapse
Affiliation(s)
- Francesco D'Egidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo".
| | - Fabrizio Ammannito
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Massimiliano Quintiliani
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| |
Collapse
|
30
|
Dowaidar M. Drug delivery based exosomes uptake pathways. Neurochem Int 2024; 179:105835. [PMID: 39147203 DOI: 10.1016/j.neuint.2024.105835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Most cells secrete a material called extracellular vesicles (EVs), which play a crucial role in cellular communication. Exosomes are one of the most studied types of EVs. Recent research has shown the many functions and substrates of cellular exosomes. Multiple studies have shown the efficacy of exosomes in transporting a wide variety of cargo to their respective target cells. As a result, they are often utilized to transport medicaments to patients. Natural exosomes as well as exosomes modified with other compounds to enhance transport capabilities have been employed. In this article, we take a look at how different types of exosomes and modified exosomes may transport different types of cargo to their respective targets. Exosomes have a lot of potential as drug delivery vehicles for many synthetic compounds, proteins, nucleic acids, and gene repair specialists because they can stay in the body for a long time, are biocompatible, and can carry natural materials. A good way to put specific protein particles into exosomes is still not clear, though, and the exosomes can't be used in many situations yet. The determinants for exosome production, as well as ways for loading certain therapeutic molecules (proteins, nucleic acids, and small compounds), were covered in this paper. Further study and the development of therapeutic exosomes may both benefit from the information collected in this review.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia; Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia.
| |
Collapse
|
31
|
Herbert A. Neurodegenerative diseases reflect the reciprocal roles played by retroelements in regulating memory and immunity. Front Neurosci 2024; 18:1445540. [PMID: 39371608 PMCID: PMC11451048 DOI: 10.3389/fnins.2024.1445540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Tetrapod endogenous retroelements (ERE) encode proteins that have been exapted to perform many roles in development and also in innate immunity, including GAG (group specific antigen) proteins from the ERE long terminal repeat (LTR) family, some of which can assemble into viral-like capsids (VLCs) and transmit mRNA across synapses. The best characterized member of this family is ARC (activity-regulated cytoskeletal gene), that is involved in memory formation. Other types of EREs, such as LINES and SINES (long and short interspersed elements), have instead been exapted for immune defenses against infectious agents. These immune EREs identify host transcripts by forming the unusual left-handed Z-DNA and Z-RNA conformations to enable self/nonself discrimination. Elevated levels of immune EREs in the brain are associated with neurodegenerative disease. Here I address the question of how pathways based on immune EREs are relate to the memory EREs that mediate neural plasticity. I propose that during infection and in other inflammatory states, ERE encoded GAG capsids deliver interferon-induced immune EREs that rapidly inhibit translation of viral RNAs in the dendritic splines by activation of protein kinase R (PKR). The response limits transmission of viruses and autonomously replicating elements, while protecting bystander cells from stress-induced cell death. Further, the PKR-dependent phosphorylation of proteins, like tau, disrupts the endocytic pathways exploited by viruses to spread to other cells. The responses come at a cost. They impair memory formation and can contribute to pathology by increasing the deposition of amyloid beta.
Collapse
|
32
|
Tang R, Guo L, Wei T, Chen T, Yang H, Ye H, Lin F, Zeng Y, Yu H, Cai Z, Liu X. Engineering PEG10assembled endogenous virus-like particles with genetically encoded neoantigen peptides for cancer vaccination. eLife 2024; 13:RP98579. [PMID: 39269893 PMCID: PMC11398863 DOI: 10.7554/elife.98579] [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] [Indexed: 09/15/2024] Open
Abstract
Tumor neoantigen peptide vaccines hold potential for boosting cancer immunotherapy, yet efficiently co-delivering peptides and adjuvants to antigen-presenting cells in vivo remains challenging. Virus-like particle (VLP), which is a kind of multiprotein structure organized as virus, can deliver therapeutic substances into cells and stimulate immune response. However, the weak targeted delivery of VLP in vivo and its susceptibility to neutralization by antibodies hinder their clinical applications. Here, we first designed a novel protein carrier using the mammalian-derived capsid protein PEG10, which can self-assemble into endogenous VLP (eVLP) with high protein loading and transfection efficiency. Then, an engineered tumor vaccine, named ePAC, was developed by packaging genetically encoded neoantigen into eVLP with further modification of CpG-ODN on its surface to serve as an adjuvant and targeting unit to dendritic cells (DCs). Significantly, ePAC can efficiently target and transport neoantigens to DCs, and promote DCs maturation to induce neoantigen-specific T cells. Moreover, in mouse orthotopic liver cancer and humanized mouse tumor models, ePAC combined with anti-TIM-3 exhibited remarkable antitumor efficacy. Overall, these results support that ePAC could be safely utilized as cancer vaccines for antitumor therapy, showing significant potential for clinical translation.
Collapse
Affiliation(s)
- Ruijing Tang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Luobin Guo
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Tingyu Wei
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Tingting Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Huan Yang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Honghao Ye
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Fangzhou Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical UniversityFuzhouChina
- The Liver Center of Fujian Province, Fujian Medical UniversityShanghaiChina
- Mengchao Med-X Center, Fuzhou UniversityFuzhouChina
| |
Collapse
|
33
|
Lallai V, Lam TT, Garcia-Milian R, Chen YC, Fowler JP, Manca L, Piomelli D, Williams K, Nairn AC, Fowler CD. Proteomic Profile of Circulating Extracellular Vesicles in the Brain after Δ9-Tetrahydrocannabinol Inhalation. Biomolecules 2024; 14:1143. [PMID: 39334909 PMCID: PMC11430348 DOI: 10.3390/biom14091143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Given the increasing use of cannabis in the US, there is an urgent need to better understand the drug's effects on central signaling mechanisms. Extracellular vesicles (EVs) have been identified as intercellular signaling mediators that contain a variety of cargo, including proteins. Here, we examined whether the main psychoactive component in cannabis, Δ9-tetrahydrocannabinol (THC), alters EV protein signaling dynamics in the brain. We first conducted in vitro studies, which found that THC activates signaling in choroid plexus epithelial cells, resulting in transcriptional upregulation of the cannabinoid 1 receptor and immediate early gene c-fos, in addition to the release of EVs containing RNA cargo. Next, male and female rats were examined for the effects of either acute or chronic exposure to aerosolized ('vaped') THC on circulating brain EVs. Cerebrospinal fluid was extracted from the brain, and EVs were isolated and processed with label-free quantitative proteomic analyses via high-resolution tandem mass spectrometry. Interestingly, circulating EV-localized proteins were differentially expressed based on acute or chronic THC exposure in a sex-specific manner. Taken together, these findings reveal that THC acts in the brain to modulate circulating EV signaling, thereby providing a novel understanding of how exogenous factors can regulate intercellular communication in the brain.
Collapse
Affiliation(s)
- Valeria Lallai
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
| | - TuKiet T. Lam
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT 06511, USA
| | - Rolando Garcia-Milian
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Bioinformatics Support Hub, Harvey Cushing/John Whitney Medical Library, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yen-Chu Chen
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - James P. Fowler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - Letizia Manca
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
| | - Daniele Piomelli
- Department and Anatomy and Neurobiology, University of California, Irvine, CA 92697, USA;
| | - Kenneth Williams
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Angus C. Nairn
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA
| | - Christie D. Fowler
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA; (V.L.); (Y.-C.C.); (J.P.F.)
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT 06511, USA; (T.T.L.); (R.G.-M.); (K.W.); (A.C.N.)
| |
Collapse
|
34
|
Andrzejewska-Romanowska A, Gumna J, Tykwińska E, Pachulska-Wieczorek K. Mapping the structural landscape of the yeast Ty3 retrotransposon RNA genome. Nucleic Acids Res 2024; 52:9821-9837. [PMID: 38864374 PMCID: PMC11381356 DOI: 10.1093/nar/gkae494] [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: 02/09/2024] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024] Open
Abstract
Long terminal repeat (LTR)-retrotransposons are significant contributors to the evolution and diversity of eukaryotic genomes. Their RNA genomes (gRNA) serve as a template for protein synthesis and reverse transcription to a DNA copy, which can integrate into the host genome. Here, we used the SHAPE-MaP strategy to explore Ty3 retrotransposon gRNA structure in yeast and under cell-free conditions. Our study reveals the structural dynamics of Ty3 gRNA and the well-folded core, formed independently of the cellular environment. Based on the detailed map of Ty3 gRNA structure, we characterized the structural context of cis-acting sequences involved in reverse transcription and frameshifting. We also identified a novel functional sequence as a potential initiator for Ty3 gRNA dimerization. Our data indicate that the dimer is maintained by direct interaction between short palindromic sequences at the 5' ends of the two Ty3 gRNAs, resembling the model characteristic for other retroelements like HIV-1 and Ty1. This work points out a range of cell-dependent and -independent Ty3 gRNA structural changes that provide a solid background for studies on RNA structure-function relationships important for retroelement biology.
Collapse
Affiliation(s)
- Angelika Andrzejewska-Romanowska
- Department of RNA Structure and Function, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Julita Gumna
- Department of RNA Structure and Function, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Ewa Tykwińska
- Department of RNA Structure and Function, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Katarzyna Pachulska-Wieczorek
- Department of RNA Structure and Function, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
35
|
Wang Y, Xu H, He Q, Wu Z, Han GZ. Natural Transposable Element Insertions Contribute to Host Fitness in Model Yeasts. Genome Biol Evol 2024; 16:evae193. [PMID: 39228319 PMCID: PMC11403283 DOI: 10.1093/gbe/evae193] [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] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024] Open
Abstract
Transposable elements (TEs) are ubiquitous in the eukaryote genomes, but their evolutionary and functional significance remains largely obscure and contentious. Here, we explore the evolution and functional impact of TEs in two model unicellular eukaryotes, the fission yeast Schizosaccharomyces pombe and the budding yeast Saccharomyces cerevisiae, which diverged around 330 to 420 million years ago. We analyze the distribution of LTR retrotransposons (LTR-RTs, the only TE order identified in both species) and their solo-LTR derivatives in 35 strains of S. pombe and 128 strains of S. cerevisiae. We find that natural LTR-RT and solo-LTR insertions exhibit high presence-absence polymorphism among individuals in both species. Population genetics analyses show that solo-LTR insertions experienced functional constraints similar to synonymous sites of host genes in both species, indicating a majority of solo-LTR insertions might have evolved in a neutral manner. When knocking out nine representative solo-LTR insertions separately in the S. pombe strain 972h- and 12 representative solo-LTR insertions separately in the S. cerevisiae strain S288C, we find that one solo-LTR insertion in S. pombe has a significant effect on the fitness and transcriptome of its host. Together, our findings indicate that a fraction of natural TE insertions likely shape their host transcriptomes and thereby contribute to their host fitness, with implications for understanding the functional significance of TEs in eukaryotes.
Collapse
Affiliation(s)
- Yan Wang
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Hao Xu
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Qinliu He
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Zhiwei Wu
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Guan-Zhu Han
- College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| |
Collapse
|
36
|
Mustafin RN. A hypothesis about interrelations of epigenetic factors and transposable elements in memory formation. Vavilovskii Zhurnal Genet Selektsii 2024; 28:476-486. [PMID: 39280851 PMCID: PMC11393658 DOI: 10.18699/vjgb-24-54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 09/18/2024] Open
Abstract
The review describes the hypothesis that the drivers of epigenetic regulation in memory formation are transposable elements that influence the expression of specific genes in the brain. The hypothesis is confirmed by research into transposon activation in neuronal stem cells during neuronal differentiation. These changes occur in the hippocampus dentate gyrus, where a pronounced activity of transposons and their insertion near neuron-specific genes have been detected. In experiments on changing the activity of histone acetyltransferase and inhibition of DNA methyltransferase and reverse transcriptase, the involvement of epigenetic factors and retroelements in the mechanisms of memory formation has been shown. Also, a number of studies on different animals have revealed the preservation of long-term memory without the participation of synaptic plasticity. The data obtained suggest that transposons, which are genome sensors highly sensitive to various environmental and internal influences, form memory at the nuclear coding level. Therefore, long-term memory is preserved after elimination of synaptic connections. This is confirmed by the fact that the proteins involved in memory formation, including the transfer of genetic information through synapses between neurons (Arc protein), originate from transposons. Long non-coding RNAs and microRNAs also originate from transposons; their role in memory consolidation has been described. Pathological activation of transposable elements is a likely cause of neurodegenerative diseases with memory impairment. Analysis of the scientific literature allowed us to identify changes in the expression of 40 microRNAs derived from transposons in Alzheimer's disease. For 24 of these microRNAs, the mechanisms of regulation of genes involved in the functioning of the brain have been described. It has been suggested that the microRNAs we identified could become potential tools for regulating transposon activity in the brain in order to improve memory.
Collapse
|
37
|
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
|
38
|
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
|
39
|
Gable SM, Bushroe NA, Mendez JM, Wilson A, Pinto BJ, Gamble T, Tollis M. Differential Conservation and Loss of Chicken Repeat 1 (CR1) Retrotransposons in Squamates Reveal Lineage-Specific Genome Dynamics Across Reptiles. Genome Biol Evol 2024; 16:evae157. [PMID: 39031594 PMCID: PMC11303007 DOI: 10.1093/gbe/evae157] [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: 02/14/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/22/2024] Open
Abstract
Transposable elements (TEs) are repetitive DNA sequences which create mutations and generate genetic diversity across the tree of life. In amniote vertebrates, TEs have been mainly studied in mammals and birds, whose genomes generally display low TE diversity. Squamates (Order Squamata; including ∼11,000 extant species of lizards and snakes) show as much variation in TE abundance and activity as they do in species and phenotypes. Despite this high TE activity, squamate genomes are remarkably uniform in size. We hypothesize that novel, lineage-specific genome dynamics have evolved over the course of squamate evolution. To understand the interplay between TEs and host genomes, we analyzed the evolutionary history of the chicken repeat 1 (CR1) retrotransposon, a TE family found in most tetrapod genomes which is the dominant TE in most reptiles. We compared 113 squamate genomes to the genomes of turtles, crocodilians, and birds and used ancestral state reconstruction to identify shifts in the rate of CR1 copy number evolution across reptiles. We analyzed the repeat landscapes of CR1 in squamate genomes and determined that shifts in the rate of CR1 copy number evolution are associated with lineage-specific variation in CR1 activity. We then used phylogenetic reconstruction of CR1 subfamilies across amniotes to reveal both recent and ancient CR1 subclades across the squamate tree of life. The patterns of CR1 evolution in squamates contrast other amniotes, suggesting key differences in how TEs interact with different host genomes and at different points across evolutionary history.
Collapse
Affiliation(s)
- Simone M Gable
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Nicholas A Bushroe
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Jasmine M Mendez
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Adam Wilson
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Brendan J Pinto
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- Department of Zoology, Milwaukee Public Museum, Milwaukee, WI, USA
| | - Tony Gamble
- Department of Zoology, Milwaukee Public Museum, Milwaukee, WI, USA
- Department of Biological Sciences, Marquette University, Milwaukee, WI, USA
- Bell Museum of Natural History, University of Minnesota, St. Paul, MN, USA
| | - Marc Tollis
- School of Informatics, Computing, and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| |
Collapse
|
40
|
Cagliani R, Forni D, Mozzi A, Fuchs R, Tussia-Cohen D, Arrigoni F, Pozzoli U, De Gioia L, Hagai T, Sironi M. Evolution of Virus-like Features and Intrinsically Disordered Regions in Retrotransposon-derived Mammalian Genes. Mol Biol Evol 2024; 41:msae154. [PMID: 39101471 PMCID: PMC11299033 DOI: 10.1093/molbev/msae154] [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: 05/29/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/06/2024] Open
Abstract
Several mammalian genes have originated from the domestication of retrotransposons, selfish mobile elements related to retroviruses. Some of the proteins encoded by these genes have maintained virus-like features; including self-processing, capsid structure formation, and the generation of different isoforms through -1 programmed ribosomal frameshifting. Using quantitative approaches in molecular evolution and biophysical analyses, we studied 28 retrotransposon-derived genes, with a focus on the evolution of virus-like features. By analyzing the rate of synonymous substitutions, we show that the -1 programmed ribosomal frameshifting mechanism in three of these genes (PEG10, PNMA3, and PNMA5) is conserved across mammals and originates alternative proteins. These genes were targets of positive selection in primates, and one of the positively selected sites affects a B-cell epitope on the spike domain of the PNMA5 capsid, a finding reminiscent of observations in infectious viruses. More generally, we found that retrotransposon-derived proteins vary in their intrinsically disordered region content and this is directly associated with their evolutionary rates. Most positively selected sites in these proteins are located in intrinsically disordered regions and some of them impact protein posttranslational modifications, such as autocleavage and phosphorylation. Detailed analyses of the biophysical properties of intrinsically disordered regions showed that positive selection preferentially targeted regions with lower conformational entropy. Furthermore, positive selection introduces variation in binary sequence patterns across orthologues, as well as in chain compaction. Our results shed light on the evolutionary trajectories of a unique class of mammalian genes and suggest a novel approach to study how intrinsically disordered region biophysical characteristics are affected by evolution.
Collapse
Affiliation(s)
- Rachele Cagliani
- Scientific Institute IRCCS E. MEDEA, Computational Biology Unit, Bosisio Parini 23842, Italy
| | - Diego Forni
- Scientific Institute IRCCS E. MEDEA, Computational Biology Unit, Bosisio Parini 23842, Italy
| | - Alessandra Mozzi
- Scientific Institute IRCCS E. MEDEA, Computational Biology Unit, Bosisio Parini 23842, Italy
| | - Rotem Fuchs
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dafna Tussia-Cohen
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Federica Arrigoni
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan 20126, Italy
| | - Uberto Pozzoli
- Scientific Institute IRCCS E. MEDEA, Computational Biology Unit, Bosisio Parini 23842, Italy
| | - Luca De Gioia
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan 20126, Italy
| | - Tzachi Hagai
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Manuela Sironi
- Scientific Institute IRCCS E. MEDEA, Computational Biology Unit, Bosisio Parini 23842, Italy
| |
Collapse
|
41
|
Roberts A, Swerdlow RH, Wang N. Adaptive and Maladaptive DNA Breaks in Neuronal Physiology and Alzheimer's Disease. Int J Mol Sci 2024; 25:7774. [PMID: 39063016 PMCID: PMC11277458 DOI: 10.3390/ijms25147774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/11/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
DNA strand breaks excessively accumulate in the brains of patients with Alzheimer's disease (AD). While traditionally considered random, deleterious events, neuron activity itself induces DNA breaks, and these "adaptive" breaks help mediate synaptic plasticity and memory formation. Recent studies mapping the brain DNA break landscape reveal that despite a net increase in DNA breaks in ectopic genomic hotspots, adaptive DNA breaks around synaptic genes are lost in AD brains, and this is associated with transcriptomic dysregulation. Additionally, relationships exist between mitochondrial dysfunction, a hallmark of AD, and DNA damage, such that mitochondrial dysfunction may perturb adaptive DNA break formation, while DNA breaks may conversely impair mitochondrial function. A failure of DNA break physiology could, therefore, potentially contribute to AD pathogenesis.
Collapse
Affiliation(s)
- Anysja Roberts
- University of Kansas Alzheimer’s Disease Research Center, Kansas City, KS 66205, USA (R.H.S.)
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Research Center, Kansas City, KS 66205, USA (R.H.S.)
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City 66160, KS, USA
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Ning Wang
- University of Kansas Alzheimer’s Disease Research Center, Kansas City, KS 66205, USA (R.H.S.)
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
42
|
Wood TW, Henriques WS, Cullen HB, Romero M, Blengini CS, Sarathy S, Sorkin J, Bekele H, Jin C, Kim S, Chemiakine A, Khondker RC, Isola JV, Stout MB, Gennarino VA, Mogessie B, Jain D, Schindler K, Suh Y, Wiedenheft B, Berchowitz LE. The retrotransposon-derived capsid genes PNMA1 and PNMA4 maintain reproductive capacity. RESEARCH SQUARE 2024:rs.3.rs-4559920. [PMID: 39041030 PMCID: PMC11261967 DOI: 10.21203/rs.3.rs-4559920/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
The human genome contains 24 gag-like capsid genes derived from deactivated retrotransposons conserved among eutherians. Although some of their encoded proteins retain the ability to form capsids and even transfer cargo, their fitness benefit has remained elusive. Here we show that the gag-like genes PNMA1 and PNMA4 support reproductive capacity during aging. Analysis of donated human ovaries shows that expression of both genes declines normally with age, while several PNMA1 and PNMA4 variants identified in genome-wide association studies are causally associated with low testosterone, altered puberty onset, or obesity. Six-week-old mice lacking either Pnma1 or Pnma4 are indistinguishable from wild-type littermates, but by six months the mutant mice become prematurely subfertile, with precipitous drops in sex hormone levels, gonadal atrophy, and abdominal obesity; overall they produce markedly fewer offspring than controls. These findings expand our understanding of factors that maintain human reproductive health and lend insight into the domestication of retrotransposon-derived genes.
Collapse
Affiliation(s)
- Thomas W.P. Wood
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - William S. Henriques
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, 59717, USA
| | - Harrison B. Cullen
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mayra Romero
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Cecilia S. Blengini
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Shreya Sarathy
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Julia Sorkin
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Hilina Bekele
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, 06511, USA
| | - Chen Jin
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Seungsoo Kim
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alexei Chemiakine
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rishad C. Khondker
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - José V.V. Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Vincenzo A. Gennarino
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
- Columbia Stem Cell Initiative, New York, NY 10032, USA
- Initiative for Columbia Ataxia and Tremor, New York, NY 10032, USA
| | - Binyam Mogessie
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, 06511, USA
| | - Devanshi Jain
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Karen Schindler
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Yousin Suh
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Blake Wiedenheft
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, 59717, USA
| | - Luke E. Berchowitz
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s and the Aging Brain, New York, NY, USA
| |
Collapse
|
43
|
Zanirati G, dos Santos PG, Alcará AM, Bruzzo F, Ghilardi IM, Wietholter V, Xavier FAC, Gonçalves JIB, Marinowic D, Shetty AK, da Costa JC. Extracellular Vesicles: The Next Generation of Biomarkers and Treatment for Central Nervous System Diseases. Int J Mol Sci 2024; 25:7371. [PMID: 39000479 PMCID: PMC11242541 DOI: 10.3390/ijms25137371] [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: 04/24/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 07/16/2024] Open
Abstract
It has been widely established that the characterization of extracellular vesicles (EVs), particularly small EVs (sEVs), shed by different cell types into biofluids, helps to identify biomarkers and therapeutic targets in neurological and neurodegenerative diseases. Recent studies are also exploring the efficacy of mesenchymal stem cell-derived extracellular vesicles naturally enriched with therapeutic microRNAs and proteins for treating various diseases. In addition, EVs released by various neural cells play a crucial function in the modulation of signal transmission in the brain in physiological conditions. However, in pathological conditions, such EVs can facilitate the spread of pathological proteins from one brain region to the other. On the other hand, the analysis of EVs in biofluids can identify sensitive biomarkers for diagnosis, prognosis, and disease progression. This review discusses the potential therapeutic use of stem cell-derived EVs in several central nervous system diseases. It lists their differences and similarities and confers various studies exploring EVs as biomarkers. Further advances in EV research in the coming years will likely lead to the routine use of EVs in therapeutic settings.
Collapse
Affiliation(s)
- Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Paula Gabrielli dos Santos
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Allan Marinho Alcará
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Fernanda Bruzzo
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Isadora Machado Ghilardi
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Vinicius Wietholter
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Fernando Antônio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Daniel Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX 77807, USA;
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90610-000, RS, Brazil; (P.G.d.S.); (F.B.); (I.M.G.); (V.W.); (F.A.C.X.); (J.I.B.G.); (D.M.); (J.C.d.C.)
| |
Collapse
|
44
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
45
|
Luo R, Le H, Wu Q, Gong C. Nanoplatform-Based In Vivo Gene Delivery Systems for Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312153. [PMID: 38441386 DOI: 10.1002/smll.202312153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Indexed: 07/26/2024]
Abstract
Gene therapy uses modern molecular biology methods to repair disease-causing genes. As a burgeoning therapeutic, it has been widely applied for cancer therapy. Since 1989, there have been numerous clinical gene therapy cases worldwide. However, a few are successful. The main challenge of clinical gene therapy is the lack of efficient and safe vectors. Although viral vectors show high transfection efficiency, their application is still limited by immune rejection and packaging capacity. Therefore, the development of non-viral vectors is overwhelming. Nanoplatform-based non-viral vectors become a hotspot in gene therapy. The reasons are mainly as follows. 1) Non-viral vectors can be engineered to be uptaken by specific types of cells or tissues, providing effective targeting capability. 2) Non-viral vectors can protect goods that need to be delivered from degradation. 3) Nanoparticles can transport large-sized cargo such as CRISPR/Cas9 plasmids and nucleoprotein complexes. 4) Nanoparticles are highly biosafe, and they are not mutagenic in themselves compared to viral vectors. 5) Nanoparticles are easy to scale preparation, which is conducive to clinical conversion and application. Here, an overview of the categories of nanoplatform-based non-viral gene vectors, the limitations on their development, and their applications in cancer therapy.
Collapse
Affiliation(s)
- Rui Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Le
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qinjie Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Changyang Gong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
46
|
Manna I, De Benedittis S, Porro D. Extracellular Vesicles in Multiple Sclerosis: Their Significance in the Development and Possible Applications as Therapeutic Agents and Biomarkers. Genes (Basel) 2024; 15:772. [PMID: 38927708 PMCID: PMC11203165 DOI: 10.3390/genes15060772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Extracellular vesicles (EVs) are "micro-shuttles" that play a role as mediators of intercellular communication. Cells release EVs into the extracellular environment in both physiological and pathological conditions and are involved in intercellular communication, due to their ability to transfer proteins, lipids, and nucleic acids, and in the modulation of the immune system and neuroinflammation. Because EVs can penetrate the blood-brain barrier and move from the central nervous system to the peripheral circulation, and vice versa, recent studies have shown a substantial role for EVs in several neurological diseases, including multiple sclerosis (MS). MS is a demyelinating disease where the main event is caused by T and B cells triggering an autoimmune reaction against myelin constituents. Recent research has elucidate the potential involvement of extracellular vesicles (EVs) in the pathophysiology of MS, although, to date, their potential role both as agents and therapeutic targets in MS is not fully defined. We present in this review a summary and comprehensive examination of EVs' involvement in the pathophysiology of multiple sclerosis, exploring their potential applications as biomarkers and indicators of therapy response.
Collapse
Affiliation(s)
- Ida Manna
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Section of Catanzaro, 88100 Catanzaro, Italy
| | - Selene De Benedittis
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 87050 Cosenza, Italy
| | - Danilo Porro
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, 20054 Milan, Italy
| |
Collapse
|
47
|
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
|
48
|
Meador K, Castells-Graells R, Aguirre R, Sawaya MR, Arbing MA, Sherman T, Senarathne C, Yeates TO. A suite of designed protein cages using machine learning and protein fragment-based protocols. Structure 2024; 32:751-765.e11. [PMID: 38513658 PMCID: PMC11162342 DOI: 10.1016/j.str.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/22/2024] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Designed protein cages and related materials provide unique opportunities for applications in biotechnology and medicine, but their creation remains challenging. Here, we apply computational approaches to design a suite of tetrahedrally symmetric, self-assembling protein cages. For the generation of docked conformations, we emphasize a protein fragment-based approach, while for sequence design of the de novo interface, a comparison of knowledge-based and machine learning protocols highlights the power and increased experimental success achieved using ProteinMPNN. An analysis of design outcomes provides insights for improving interface design protocols, including prioritizing fragment-based motifs, balancing interface hydrophobicity and polarity, and identifying preferred polar contact patterns. In all, we report five structures for seven protein cages, along with two structures of intermediate assemblies, with the highest resolution reaching 2.0 Å using cryo-EM. This set of designed cages adds substantially to the body of available protein nanoparticles, and to methodologies for their creation.
Collapse
Affiliation(s)
- Kyle Meador
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | | | - Roman Aguirre
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Michael R Sawaya
- UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA 90095, USA
| | - Mark A Arbing
- UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA 90095, USA
| | - Trent Sherman
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Chethaka Senarathne
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Todd O Yeates
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA; UCLA-DOE Institute for Genomics and Proteomics, Los Angeles, CA 90095, USA.
| |
Collapse
|
49
|
Ma L, Wu Q, You Y, Zhang P, Tan D, Liang M, Huang Y, Gao Y, Ban Y, Chen Y, Yuan J. Neuronal small extracellular vesicles carrying miR-181c-5p contribute to the pathogenesis of epilepsy by regulating the protein kinase C-δ/glutamate transporter-1 axis in astrocytes. Glia 2024; 72:1082-1095. [PMID: 38385571 DOI: 10.1002/glia.24517] [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: 09/25/2023] [Revised: 01/17/2024] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
Information exchange between neurons and astrocytes mediated by extracellular vesicles (EVs) is known to play a key role in the pathogenesis of central nervous system diseases. A key driver of epilepsy is the dysregulation of intersynaptic excitatory neurotransmitters mediated by astrocytes. Thus, we investigated the potential association between neuronal EV microRNAs (miRNAs) and astrocyte glutamate uptake ability in epilepsy. Here, we showed that astrocytes were able to engulf epileptogenic neuronal EVs, inducing a significant increase in the glutamate concentration in the extracellular fluid of astrocytes, which was linked to a decrease in glutamate transporter-1 (GLT-1) protein expression. Using sequencing and gene ontology (GO) functional analysis, miR-181c-5p was found to be the most significantly upregulated miRNA in epileptogenic neuronal EVs and was linked to glutamate metabolism. Moreover, we found that neuronal EV-derived miR-181c-5p interacted with protein kinase C-delta (PKCδ), downregulated PKCδ and GLT-1 protein expression and increased glutamate concentrations in astrocytes both in vitro and in vivo. Our findings demonstrated that epileptogenic neuronal EVs carrying miR-181c-5p decrease the glutamate uptake ability of astrocytes, thus promoting susceptibility to epilepsy.
Collapse
Affiliation(s)
- Limin Ma
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Qingyuan Wu
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yu You
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Zhang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dandan Tan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Minxue Liang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunyi Huang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Gao
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuenan Ban
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yangmei Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinxian Yuan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
50
|
Lin X, Yin J, Wang Y, Yao J, Li QQ, Latzel V, Bossdorf O, Zhang YY. Environment-induced heritable variations are common in Arabidopsis thaliana. Nat Commun 2024; 15:4615. [PMID: 38816460 PMCID: PMC11139905 DOI: 10.1038/s41467-024-49024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Parental or ancestral environments can induce heritable phenotypic changes, but whether such environment-induced heritable changes are a common phenomenon remains unexplored. Here, we subject 14 genotypes of Arabidopsis thaliana to 10 different environmental treatments and observe phenotypic and genome-wide gene expression changes over four successive generations. We find that all treatments caused heritable phenotypic and gene expression changes, with a substantial proportion stably transmitted over all observed generations. Intriguingly, the susceptibility of a genotype to environmental inductions could be predicted based on the transposon abundance in the genome. Our study thus challenges the classic view that the environment only participates in the selection of heritable variation and suggests that the environment can play a significant role in generating of heritable variations.
Collapse
Affiliation(s)
- Xiaohe Lin
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, China
| | - Junjie Yin
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, China
| | - Yifan Wang
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, China
| | - Jing Yao
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, China
| | - Qingshun Q Li
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, China
- Biomedical Sciences, College of Dental Medicine, Western University of Health Sciences, Pomona, CA, USA
| | - Vit Latzel
- Institute of Botany of the CAS, Zamek 1, 252 43, Pruhonice, Czech Republic
| | - Oliver Bossdorf
- Institute of Evolution & Ecology, University of Tübingen, Auf der Morgenstelle 5, 72076, Tübingen, Germany
| | - Yuan-Ye Zhang
- Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, College of the Environment and Ecology, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|