1
|
Huggon L, Clayton EL. Beginning from the end: the presynaptic terminal as a pathomechanism hub in frontotemporal dementia and amyotrophic lateral sclerosis. Neural Regen Res 2025; 20:3217-3218. [PMID: 39715090 PMCID: PMC11881734 DOI: 10.4103/nrr.nrr-d-24-00639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/12/2024] [Accepted: 09/03/2024] [Indexed: 12/25/2024] Open
Affiliation(s)
- Laura Huggon
- UK Dementia Research Institute at King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, UK
| | - Emma L. Clayton
- UK Dementia Research Institute at King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology & Neuroscience, King’s College London, London, UK
| |
Collapse
|
2
|
Zeng Y, Antoniou A. Regulation of synaptic mitochondria by extracellular vesicles and its implications for neuronal metabolism and synaptic plasticity. J Cereb Blood Flow Metab 2025:271678X251337630. [PMID: 40367393 PMCID: PMC12078259 DOI: 10.1177/0271678x251337630] [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: 01/30/2025] [Revised: 03/17/2025] [Accepted: 03/28/2025] [Indexed: 05/16/2025]
Abstract
Mitochondrial metabolism in neurons is necessary for energetically costly processes like synaptic transmission and plasticity. As post-mitotic cells, neurons are therefore faced with the challenge of maintaining healthy functioning mitochondria throughout lifetime. The precise mechanisms of mitochondrial maintenance in neurons, and particularly in morphologically complex dendrites and axons, are not fully understood. Evidence from several biological systems suggests the regulation of cellular metabolism by extracellular vesicles (EVs), secretory lipid-enclosed vesicles that have emerged as important mediators of cell communication. In the nervous system, neuronal and glial EVs were shown to regulate neuronal circuit development and function, at least in part via the transfer of protein and RNA cargo. Interestingly, EVs have been implicated in diseases characterized by altered metabolism, such as cancer and neurodegenerative diseases. Furthermore, nervous system EVs were shown to contain proteins related to metabolic processes, mitochondrial proteins and even intact mitochondria. Here, we present the current knowledge of the mechanisms underlying neuronal mitochondrial maintenance, and highlight recent evidence suggesting the regulation of synaptic mitochondria by neuronal and glial cell EVs. We further discuss the potential implications of EV-mediated regulation of mitochondrial maintenance and function in neuronal circuit development and synaptic plasticity.
Collapse
Affiliation(s)
- Yuzhou Zeng
- Medical Faculty, University of Bonn, Bonn, Germany
| | - Anna Antoniou
- Medical Faculty, University of Bonn, Bonn, Germany
- Faculty of Life Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Zilio E, Schlegel T, Zaninello M, Rugarli EI. The role of mitochondrial mRNA translation in cellular communication. J Cell Sci 2025; 138:jcs263753. [PMID: 40326563 DOI: 10.1242/jcs.263753] [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: 05/07/2025] Open
Abstract
Mitochondria are dynamic and heterogeneous organelles that rewire their network and metabolic functions in response to changing cellular needs. To this end, mitochondria integrate a plethora of incoming signals to influence cell fate and survival. A crucial and highly regulated node of cell-mitochondria communication is the translation of nuclear-encoded mitochondrial mRNAs. By controlling and monitoring the spatio-temporal translation of these mRNAs, cells can rapidly adjust mitochondrial function to meet metabolic demands, optimise ATP production and regulate organelle biogenesis and turnover. In this Review, we focus on how RNA-binding proteins that recognise nuclear-encoded mitochondrial mRNAs acutely modulate the rate of translation in response to nutrient availability. We further discuss the relevance of localised translation of these mRNAs for subsets of mitochondria in polarised cells. Finally, we highlight quality control mechanisms that monitor the translation process at the mitochondrial surface and their connections to mitophagy and stress responses. We propose that these processes collectively contribute to mitochondrial specialisation and signalling function.
Collapse
Affiliation(s)
- Eleonora Zilio
- Institute for Genetics University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Tim Schlegel
- Institute for Genetics University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Marta Zaninello
- Institute for Genetics University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Elena I Rugarli
- Institute for Genetics University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| |
Collapse
|
4
|
Mohar B, Michel G, Wang YZ, Hernandez V, Grimm JB, Park JY, Patel R, Clarke M, Brown TA, Bergmann C, Gebis KK, Wilen AP, Liu B, Johnson R, Graves A, Tchumatchenko T, Savas JN, Fornasiero EF, Huganir RL, Tillberg PW, Lavis LD, Svoboda K, Spruston N. DELTA: a method for brain-wide measurement of synaptic protein turnover reveals localized plasticity during learning. Nat Neurosci 2025; 28:1089-1098. [PMID: 40164741 DOI: 10.1038/s41593-025-01923-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
Synaptic plasticity alters neuronal connections in response to experience, which is thought to underlie learning and memory. However, the loci of learning-related synaptic plasticity, and the degree to which plasticity is localized or distributed, remain largely unknown. Here we describe a new method, DELTA, for mapping brain-wide changes in synaptic protein turnover with single-synapse resolution, based on Janelia Fluor dyes and HaloTag knock-in mice. During associative learning, the turnover of the ionotropic glutamate receptor subunit GluA2, an indicator of synaptic plasticity, was enhanced in several brain regions, most markedly hippocampal area CA1. More broadly distributed increases in the turnover of synaptic proteins were observed in response to environmental enrichment. In CA1, GluA2 stability was regulated in an input-specific manner, with more turnover in layers containing input from CA3 compared to entorhinal cortex. DELTA will facilitate exploration of the molecular and circuit basis of learning and memory and other forms of plasticity at scales ranging from single synapses to the entire brain.
Collapse
Affiliation(s)
- Boaz Mohar
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA.
| | - Gabriela Michel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Veronica Hernandez
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jonathan B Grimm
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jin-Yong Park
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Ronak Patel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Morgan Clarke
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Timothy A Brown
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Cornelius Bergmann
- Institute for Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kamil K Gebis
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anika P Wilen
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bian Liu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard Johnson
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Austin Graves
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tatjana Tchumatchenko
- Institute for Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eugenio F Fornasiero
- Department Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen (UMG), Göttingen, Germany
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Richard L Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul W Tillberg
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Karel Svoboda
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Nelson Spruston
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| |
Collapse
|
5
|
Wingfield JL, Puthanveettil SV. Decoding the complex journeys of RNAs along neurons. Nucleic Acids Res 2025; 53:gkaf293. [PMID: 40243060 PMCID: PMC12004114 DOI: 10.1093/nar/gkaf293] [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: 12/29/2024] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Neurons are highly polarized, specialized cells that must overcome immense challenges to ensure the health and survival of the organism in which they reside. They can spread over meters and persist for decades yet communicate at sub-millisecond and millimeter scales. Thus, neurons require extreme levels of spatial-temporal control. Neurons employ molecular motors to transport coding and noncoding RNAs to distal synapses. Intracellular trafficking of RNAs enables neurons to locally regulate protein synthesis and synaptic activity. The way in which RNAs get loaded onto molecular motors and transported to their target locations, particularly following synaptic plasticity, is explored below.
Collapse
Affiliation(s)
- Jenna L Wingfield
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, United States
| | - Sathyanarayanan V Puthanveettil
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, United States
| |
Collapse
|
6
|
Rupert J, Milovanovic D. At the crossroads of calcium signaling, protein synthesis and neuropeptide release. eLife 2025; 14:e106553. [PMID: 40172946 PMCID: PMC11964446 DOI: 10.7554/elife.106553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025] Open
Abstract
By influencing calcium homeostasis, local protein synthesis and the endoplasmic reticulum, a small protein called Rab10 emerges as a crucial cytoplasmic regulator of neuropeptide secretion.
Collapse
Affiliation(s)
- Jakob Rupert
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE)Berlin and BonnGermany
| | - Dragomir Milovanovic
- Laboratory of Molecular Neuroscience, German Center for Neurodegenerative Diseases (DZNE)Berlin and BonnGermany
| |
Collapse
|
7
|
Bergadà-Martínez A, de Los Reyes-Ramírez L, Martínez-Torres S, Ciaran-Alfano L, Martínez-Gallego I, Maldonado R, Rodríguez-Moreno A, Ozaita A. Sub-chronic administration of AM6545 enhances cognitive performance and induces hippocampal synaptic plasticity changes in naïve mice. Br J Pharmacol 2025. [PMID: 40102206 DOI: 10.1111/bph.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND AND PURPOSE There is evidence of crosstalk between the brain and peripheral tissues. However, how the periphery contributes to brain function is not well understood. The cannabinoid CB1 receptor is classically pictured to have a relevant role in cognitive function. We previously demonstrated a novel mechanism where acute administration of the CB1 receptor antagonist AM6545, largely restricted to the periphery, prolonged memory persistence in mice. Here, we have assessed the effects of repeated exposure to AM6545 on cognitive improvements. EXPERIMENTAL APPROACH We evaluated, in young adult male and female mice, the behavioural consequences of sub-chronic treatment with AM6545. An unbiased transcriptomic analysis, as well as electrophysiological and biochemical studies, was carried out to elucidate the central cellular and molecular consequences of such action at peripheral receptors. KEY RESULTS Sub-chronic AM6545 enhanced memory in low and high arousal conditions in male and female mice. Executive function was facilitated after repeated AM6545 administration in male mice. Transcriptional analysis of hippocampal synaptoneurosomes from treated mice revealed a preliminary, sex-dependent, modulation of synaptic transcripts by AM6545. Notably, AM6545 occluded long-term potentiation in CA3-CA1 synapses while enhancing input-output relation in male mice. This was accompanied by an increase in hippocampal expression of Bdnf and Ngf. CONCLUSION AND IMPLICATIONS Our results showed that repeated administration of AM6545 contributed to the modulation of memory persistence, executive function and hippocampal synaptic plasticity in mice, further indicating that peripheral CB1 receptors could act as a target for a novel class of nootropic compounds.
Collapse
Affiliation(s)
- Araceli Bergadà-Martínez
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lucía de Los Reyes-Ramírez
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Group in Biology of Cognition, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Sara Martínez-Torres
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laura Ciaran-Alfano
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Group in Biology of Cognition, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Irene Martínez-Gallego
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Programme in Neurosciences, IMIM Hospital del Mar Research Institute, Barcelona, Spain
| | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain
| | - Andrés Ozaita
- Laboratory of Neuropharmacology-NeuroPhar, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Group in Biology of Cognition, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Research Programme in Neurosciences, IMIM Hospital del Mar Research Institute, Barcelona, Spain
| |
Collapse
|
8
|
de Queiroz BR, Laghrissi H, Rajeev S, Blot L, De Graeve F, Dehecq M, Hallegger M, Dag U, Dunoyer de Segonzac M, Ramialison M, Cazevieille C, Keleman K, Ule J, Hubstenberger A, Besse F. Axonal RNA localization is essential for long-term memory. Nat Commun 2025; 16:2560. [PMID: 40089499 PMCID: PMC11910521 DOI: 10.1038/s41467-025-57651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
Localization of mRNAs to neuronal terminals, coupled to local translation, has emerged as a prevalent mechanism controlling the synaptic proteome. However, the physiological regulation and function of this process in the context of mature in vivo memory circuits has remained unclear. Here, we combined synaptosome RNA profiling with whole brain high-resolution imaging to uncover mRNAs with different localization patterns in the axons of Drosophila Mushroom Body memory neurons, some exhibiting regionalized, input-dependent, recruitment along axons. By integrating transcriptome-wide binding approaches and functional assays, we show that the conserved Imp RNA binding protein controls the transport of mRNAs to Mushroom Body axons and characterize a mutant in which this transport is selectively impaired. Using this unique mutant, we demonstrate that axonal mRNA localization is required for long-term, but not short-term, behavioral memory. This work uncovers circuit-dependent mRNA targeting in vivo and demonstrates the importance of local RNA regulation in memory consolidation.
Collapse
Affiliation(s)
- Bruna R de Queiroz
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France
| | - Hiba Laghrissi
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France
| | - Seetha Rajeev
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France
| | - Lauren Blot
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France
| | - Fabienne De Graeve
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France
| | - Marine Dehecq
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France
| | - Martina Hallegger
- The Francis Crick Institute, London, UK
- UK Dementia Research Institute at King's College London, London, UK
- Oxford-GSK Institute of Molecular and Computational Medicine (IMCM), Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ugur Dag
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | | | - Mirana Ramialison
- Murdoch Children's Research Institute, Department of Paediatrics, Royal Children's Hospital, University of Melbourne, Parkville, VIC, Australia
- Australian Regenerative Medicine Institute, Clayton, VIC, Australia
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, University of Melbourne, Parkville, VIC, Australia
| | | | - Krystyna Keleman
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Jernej Ule
- The Francis Crick Institute, London, UK
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Arnaud Hubstenberger
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France
| | - Florence Besse
- Institut de Biologie Valrose, Université Côte d'Azur, CNRS, Inserm, Nice, France.
| |
Collapse
|
9
|
Zhang Z, Xu A, Bai Y, Chen Y, Cates K, Kerr C, Bermudez A, Susanto TT, Wysong K, García Marqués FJ, Nolan GP, Pitteri S, Barna M. A subcellular map of translational machinery composition and regulation at the single-molecule level. Science 2025; 387:eadn2623. [PMID: 40048539 DOI: 10.1126/science.adn2623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 10/09/2024] [Accepted: 12/16/2024] [Indexed: 04/23/2025]
Abstract
Millions of ribosomes are packed within mammalian cells, yet we lack tools to visualize them in toto and characterize their subcellular composition. In this study, we present ribosome expansion microscopy (RiboExM) to visualize individual ribosomes and an optogenetic proximity-labeling technique (ALIBi) to probe their composition. We generated a super-resolution ribosomal map, revealing subcellular translational hotspots and enrichment of 60S subunits near polysomes at the endoplasmic reticulum (ER). We found that Lsg1 tethers 60S to the ER and regulates translation of select proteins. Additionally, we discovered ribosome heterogeneity at mitochondria guiding translation of metabolism-related transcripts. Lastly, we visualized ribosomes in neurons, revealing a dynamic switch between monosomes and polysomes in neuronal translation. Together, these approaches enable exploration of ribosomal localization and composition at unprecedented resolution.
Collapse
Affiliation(s)
- Zijian Zhang
- Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Adele Xu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Yunhao Bai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Yuxiang Chen
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Kitra Cates
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Craig Kerr
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Abel Bermudez
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
| | | | - Kelsie Wysong
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Garry P Nolan
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Sharon Pitteri
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
| | - Maria Barna
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| |
Collapse
|
10
|
Milenkovic I, Novoa EM. Ribosomal protein paralogues in ribosome specialization. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230387. [PMID: 40045786 PMCID: PMC11883438 DOI: 10.1098/rstb.2023.0387] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 03/09/2025] Open
Abstract
Ribosomes are macromolecular complexes responsible for protein synthesis, comprising ribosomal proteins (RPs) and ribosomal RNA. While most RPs are present as single copies in higher eukaryotes, a handful of them have paralogues that emerged through duplication events. However, it is still unclear why a small subset of RP paralogues were preserved through evolution, and whether they can endow ribosomes with specialized functions. In this review, we focus on RP paralogue pairs present in humans, providing an overview of the most recent findings on RP paralogue functions and their roles in ribosome specialization.This article is part of the discussion meeting issue 'Ribosome diversity and its impact on protein synthesis, development and disease'.
Collapse
Affiliation(s)
- Ivan Milenkovic
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona08003, Spain
| | - Eva Maria Novoa
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona08003, Spain
- ICREA, Pg. Lluís Companys 23, Barcelona08010, Spain
| |
Collapse
|
11
|
Sahoo PK, Agrawal M, Hanovice N, Ward PJ, Desai M, Smith TP, SiMa H, Dulin JN, Vaughn LS, Tuszynski MH, Welshhans K, Benowitz LI, English AW, Houle JD, Twiss JL. Disruption of G3BP1 granules promotes mammalian CNS and PNS axon regeneration. Proc Natl Acad Sci U S A 2025; 122:e2411811122. [PMID: 40014573 PMCID: PMC11892601 DOI: 10.1073/pnas.2411811122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 01/25/2025] [Indexed: 03/01/2025] Open
Abstract
Depletion or inhibition of core stress granule proteins, G3BP1 in mammals and TIAR-2 in Caenorhabditis elegans, increases the growth of spontaneously regenerating axons. Inhibition of G3BP1 by expression of its acidic or "B-domain" accelerates axon regeneration after nerve injury, bringing a potential therapeutic strategy for peripheral nerve repair. Here, we asked whether G3BP1 inhibition is a viable strategy to promote regeneration in injured mammalian central nervous system (CNS) where axons do not regenerate spontaneously. G3BP1 B-domain expression was found to promote axon regeneration in the transected spinal cord provided with a permissive peripheral nerve graft (PNG) as well as in crushed optic nerve. Moreover, a cell-permeable peptide (CPP) to a subregion of B-domain (rodent G3BP1 amino acids 190 to 208) accelerated axon regeneration after peripheral nerve injury and promoted regrowth of reticulospinal axons into the distal transected spinal cord through a bridging PNG. G3BP1 CPP promoted axon growth from rodent and human neurons cultured on permissive substrates, and this function required alternating Glu/Asp-Pro repeats that impart a unique predicted tertiary structure. The G3BP1 CPP disassembles axonal G3BP1, G3BP2, and FMRP, but not FXR1, granules and selectively increases axonal protein synthesis in cortical neurons. These studies identify G3BP1 granules as a key regulator of axon growth in CNS neurons and demonstrate that disassembly of these granules promotes retinal axon regeneration in injured optic nerve and reticulospinal axon elongation into permissive environments after CNS injury. This work highlights G3BP1 granule disassembly as a potential therapeutic strategy for enhancing axon growth and neural repair.
Collapse
Affiliation(s)
- Pabitra K. Sahoo
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Rutgers University–Newark, Newark, NJ07102
| | - Manasi Agrawal
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Rutgers University–Newark, Newark, NJ07102
- Department of Biological Sciences, School of Biomedical Sciences, Kent State University, Kent, OH44242
| | - Nicholas Hanovice
- Departments of Neurosurgery and Ophthalmology, Boston Children’s Hospital, Cambridge, MA02115
| | - Patricia J. Ward
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, GA30332
| | - Meghal Desai
- Department of Biological Sciences, Rutgers University–Newark, Newark, NJ07102
| | - Terika P. Smith
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
| | - HaoMin SiMa
- Departments of Neurosurgery and Ophthalmology, Boston Children’s Hospital, Cambridge, MA02115
| | - Jennifer N. Dulin
- Department of Neurosciences, University of California–San Diego, La Jolla, CA92093
- Department of Biology, Texas A&M University, College Station, TX77843
| | - Lauren S. Vaughn
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
| | - Mark H. Tuszynski
- Department of Neurosciences, University of California–San Diego, La Jolla, CA92093
| | - Kristy Welshhans
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC29208
| | - Larry I. Benowitz
- Departments of Neurosurgery and Ophthalmology, Boston Children’s Hospital, Cambridge, MA02115
| | - Arthur W. English
- Department of Cell Biology, School of Medicine, Emory University, Atlanta, GA30332
| | - John D. Houle
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA19129
| | - Jeffery L. Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, SC29208
- Department of Biological Sciences, Carolina Autism and Neurodevelopment Research Center, University of South Carolina, Columbia, SC29208
| |
Collapse
|
12
|
de la Cruz-Gambra A, Baleriola J. Astrocyte-secreted factors modulate synaptic protein synthesis as revealed by puromycin labeling of isolated synaptosomes. Front Mol Neurosci 2025; 18:1427036. [PMID: 40051914 PMCID: PMC11882599 DOI: 10.3389/fnmol.2025.1427036] [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: 05/02/2024] [Accepted: 01/24/2025] [Indexed: 03/09/2025] Open
Abstract
The synaptic proteome can be shaped by proteins transported from the neuronal soma and/or by mRNAs that are delivered to synapses where proteins are locally synthesized. This last mechanism is known as local translation. Local translation has been extensively studied in neurons in physiological conditions and, more recently, in neurological disorders, in which local transcriptomes and translatomes become dysregulated. It is widely believed that in neurons, the main source of localized transcripts is the neuronal soma and that localized translation is primarily regulated by the neuron itself. However, we wondered whether glial cells, especially astrocytes, could contribute to the modulation of synaptic local protein synthesis. To address this question, we compared levels of proteins produced in synaptic compartments in neuronal and neuron-astrocyte co-cultures using modified Boyden chambers or astrocyte-conditioned medium. We developed a methodology to measure local protein synthesis by puromycin labeling of isolated synaptosomes devoid of somatic input. Our results show that synaptic local translation is enhanced or retained when neurons are cultured in the presence of astrocytes and in response to astrocyte-conditioned medium. Puromycin labeling coupled with proximity ligation identified Rpl26 as one of the proteins whose local synthesis is regulated by astrocyte-secreted factors. Our results thus unravel the contribution of glia to synaptic protein synthesis and point to a previously unexplored extra layer of complexity in the regulation of local translation in neurons.
Collapse
Affiliation(s)
- Aida de la Cruz-Gambra
- Laboratory of Local Translation in Neurons and Glia, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Neuroscience Department, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jimena Baleriola
- Laboratory of Local Translation in Neurons and Glia, Achucarro Basque Center for Neuroscience, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
13
|
Hahn A, Reed MB, Murgaš M, Vraka C, Klug S, Schmidt C, Godbersen GM, Eggerstorfer B, Gomola D, Silberbauer LR, Nics L, Philippe C, Hacker M, Lanzenberger R. Dynamics of human serotonin synthesis differentially link to reward anticipation and feedback. Mol Psychiatry 2025; 30:600-607. [PMID: 39179904 PMCID: PMC11746133 DOI: 10.1038/s41380-024-02696-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/26/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024]
Abstract
Serotonin (5-HT) plays an essential role in reward processing, however, the possibilities to investigate 5-HT action in humans during emotional stimulation are particularly limited. Here we demonstrate the feasibility of assessing reward-specific dynamics in 5-HT synthesis using functional PET (fPET), combining its molecular specificity with the high temporal resolution of blood oxygen level dependent (BOLD) fMRI. Sixteen healthy volunteers underwent simultaneous fPET/fMRI with the radioligand [11C]AMT, a substrate for tryptophan hydroxylase. During the scan, participants completed the monetary incentive delay task and arterial blood samples were acquired for quantifying 5-HT synthesis rates. BOLD fMRI was recorded as a proxy of neuronal activation, allowing differentiation of reward anticipation and feedback. Monetary gain and loss resulted in substantial increases in 5-HT synthesis in the ventral striatum (VStr, +21% from baseline) and the anterior insula (+41%). In the VStr, task-specific 5-HT synthesis was further correlated with BOLD signal changes during reward feedback (ρ = -0.65), but not anticipation. Conversely, 5-HT synthesis in the anterior insula correlated with BOLD reward anticipation (ρ = -0.61), but not feedback. In sum, we provide a robust tool to identify task-induced changes in 5-HT action in humans, linking the dynamics of 5-HT synthesis to distinct phases of reward processing in a regionally specific manner. Given the relevance of altered reward processing in psychiatric disorders such as addiction, depression and schizophrenia, our approach offers a tailored assessment of impaired 5-HT signaling during cognitive and emotional processing.
Collapse
Affiliation(s)
- Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria.
| | - Murray B Reed
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Matej Murgaš
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Chrysoula Vraka
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Sebastian Klug
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Clemens Schmidt
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Godber M Godbersen
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Benjamin Eggerstorfer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - David Gomola
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Leo R Silberbauer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Cécile Philippe
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
- Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
14
|
van Oostrum M, Schuman EM. Understanding the molecular diversity of synapses. Nat Rev Neurosci 2025; 26:65-81. [PMID: 39638892 DOI: 10.1038/s41583-024-00888-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Synapses are composed of thousands of proteins, providing the potential for extensive molecular diversity to shape synapse type-specific functional specializations. In this Review, we explore the landscape of synaptic diversity and describe the mechanisms that expand the molecular complexity of synapses, from the genotype to the regulation of gene expression to the production of specific proteoforms and the formation of localized protein complexes. We emphasize the importance of examining every molecular layer and adopting a systems perspective to understand how these interconnected mechanisms shape the diverse functional and structural properties of synapses. We explore current frameworks for classifying synapses and methodologies for investigating different synapse types at varying scales, from synapse-type-specific proteomics to advanced imaging techniques with single-synapse resolution. We highlight the potential of synapse-type-specific approaches for integrating molecular data with cellular functions, circuit organization and organismal phenotypes to enable a more holistic exploration of neuronal phenomena across different scales.
Collapse
Affiliation(s)
- Marc van Oostrum
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
- Biozentrum, University of Basel, Basel, Switzerland
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany.
| |
Collapse
|
15
|
Winden KD, Ruiz JF, Sahin M. Construction destruction: Contribution of dyregulated proteostasis to neurodevelopmental disorders. Curr Opin Neurobiol 2025; 90:102934. [PMID: 39612590 PMCID: PMC11839335 DOI: 10.1016/j.conb.2024.102934] [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: 09/04/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
Genetic causes of neurodevelopmental disorders (NDDs) such as epilepsy and autism spectrum disorder are rapidly being uncovered. The genetic risk factors that are responsible for various NDDs fall into many categories, and while some genes such as those involved in synaptic transmission are expected, there are several other classes of genes whose involvement in these disorders is not intuitive. One such group of genes is involved in protein synthesis and degradation, and the balance between these opposing pathways is termed proteostasis. Here, we review these pathways, the genetics of the related neurological disorders, and some potential disease mechanisms. Improved understanding of this collection of genetic disorders will be informative for the pathogenesis of these disorders and imply novel therapeutic strategies.
Collapse
Affiliation(s)
- Kellen D Winden
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Juan F Ruiz
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Wijegunawardana D, Nayak A, Vishal SS, Venkatesh N, Gopal PP. Ataxin-2 polyglutamine expansions aberrantly sequester TDP-43 ribonucleoprotein condensates disrupting mRNA transport and local translation in neurons. Dev Cell 2025; 60:253-269.e5. [PMID: 39419034 PMCID: PMC12063900 DOI: 10.1016/j.devcel.2024.09.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/10/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
Altered RNA metabolism and misregulation of transactive response DNA-binding protein of 43 kDa (TDP-43), an essential RNA-binding protein (RBP), define amyotrophic lateral sclerosis (ALS). Intermediate-length polyglutamine (polyQ) expansions of Ataxin-2, a like-Sm (LSm) RBP, are associated with increased risk for ALS, but the underlying biological mechanisms remain unknown. Here, we studied the spatiotemporal dynamics and mRNA regulatory functions of TDP-43 and Ataxin-2 ribonucleoprotein (RNP) condensates in rodent (rat) primary cortical neurons and mouse motor neuron axons in vivo. We report that Ataxin-2 polyQ expansions aberrantly sequester TDP-43 within RNP condensates and disrupt both its motility along the axon and liquid-like properties. We provide evidence that Ataxin-2 governs motility and translation of neuronal RNP condensates and that Ataxin-2 polyQ expansions fundamentally perturb spatial localization of mRNA and suppress local translation. Overall, our results support a model in which Ataxin-2 polyQ expansions disrupt stability, localization, and/or translation of critical axonal and cytoskeletal mRNAs, particularly important for motor neuron integrity.
Collapse
Affiliation(s)
- Denethi Wijegunawardana
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA
| | - Asima Nayak
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sonali S Vishal
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Neha Venkatesh
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA; College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Pallavi P Gopal
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
17
|
Paget-Blanc V, Pronot M, Pfeffer ME, Angelo MF, Herzog E. Purification of Afference-Specific Synaptosome Populations Using Fluorescence-Activated Synaptosome Sorting. Methods Mol Biol 2025; 2910:87-104. [PMID: 40220095 DOI: 10.1007/978-1-0716-4446-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
The central nervous system contains a complex intermingled network of neuronal, glial, and vascular cells, and for several decades, neurobiologists have used subcellular fractionation methods to analyze the molecular structure and functional features of the different cell populations. Biochemists have optimized fractionation protocols that enrich specific compartments such as synapses (called "synaptosomes") and synaptic vesicles to reduce this complexity. However, these approaches suffered from a lack of specificity and purity, which is why we previously extended the conventional synaptosome preparation to purify fluorescent synaptosomes from VGLUT1venus knock-in mice on a cell sorter. We adapted our previous protocol to sort from single neuronal projections and small target regions of the brain as we did in the present example by labeling dopaminergic projections to the striatum. We proved that our newest method allows a steep enrichment in fluorescent dopaminergic synaptosomes containing presynaptic varicosities and associated postsynaptic elements and a substantial depletion in glial contaminants. Here we propose a detailed procedure for implementing projection-specific fluorescence-activated synaptosome sorting.
Collapse
Affiliation(s)
- Vincent Paget-Blanc
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Marie Pronot
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Marlene E Pfeffer
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Maria Florencia Angelo
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Etienne Herzog
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
18
|
Hardy PB, Wang BY, Chan KM, Webber CA, Senger JLB. The use of electrical stimulation to enhance recovery following peripheral nerve injury. Muscle Nerve 2024; 70:1151-1162. [PMID: 39347555 DOI: 10.1002/mus.28262] [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: 01/17/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024]
Abstract
Peripheral nerve injury is common and can have devastating consequences. In severe cases, functional recovery is often poor despite surgery. This is primarily due to the exceedingly slow rate of nerve regeneration at only 1-3 mm/day. The local environment in the distal nerve stump supportive of nerve regrowth deteriorates over time and the target end organs become atrophic. To overcome these challenges, investigations into treatments capable of accelerating nerve regrowth are of great clinical relevance and are an active area of research. One intervention that has shown great promise is perioperative electrical stimulation. Postoperative stimulation helps to expedite the Wallerian degeneration process and reduces delays caused by staggered regeneration at the site of nerve injury. By contrast, preoperative "conditioning" stimulation increases the rate of nerve regrowth along the nerve trunk. Over the past two decades, a rich body of literature has emerged that provides molecular insights into the mechanism by which electrical stimulation impacts nerve regeneration. The end result is upregulation of regeneration-associated genes in the neuronal body and accelerated transport to the axon front for regrowth. The efficacy of brief electrical stimulation on patients with peripheral nerve injuries was demonstrated in a number of randomized controlled trials on compressive, transection and traction injuries. As approved equipment to deliver this treatment is becoming available, it may be feasible to deploy this novel treatment in a wide range of clinical settings.
Collapse
Affiliation(s)
- Paige B Hardy
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Bonnie Y Wang
- Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, Alberta, Canada
| | - K Ming Chan
- Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, Alberta, Canada
| | | | - Jenna-Lynn B Senger
- Division of Plastic & Reconstructive Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
19
|
Wang J, Li Y, Struebing FL, Jardines S, Lin ST, Lin F, Geisert EE. Dnajc3 (HSP40) Enhances Axon Regeneration in the Mouse Optic Nerve. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617251. [PMID: 39416210 PMCID: PMC11482814 DOI: 10.1101/2024.10.08.617251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
A forward genetics approach was used to identify genomic elements enhancing axon regeneration in the BXD recombinant mouse strains. Axon regeneration was induced by knocking down Pten in retinal ganglion cells (RGCs) using adeno-associated virus (AAV) to deliver an shRNA followed by an intravitreal injection of Zymosan with CPT-cAMP that produced a mild inflammatory response. RGC axons were damaged by optic nerve crush (ONC). Following a 12-day survival period, regenerating axons were labeled by intravitreal injection of Cholera Toxin B (CTB) conjugated with Alexa Fluor 647. Two days later, labeled axons within the optic nerve were examined to determine the number of regenerating axons and the distance they traveled down the optic nerve. The analysis revealed a surprising difference in the amount of axonal regeneration across all 33 BXD strains. There was a 7.5-fold difference in the number of regenerating axons and a 4-fold difference in distance traveled by regenerating axons. These data were used to generate an integral map defining genomic loci modulating the enhanced axonal regeneration. A quantitative trait locus modulating axon regeneration was identified on Chromosome 14 (115 to 119 Mb). Within this locus were 16 annotated genes. Subsequent testing revealed that one candidate gene, Dnajc3 , modulates axonal regeneration. Dnajc3 encodes Heat Shock Protein 40 (HSP40), which is a molecular chaperone. Knocking down Dnajc3 in the high regenerative strain (BXD90) led to a decreased regeneration response, while overexpression of Dnajc3 in a low regenerative strain (BXD34) resulted in an increased regeneration response. These findings suggest that Dnajc3 not only increases the number of regenerating axons, it also increases the distance those axons travel. This may prove to be critical for functional recovery in large mammals, where the distance axons travel to their target is considerably longer than that of the mouse. Thus, Dnajc3 may play a critical role for functional recovery in humans by increasing the number of regenerating axons and the distance the regenerating axons travel.
Collapse
|
20
|
Tomé D, Almeida RD. The injured axon: intrinsic mechanisms driving axonal regeneration. Trends Neurosci 2024; 47:875-891. [PMID: 39438216 DOI: 10.1016/j.tins.2024.09.009] [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: 04/15/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Injury to the central nervous system (CNS) often results in permanent neurological impairments because axons fail to regenerate and re-establish lost synaptic contacts. By contrast, peripheral neurons can activate a pro-regenerative program and regenerate following a nerve lesion. This relies on an intricate intracellular communication system between the severed axon and the cell body. Locally activated signaling molecules are retrogradely transported to the soma to promote the epigenetic and transcriptional changes required for the injured neuron to regain growth competence. These signaling events rely heavily on intra-axonal translation and mitochondrial trafficking into the severed axon. Here, we discuss the interplay between these mechanisms and the main intrinsic barriers to axonal regeneration. We also examine the potential of manipulating these processes for driving CNS repair.
Collapse
Affiliation(s)
- Diogo Tomé
- iBiMED- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | - Ramiro D Almeida
- iBiMED- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
21
|
Kiebler MA, Bauer KE. RNA granules in flux: dynamics to balance physiology and pathology. Nat Rev Neurosci 2024; 25:711-725. [PMID: 39367081 DOI: 10.1038/s41583-024-00859-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2024] [Indexed: 10/06/2024]
Abstract
The life cycle of an mRNA is a complex process that is tightly regulated by interactions between the mRNA and RNA-binding proteins, forming molecular machines known as RNA granules. Various types of these membrane-less organelles form inside cells, including neurons, and contribute critically to various physiological processes. RNA granules are constantly in flux, change dynamically and adapt to their local environment, depending on their intracellular localization. The discovery that RNA condensates can form by liquid-liquid phase separation expanded our understanding of how compartments may be generated in the cell. Since then, a plethora of new functions have been proposed for distinct condensates in cells that await their validation in vivo. The finding that dysregulation of RNA granules (for example, stress granules) is likely to affect neurodevelopmental and neurodegenerative diseases further boosted interest in this topic. RNA granules have various physiological functions in neurons and in the brain that we would like to focus on. We outline examples of state-of-the-art experiments including timelapse microscopy in neurons to unravel the precise functions of various types of RNA granule. Finally, we distinguish physiologically occurring RNA condensation from aberrant aggregation, induced by artificial RNA overexpression, and present visual examples to discriminate both forms in neurons.
Collapse
Affiliation(s)
- Michael A Kiebler
- Biomedical Center (BMC), Department of Cell Biology and Anatomy, Medical Faculty, Ludwig-Maximilians-University, Planegg-Martinsried, Germany.
| | - Karl E Bauer
- Biomedical Center (BMC), Department of Cell Biology and Anatomy, Medical Faculty, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| |
Collapse
|
22
|
Navarri X, Robertson DN, Charfi I, Wünnemann F, Sâmia Fernandes do Nascimento A, Trottier G, Leclerc S, Andelfinger GU, Di Cristo G, Richer L, Pike GB, Pausova Z, Piñeyro G, Paus T. Cells and Molecules Underpinning Cannabis-Related Variations in Cortical Thickness during Adolescence. J Neurosci 2024; 44:e2256232024. [PMID: 39214708 PMCID: PMC11466068 DOI: 10.1523/jneurosci.2256-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
During adolescence, cannabis experimentation is common, and its association with interindividual variations in brain maturation well studied. Cellular and molecular underpinnings of these system-level relationships are, however, unclear. We thus conducted a three-step study. First, we exposed adolescent male mice to Δ-9-tetrahydrocannabinol (THC) or a synthetic cannabinoid WIN 55,212-2 (WIN) and assessed differentially expressed genes (DEGs), spine numbers, and dendritic complexity in their frontal cortex. Second, in human (male) adolescents, we examined group differences in cortical thickness in 34 brain regions, using magnetic resonance imaging, between those who experimented with cannabis before age 16 (n = 140) and those who did not (n = 327). Finally, we correlated spatially these group differences with gene expression of human homologs of mouse-identified DEGs. The spatial expression of 13 THC-related human homologs of DEGs correlated with cannabis-related variations in cortical thickness, and virtual histology revealed coexpression patterns of these 13 genes with cell-specific markers of astrocytes, microglia, and a type of pyramidal cells enriched in dendrite-regulating genes. Similarly, the spatial expression of 18 WIN-related human homologs of DEGs correlated with group differences in cortical thickness and showed coexpression patterns with the same three cell types. Gene ontology analysis indicated that 37 THC-related human homologs are enriched in neuron projection development, while 33 WIN-related homologs are enriched in processes associated with learning and memory. In mice, we observed spine loss and lower dendritic complexity in pyramidal cells of THC-exposed animals (vs controls). Experimentation with cannabis during adolescence may influence cortical thickness by impacting glutamatergic synapses and dendritic arborization.
Collapse
Affiliation(s)
- Xavier Navarri
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
| | | | - Iness Charfi
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Florian Wünnemann
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | | | - Giacomo Trottier
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Sévérine Leclerc
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Gregor U Andelfinger
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Graziella Di Cristo
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Louis Richer
- Department of Health Sciences, Université du Québec à Chicoutimi, Chicoutimi, Quebec G7H 2B1, Canada
| | - G Bruce Pike
- Departments of Radiology and Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Zdenka Pausova
- Departments of Physiology and Nutritional Sciences, Hospital for Sick Children, University of Toronto, Peter Gilgan Centre for Research and Learning, Toronto, Ontario M5G 0A4, Canada
| | - Graciela Piñeyro
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Tomáš Paus
- Department of Neuroscience, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
- CHU Ste-Justine Research Centre, Montréal, Quebec H3T 1C5, Canada
- Department of Psychiatry and Addictology, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
23
|
Ren J, Luo S, Shi H, Wang X. Spatial omics advances for in situ RNA biology. Mol Cell 2024; 84:3737-3757. [PMID: 39270643 PMCID: PMC11455602 DOI: 10.1016/j.molcel.2024.08.002] [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: 05/21/2024] [Revised: 07/07/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024]
Abstract
Spatial regulation of RNA plays a critical role in gene expression regulation and cellular function. Understanding spatially resolved RNA dynamics and translation is vital for bringing new insights into biological processes such as embryonic development, neurobiology, and disease pathology. This review explores past studies in subcellular, cellular, and tissue-level spatial RNA biology driven by diverse methodologies, ranging from cell fractionation, in situ and proximity labeling, imaging, spatially indexed next-generation sequencing (NGS) approaches, and spatially informed computational modeling. Particularly, recent advances have been made for near-genome-scale profiling of RNA and multimodal biomolecules at high spatial resolution. These methods enabled new discoveries into RNA's spatiotemporal kinetics, RNA processing, translation status, and RNA-protein interactions in cells and tissues. The evolving landscape of experimental and computational strategies reveals the complexity and heterogeneity of spatial RNA biology with subcellular resolution, heralding new avenues for RNA biology research.
Collapse
Affiliation(s)
- Jingyi Ren
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shuchen Luo
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailing Shi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Xiao Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
24
|
Caglayan E, Konopka G. Evolutionary neurogenomics at single-cell resolution. Curr Opin Genet Dev 2024; 88:102239. [PMID: 39094380 DOI: 10.1016/j.gde.2024.102239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/20/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
The human brain is composed of increasingly recognized heterogeneous cell types. Applying single-cell genomics to brain tissue can elucidate relative cell type proportions as well as differential gene expression and regulation among humans and other species. Here, we review recent studies that utilized high-throughput genomics approaches to compare brains among species at single-cell resolution. These studies identified genomic elements that are similar among species as well as evolutionary novelties on the human lineage. We focus on those human-relevant innovations and discuss the biological implications of these modifications. Finally, we discuss areas of comparative single-cell genomics that remain unexplored either due to needed technological advances or due to biological availability at the brain region or species level.
Collapse
Affiliation(s)
- Emre Caglayan
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Genevieve Konopka
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
25
|
Zhu B, Zhu J, Liu A, Yao B, Liao F, Yang S. Transcriptomic and metabolomic analysis based on different aggressive pecking phenotype in duck. Sci Rep 2024; 14:22321. [PMID: 39333746 PMCID: PMC11436778 DOI: 10.1038/s41598-024-73726-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Aggressive pecking is an important welfare and production efficiency issue in poultry farming. The precise mechanisms underlying the occurrence of aggressive pecking remain poorly understood. In this study, we selected Sansui ducks that performed aggressive pecking and ducks that did not perform aggressive pecking from video recordings. Transcriptomic and metabolomic analyses of the whole brains of aggressive pecking ducks and normal ducks revealed 504 differentially expressed genes and 5 differentially altered metabolites (adenosine, guanidinopropionic acid, Met-Leu, Glu-Ile and 5,6,8-trihydroxy-2-methylbenzo[g]chromen-4-one). By jointly analysing the transcriptomics and metabolomics results, we discovered 8 candidate genes (ADCYAP1, GAL, EDN2, EDN1, MC5R, S1PR4, LOC113843450, and IAPP) and one candidate metabolite (adenosine) that regulates aggressive pecking behaviour in ducks. The candidate genes and metabolites may be involved in regulating aggressive pecking behaviour by inducing neurodegeneration and disrupting neural excitatory-inhibitory homeostasis, which in turn affects central nervous system function in aggressive pecking and normal ducks. Our findings provide a new reference for revealing the underlying mechanism of aggressive pecking behaviour in ducks.
Collapse
Affiliation(s)
- Baoguo Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Jinjin Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Ai Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Bingnong Yao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Fuyou Liao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou province, Guizhou University, Guiyang, 550025, China
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Shenglin Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, 550025, China.
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Guizhou province, Guizhou University, Guiyang, 550025, China.
- College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
26
|
Tu WY, Xu W, Bai L, Liu J, Han Y, Luo B, Wang B, Zhang K, Shen C. Local protein synthesis at neuromuscular synapses is required for motor functions. Cell Rep 2024; 43:114661. [PMID: 39178112 DOI: 10.1016/j.celrep.2024.114661] [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: 10/17/2023] [Revised: 06/27/2024] [Accepted: 08/05/2024] [Indexed: 08/25/2024] Open
Abstract
Motor neurons are highly polarized, and their axons extend over great distances to form connections with myofibers via neuromuscular junctions (NMJs). Local translation at the NMJs in vivo has not been identified. Here, we utilized motor neuron-labeled RiboTag mice and the TRAP (translating ribosome affinity purification) technique to spatiotemporally profile the translatome at NMJs. We found that mRNAs associated with glucose catabolism, synaptic connection, and protein homeostasis are enriched at presynapses. Local translation at the synapse shifts from the assembly of cytoskeletal components during early developmental stages to energy production in adulthood. The mRNA of neuronal Agrin (Agrn), the key molecule for NMJ assembly, is present at motor axon terminals and locally translated. Disrupting the axonal location of Agrn mRNA causes impairment of synaptic transmission and motor functions in adult mice. Our findings indicate that spatiotemporal regulation of mRNA local translation at NMJs plays critical roles in synaptic transmission and motor functions in vivo.
Collapse
Affiliation(s)
- Wen-Yo Tu
- Department of Neurobiology of First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Wentao Xu
- Department of Neurobiology of First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Lei Bai
- Department of Neurobiology of First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Jun Liu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Han
- Department of Neurobiology of First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Benyan Luo
- Department of Neurobiology of First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Bingwei Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Kejing Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Hangzhou 310006, China.
| | - Chengyong Shen
- Department of Neurobiology of First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China; MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
27
|
Li Y, Uhelski ML, North RY, Mwirigi JM, Tatsui CE, McDonough KE, Cata JP, Corrales G, Dussor G, Price TJ, Dougherty PM. Tomivosertib reduces ectopic activity in dorsal root ganglion neurons from patients with radiculopathy. Brain 2024; 147:2991-2997. [PMID: 39046204 PMCID: PMC11370786 DOI: 10.1093/brain/awae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 07/25/2024] Open
Abstract
Spontaneous activity in dorsal root ganglion (DRG) neurons is a key driver of neuropathic pain in patients suffering from this largely untreated disease. While many intracellular signalling mechanisms have been examined in preclinical models that drive spontaneous activity, none have been tested directly on spontaneously active human nociceptors. Using cultured DRG neurons recovered during thoracic vertebrectomy surgeries, we showed that inhibition of mitogen-activated protein kinase interacting kinase (MNK) with tomivosertib (eFT508, 25 nM) reversibly suppresses spontaneous activity in human sensory neurons that are likely nociceptors based on size and action potential characteristics associated with painful dermatomes within minutes of treatment. Tomivosertib treatment also decreased action potential amplitude and produced alterations in the magnitude of after hyperpolarizing currents, suggesting modification of Na+ and K+ channel activity as a consequence of drug treatment. Parallel to the effects on electrophysiology, eFT508 treatment led to a profound loss of eIF4E serine 209 phosphorylation in primary sensory neurons, a specific substrate of MNK, within 2 min of drug treatment. Our results create a compelling case for the future testing of MNK inhibitors in clinical trials for neuropathic pain.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Megan L Uhelski
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Y North
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juliet M Mwirigi
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Claudio E Tatsui
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kathleen E McDonough
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Juan P Cata
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - German Corrales
- Department of Anesthesiology & Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Greg Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Patrick M Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
28
|
Zare A, Salehi S, Bader J, Schneider C, Fischer U, Veh A, Arampatzi P, Mann M, Briese M, Sendtner M. hnRNP R promotes O-GlcNAcylation of eIF4G and facilitates axonal protein synthesis. Nat Commun 2024; 15:7430. [PMID: 39198412 PMCID: PMC11358521 DOI: 10.1038/s41467-024-51678-y] [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: 10/16/2023] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Motoneurons critically depend on precise spatial and temporal control of translation for axon growth and the establishment and maintenance of neuromuscular connections. While defects in local translation have been implicated in the pathogenesis of motoneuron disorders, little is known about the mechanisms regulating axonal protein synthesis. Here, we report that motoneurons derived from Hnrnpr knockout mice show reduced axon growth accompanied by lowered synthesis of cytoskeletal and synaptic components in axons. Mutant mice display denervated neuromuscular junctions and impaired motor behavior. In axons, hnRNP R is a component of translation initiation complexes and, through interaction with O-linked β-N-acetylglucosamine (O-GlcNAc) transferase (Ogt), modulates O-GlcNAcylation of eIF4G. Restoring axonal O-GlcNAc levels rescued local protein synthesis and axon growth defects of hnRNP R knockout motoneurons. Together, these findings demonstrate a function of hnRNP R in controlling the local production of key factors required for axon growth and formation of neuromuscular innervations.
Collapse
Affiliation(s)
- Abdolhossein Zare
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Saeede Salehi
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Jakob Bader
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Cornelius Schneider
- Department of Biochemistry, Theodor Boveri Institute, University of Wuerzburg, Wuerzburg, Germany
| | - Utz Fischer
- Department of Biochemistry, Theodor Boveri Institute, University of Wuerzburg, Wuerzburg, Germany
| | - Alexander Veh
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | | | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Briese
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany.
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
29
|
Tarannum R, Mun G, Quddos F, Swanger SA, Steward O, Farris S. Dendritically localized RNAs are packaged as diversely composed ribonucleoprotein particles with heterogeneous copy number states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603387. [PMID: 39071419 PMCID: PMC11275876 DOI: 10.1101/2024.07.13.603387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Localization of mRNAs to dendrites is a fundamental mechanism by which neurons achieve spatiotemporal control of gene expression. Translationally repressed neuronal mRNA transport granules, also referred to as ribonucleoprotein particles (RNPs), have been shown to be trafficked as single or low copy number RNPs and as larger complexes with multiple copies and/or species of mRNAs. However, there is little evidence of either population in intact neuronal circuits. Using single molecule fluorescence in situ hybridization studies in the dendrites of adult rat and mouse hippocampus, we provide evidence that supports the existence of multi-transcript RNPs with the constituents varying in amounts for each RNA species. By competing-off fluorescently labeled probe with serial increases of unlabeled probe, we detected stepwise decreases in Arc RNP number and fluorescence intensity, suggesting Arc RNAs localize to dendrites in both low- and multiple-copy number RNPs. When probing for multiple mRNAs, we find that localized RNPs are heterogeneous in size and colocalization patterns that vary per RNA. Further, localized RNAs that are targeted by the same trans-acting element (FMRP) display greater levels of colocalization compared to an RNA not targeted by FMRP. Simultaneous visualization of a dozen FMRP-targeted mRNA species using highly multiplexed imaging demonstrates that dendritic RNAs are mostly trafficked as heteromeric cargoes of multiple types of RNAs (at least one or more RNAs). Moreover, the composition of these RNA cargoes, as assessed by colocalization, correlates with the abundance of the transcripts even after accounting for the expected differences in colocalization based on expression. Collectively, these results suggest that dendritic RNPs are packaged as heterogeneous co-assemblies of different mRNAs and that RNP contents may be driven, at least partially, by highly abundant dendritic RNAs; a model that favors efficiency over fine-tuned control for sustaining long-distance trafficking of thousands of messenger molecules.
Collapse
Affiliation(s)
- Renesa Tarannum
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, Virginia
- Translational Biology, Medicine & Health Graduate Program, Virginia Tech, Blacksburg, Virginia
| | - Grace Mun
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, Virginia
| | - Fatima Quddos
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, Virginia
- Translational Biology, Medicine & Health Graduate Program, Virginia Tech, Blacksburg, Virginia
| | - Sharon A. Swanger
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, Virginia
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia
| | | | - Shannon Farris
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Neurobiology Research, Roanoke, Virginia
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia
| |
Collapse
|
30
|
Shi Z, Wen K, Zou Z, Fu W, Guo K, Sammudin NH, Ruan X, Sullere S, Wang S, Zhang X, Thinakaran G, He C, Zhuang X. YTHDF1 mediates translational control by m6A mRNA methylation in adaptation to environmental challenges. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.607063. [PMID: 39149343 PMCID: PMC11326287 DOI: 10.1101/2024.08.07.607063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Animals adapt to environmental challenges with long-term changes at the behavioral, circuit, cellular, and synaptic levels which often require new protein synthesis. The discovery of reversible N6-methyladenosine (m6A) modifications of mRNA has revealed an important layer of post-transcriptional regulation which affects almost every phase of mRNA metabolism and therefore translational control. Many in vitro and in vivo studies have demonstrated the significant role of m6A in cell differentiation and survival, but its role in adult neurons is understudied. We used cell-type specific gene deletion of Mettl14, which encodes one of the subunits of the m6A methyltransferase, and Ythdf1, which encodes one of the cytoplasmic m6A reader proteins, in dopamine D1 receptor expressing or D2 receptor expressing neurons. Mettl14 or Ythdf1 deficiency blunted responses to environmental challenges at the behavioral, cellular, and molecular levels. In three different behavioral paradigms, gene deletion of either Mettl14 or Ythdf1 in D1 neurons impaired D1-dependent learning, whereas gene deletion of either Mettl14 or Ythdf1 in D2 neurons impaired D2-dependent learning. At the cellular level, modulation of D1 and D2 neuron firing in response to changes in environments was blunted in all three behavioral paradigms in mutant mice. Ythdf1 deletion resembled impairment caused by Mettl14 deletion in a cell type-specific manner, suggesting YTHDF1 is the main mediator of the functional consequences of m6A mRNA methylation in the striatum. At the molecular level, while striatal neurons in control mice responded to elevated cAMP by increasing de novo protein synthesis, striatal neurons in Ythdf1 knockout mice didn't. Finally, boosting dopamine release by cocaine drastically increased YTHDF1 binding to many mRNA targets in the striatum, especially those that encode structural proteins, suggesting the initiation of long-term neuronal and/or synaptic structural changes. While the m6A-YTHDF1 pathway has similar functional significance at cellular level, its cell type specific deficiency in D1 and D2 neurons often resulted in contrasting behavioral phenotypes, allowing us to cleanly dissociate the opposing yet cooperative roles of D1 and D2 neurons.
Collapse
Affiliation(s)
- Zhuoyue Shi
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Kailong Wen
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Zhongyu Zou
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Wenqin Fu
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Kathryn Guo
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Nabilah H Sammudin
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Xiangbin Ruan
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Shivang Sullere
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Shuai Wang
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
| | - Xiaochang Zhang
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- The Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| | - Gopal Thinakaran
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL 33613, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Chuan He
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Howard Hughes Medical Institute, The University of Chicago, IL 60637, USA
| | - Xiaoxi Zhuang
- The Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
- The Neuroscience Institute, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
31
|
Scott-Hewitt N, Mahoney M, Huang Y, Korte N, Yvanka de Soysa T, Wilton DK, Knorr E, Mastro K, Chang A, Zhang A, Melville D, Schenone M, Hartigan C, Stevens B. Microglial-derived C1q integrates into neuronal ribonucleoprotein complexes and impacts protein homeostasis in the aging brain. Cell 2024; 187:4193-4212.e24. [PMID: 38942014 DOI: 10.1016/j.cell.2024.05.058] [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: 05/31/2023] [Revised: 01/08/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
Neuroimmune interactions mediate intercellular communication and underlie critical brain functions. Microglia, CNS-resident macrophages, modulate the brain through direct physical interactions and the secretion of molecules. One such secreted factor, the complement protein C1q, contributes to complement-mediated synapse elimination in both developmental and disease models, yet brain C1q protein levels increase significantly throughout aging. Here, we report that C1q interacts with neuronal ribonucleoprotein (RNP) complexes in an age-dependent manner. Purified C1q protein undergoes RNA-dependent liquid-liquid phase separation (LLPS) in vitro, and the interaction of C1q with neuronal RNP complexes in vivo is dependent on RNA and endocytosis. Mice lacking C1q have age-specific alterations in neuronal protein synthesis in vivo and impaired fear memory extinction. Together, our findings reveal a biophysical property of C1q that underlies RNA- and age-dependent neuronal interactions and demonstrate a role of C1q in critical intracellular neuronal processes.
Collapse
Affiliation(s)
- Nicole Scott-Hewitt
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Matthew Mahoney
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Youtong Huang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nils Korte
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - T Yvanka de Soysa
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel K Wilton
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Emily Knorr
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kevin Mastro
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Allison Chang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Allison Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - David Melville
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Monica Schenone
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christina Hartigan
- The Broad Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA; The Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Investigator, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Lederbauer J, Das S, Piton A, Lessel D, Kreienkamp HJ. The role of DEAD- and DExH-box RNA helicases in neurodevelopmental disorders. Front Mol Neurosci 2024; 17:1414949. [PMID: 39149612 PMCID: PMC11324592 DOI: 10.3389/fnmol.2024.1414949] [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: 04/09/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024] Open
Abstract
Neurodevelopmental disorders (NDDs) represent a large group of disorders with an onset in the neonatal or early childhood period; NDDs include intellectual disability (ID), autism spectrum disorders (ASD), attention deficit hyperactivity disorders (ADHD), seizures, various motor disabilities and abnormal muscle tone. Among the many underlying Mendelian genetic causes for these conditions, genes coding for proteins involved in all aspects of the gene expression pathway, ranging from transcription, splicing, translation to the eventual RNA decay, feature rather prominently. Here we focus on two large families of RNA helicases (DEAD- and DExH-box helicases). Genetic variants in the coding genes for several helicases have recently been shown to be associated with NDD. We address genetic constraints for helicases, types of pathological variants which have been discovered and discuss the biological pathways in which the affected helicase proteins are involved.
Collapse
Affiliation(s)
- Johannes Lederbauer
- Institute of Human Genetics, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarada Das
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Amelie Piton
- Department of Translational Medicine and Neurogenetics, Institute of Genetics and Molecular and Cellular Biology, Strasbourg University, CNRS UMR7104, INSERM U1258, Illkirch, France
| | - Davor Lessel
- Institute of Human Genetics, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Jürgen Kreienkamp
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
33
|
Campbell KR, Hallada LP, Huang YS, Solecki DJ. From Blur to Brilliance: The Ascendance of Advanced Microscopy in Neuronal Cell Biology. Annu Rev Neurosci 2024; 47:235-253. [PMID: 38608643 DOI: 10.1146/annurev-neuro-111020-090208] [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] [Indexed: 04/14/2024]
Abstract
The intricate network of the brain's neurons and synapses poses unparalleled challenges for research, distinct from other biological studies. This is particularly true when dissecting how neurons and their functional units work at a cell biological level. While traditional microscopy has been foundational, it was unable to reveal the deeper complexities of neural interactions. However, an imaging renaissance has transformed our capabilities. Advancements in light and electron microscopy, combined with correlative imaging, now achieve unprecedented resolutions, uncovering the most nuanced neural structures. Maximizing these tools requires more than just technical proficiency. It is crucial to align research aims, allocate resources wisely, and analyze data effectively. At the heart of this evolution is interdisciplinary collaboration, where various experts come together to translate detailed imagery into significant biological insights. This review navigates the latest developments in microscopy, underscoring both the promise of and prerequisites for bending this powerful tool set to understanding neuronal cell biology.
Collapse
Affiliation(s)
- Kirby R Campbell
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Liam P Hallada
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Yu-Shan Huang
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - David J Solecki
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| |
Collapse
|
34
|
Lu WH, Chang TT, Chang YM, Liu YH, Lin CH, Suen CS, Hwang MJ, Huang YS. CPEB2-activated axonal translation of VGLUT2 mRNA promotes glutamatergic transmission and presynaptic plasticity. J Biomed Sci 2024; 31:69. [PMID: 38992696 PMCID: PMC11241979 DOI: 10.1186/s12929-024-01061-2] [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: 11/13/2023] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Local translation at synapses is important for rapidly remodeling the synaptic proteome to sustain long-term plasticity and memory. While the regulatory mechanisms underlying memory-associated local translation have been widely elucidated in the postsynaptic/dendritic region, there is no direct evidence for which RNA-binding protein (RBP) in axons controls target-specific mRNA translation to promote long-term potentiation (LTP) and memory. We previously reported that translation controlled by cytoplasmic polyadenylation element binding protein 2 (CPEB2) is important for postsynaptic plasticity and memory. Here, we investigated whether CPEB2 regulates axonal translation to support presynaptic plasticity. METHODS Behavioral and electrophysiological assessments were conducted in mice with pan neuron/glia- or glutamatergic neuron-specific knockout of CPEB2. Hippocampal Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 pathways were electro-recorded to monitor synaptic transmission and LTP evoked by 4 trains of high-frequency stimulation. RNA immunoprecipitation, coupled with bioinformatics analysis, were used to unveil CPEB2-binding axonal RNA candidates associated with learning, which were further validated by Western blotting and luciferase reporter assays. Adeno-associated viruses expressing Cre recombinase were stereotaxically delivered to the pre- or post-synaptic region of the TA circuit to ablate Cpeb2 for further electrophysiological investigation. Biochemically isolated synaptosomes and axotomized neurons cultured on a microfluidic platform were applied to measure axonal protein synthesis and FM4-64FX-loaded synaptic vesicles. RESULTS Electrophysiological analysis of hippocampal CA1 neurons detected abnormal excitability and vesicle release probability in CPEB2-depleted SC and TA afferents, so we cross-compared the CPEB2-immunoprecipitated transcriptome with a learning-induced axonal translatome in the adult cortex to identify axonal targets possibly regulated by CPEB2. We validated that Slc17a6, encoding vesicular glutamate transporter 2 (VGLUT2), is translationally upregulated by CPEB2. Conditional knockout of CPEB2 in VGLUT2-expressing glutamatergic neurons impaired consolidation of hippocampus-dependent memory in mice. Presynaptic-specific ablation of Cpeb2 in VGLUT2-dominated TA afferents was sufficient to attenuate protein synthesis-dependent LTP. Moreover, blocking activity-induced axonal Slc17a6 translation by CPEB2 deficiency or cycloheximide diminished the releasable pool of VGLUT2-containing synaptic vesicles. CONCLUSIONS We identified 272 CPEB2-binding transcripts with altered axonal translation post-learning and established a causal link between CPEB2-driven axonal synthesis of VGLUT2 and presynaptic translation-dependent LTP. These findings extend our understanding of memory-related translational control mechanisms in the presynaptic compartment.
Collapse
Affiliation(s)
- Wen-Hsin Lu
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Tzu-Tung Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yi-Hsiang Liu
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Chia-Hsuan Lin
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, 11529, Taiwan
| | - Ching-Shu Suen
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Ming-Jing Hwang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan.
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, 11529, Taiwan.
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
35
|
Russo C, Valle MS, D’Angeli F, Surdo S, Malaguarnera L. Resveratrol and Vitamin D: Eclectic Molecules Promoting Mitochondrial Health in Sarcopenia. Int J Mol Sci 2024; 25:7503. [PMID: 39062745 PMCID: PMC11277153 DOI: 10.3390/ijms25147503] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Sarcopenia refers to the progressive loss and atrophy of skeletal muscle function, often associated with aging or secondary to conditions involving systemic inflammation, oxidative stress, and mitochondrial dysfunction. Recent evidence indicates that skeletal muscle function is not only influenced by physical, environmental, and genetic factors but is also significantly impacted by nutritional deficiencies. Natural compounds with antioxidant properties, such as resveratrol and vitamin D, have shown promise in preventing mitochondrial dysfunction in skeletal muscle cells. These antioxidants can slow down muscle atrophy by regulating mitochondrial functions and neuromuscular junctions. This review provides an overview of the molecular mechanisms leading to skeletal muscle atrophy and summarizes recent advances in using resveratrol and vitamin D supplementation for its prevention and treatment. Understanding these molecular mechanisms and implementing combined interventions can optimize treatment outcomes, ensure muscle function recovery, and improve the quality of life for patients.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy;
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Floriana D’Angeli
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Sofia Surdo
- Italian Center for the Study of Osteopathy (CSDOI), 95124 Catania, Italy;
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
36
|
Clayton EL, Huggon L, Cousin MA, Mizielinska S. Synaptopathy: presynaptic convergence in frontotemporal dementia and amyotrophic lateral sclerosis. Brain 2024; 147:2289-2307. [PMID: 38451707 PMCID: PMC11224618 DOI: 10.1093/brain/awae074] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Frontotemporal dementia and amyotrophic lateral sclerosis are common forms of neurodegenerative disease that share overlapping genetics and pathologies. Crucially, no significantly disease-modifying treatments are available for either disease. Identifying the earliest changes that initiate neuronal dysfunction is important for designing effective intervention therapeutics. The genes mutated in genetic forms of frontotemporal dementia and amyotrophic lateral sclerosis have diverse cellular functions, and multiple disease mechanisms have been proposed for both. Identification of a convergent disease mechanism in frontotemporal dementia and amyotrophic lateral sclerosis would focus research for a targetable pathway, which could potentially effectively treat all forms of frontotemporal dementia and amyotrophic lateral sclerosis (both familial and sporadic). Synaptopathies are diseases resulting from physiological dysfunction of synapses, and define the earliest stages in multiple neuronal diseases, with synapse loss a key feature in dementia. At the presynapse, the process of synaptic vesicle recruitment, fusion and recycling is necessary for activity-dependent neurotransmitter release. The unique distal location of the presynaptic terminal means the tight spatio-temporal control of presynaptic homeostasis is dependent on efficient local protein translation and degradation. Recently, numerous publications have shown that mutations associated with frontotemporal dementia and amyotrophic lateral sclerosis present with synaptopathy characterized by presynaptic dysfunction. This review will describe the complex local signalling and membrane trafficking events that occur at the presynapse to facilitate neurotransmission and will summarize recent publications linking frontotemporal dementia/amyotrophic lateral sclerosis genetic mutations to presynaptic function. This evidence indicates that presynaptic synaptopathy is an early and convergent event in frontotemporal dementia and amyotrophic lateral sclerosis and illustrates the need for further research in this area, to identify potential therapeutic targets with the ability to impact this convergent pathomechanism.
Collapse
Affiliation(s)
- Emma L Clayton
- UK Dementia Research Institute at King’s College London, London SE5 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| | - Laura Huggon
- UK Dementia Research Institute at King’s College London, London SE5 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| | - Michael A Cousin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Sarah Mizielinska
- UK Dementia Research Institute at King’s College London, London SE5 9RT, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RT, UK
| |
Collapse
|
37
|
Abstract
The translation of messenger RNA (mRNA) into proteins represents the culmination of gene expression. Recent technological advances have revolutionized our ability to investigate this process with unprecedented precision, enabling the study of translation at the single-molecule level in real time within live cells. In this review, we provide an overview of single-mRNA translation reporters. We focus on the core technology, as well as the rapid development of complementary probes, tags, and accessories that enable the visualization and quantification of a wide array of translation dynamics. We then highlight notable studies that have utilized these reporters in model systems to address key biological questions. The high spatiotemporal resolution of these studies is shedding light on previously unseen phenomena, uncovering the full heterogeneity and complexity of translational regulation.
Collapse
Affiliation(s)
- Tatsuya Morisaki
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA;
| | - O'Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA;
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA;
- Cell Biology Center and World Research Hub Initiative, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
38
|
Oliver Goral R, Lamb PW, Yakel JL. Acetylcholine Neurons Become Cholinergic during Three Time Windows in the Developing Mouse Brain. eNeuro 2024; 11:ENEURO.0542-23.2024. [PMID: 38942474 PMCID: PMC11253243 DOI: 10.1523/eneuro.0542-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/12/2024] [Accepted: 06/22/2024] [Indexed: 06/30/2024] Open
Abstract
Acetylcholine (ACh) neurons in the central nervous system are required for the coordination of neural network activity during higher brain functions, such as attention, learning, and memory, as well as locomotion. Disturbed cholinergic signaling has been described in many neurodevelopmental and neurodegenerative disorders. Furthermore, cotransmission of other signaling molecules, such as glutamate and GABA, with ACh has been associated with essential roles in brain function or disease. However, it is unknown when ACh neurons become cholinergic during development. Thus, understanding the timeline of how the cholinergic system develops and becomes active in the healthy brain is a crucial part of understanding brain development. To study this, we used transgenic mice to selectively label ACh neurons with tdTomato. We imaged serial sectioned brains and generated whole-brain reconstructions at different time points during pre- and postnatal development. We found three crucial time windows-two in the prenatal and one in the postnatal brain-during which most ACh neuron populations become cholinergic in the brain. We also found that cholinergic gene expression is initiated in cortical ACh interneurons, while the cerebral cortex is innervated by cholinergic projection neurons from the basal forebrain. Taken together, we show that ACh neuron populations are present and become cholinergic before postnatal day 12, which is the onset of major sensory processes, such as hearing and vision. We conclude that the birth of ACh neurons and initiation of cholinergic gene expression are temporally separated during development but highly coordinated by brain anatomical structure.
Collapse
Affiliation(s)
- Rene Oliver Goral
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
- Center on Compulsive Behaviors, National Institutes of Health, Bethesda, Maryland 20892
| | - Patricia W Lamb
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| |
Collapse
|
39
|
Rojas P, Piro O, Garcia ME. Biological Rhythms Generated by a Single Activator-Repressor Loop with Inhomogeneity and Diffusion. PHYSICAL REVIEW LETTERS 2024; 132:268401. [PMID: 38996302 DOI: 10.1103/physrevlett.132.268401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/19/2024] [Indexed: 07/14/2024]
Abstract
Common models of circadian rhythms are typically constructed as compartmental reactions of well-mixed biochemicals, incorporating a negative-feedback loop consisting of several intermediate reaction steps essentially required to produce oscillations. Spatial transport of each reactant is often represented as an extra compartmental reaction step. Contrary to this traditional understanding, in this Letter we demonstrate that a single activation-repression biochemical reaction pair is sufficient to generate sustained oscillations if the sites of both reactions are spatially separated and molecular transport is mediated by diffusion. Our proposed scenario represents the simplest configuration in terms of the participating chemical reactions and offers a conceptual basis for understanding biological oscillations and inspiring in vitro assays aimed at constructing minimal clocks.
Collapse
Affiliation(s)
- Pablo Rojas
- Theoretical Physics and Center for Interdisciplinary Nanostructure Science and Technology (CINSaT), University of Kassel, Kassel, Germany
| | - Oreste Piro
- Theoretical Physics and Center for Interdisciplinary Nanostructure Science and Technology (CINSaT), University of Kassel, Kassel, Germany
- Departament de Física, Universitat de les Illes Balears, Palma de Mallorca, Spain
- Institut Mediterrani d'Estudis Avançats, IMEDEA (CSIC-UIB), Esporles, Spain
| | - Martin E Garcia
- Theoretical Physics and Center for Interdisciplinary Nanostructure Science and Technology (CINSaT), University of Kassel, Kassel, Germany
| |
Collapse
|
40
|
Martin-Solana E, Carter SD, Donahue EK, Ning J, Glausier JR, Preisegger MA, Eisenman L, Joseph PN, Bouchet-Marquis C, Wu K, Mobini CL, Frantz AN, Puig S, Hampton CM, Kabbani N, Jensen GJ, Watkins SC, Deisseroth K, Fenno LE, Gold MS, Wills ZP, Burkewitz K, Das S, Freyberg Z. Ribosome-Associated Vesicles promote activity-dependent local translation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.598007. [PMID: 38895376 PMCID: PMC11185778 DOI: 10.1101/2024.06.07.598007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Local protein synthesis in axons and dendrites underpins synaptic plasticity. However, the composition of the protein synthesis machinery in distal neuronal processes and the mechanisms for its activity-driven deployment to local translation sites remain unclear. Here, we employed cryo-electron tomography, volume electron microscopy, and live-cell imaging to identify Ribosome-Associated Vesicles (RAVs) as a dynamic platform for moving ribosomes to distal processes. Stimulation via chemically-induced long-term potentiation causes RAV accumulation in distal sites to drive local translation. We also demonstrate activity-driven changes in RAV generation and dynamics in vivo, identifying tubular ER shaping proteins in RAV biogenesis. Together, our work identifies a mechanism for ribosomal delivery to distal sites in neurons to promote activity-dependent local translation.
Collapse
Affiliation(s)
- Eva Martin-Solana
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen D. Carter
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Eric K.F. Donahue
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Jiying Ning
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jill R. Glausier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Leanna Eisenman
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paul N. Joseph
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Ken Wu
- Materials and Structural Analysis, Thermo Fisher Scientific, Hillsboro, OR, USA
| | | | - Amber N. Frantz
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephanie Puig
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Cheri M. Hampton
- UES, Inc., Dayton, OH, USA
- Materials and Manufacturing Directorate, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, OH, USA
| | - Nadine Kabbani
- Interdisciplinary Program in Neuroscience, George Mason University, Fairfax, VA, USA
- School of Systems Biology, George Mason University, Fairfax, VA, USA
| | - Grant J. Jensen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Simon C. Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Lief E. Fenno
- Departments of Psychiatry and Neuroscience, University of Texas Austin Dell Medical School, Austin, TX, USA
| | - Michael S. Gold
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary P. Wills
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kristopher Burkewitz
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Sulagna Das
- Department of Cell Biology, Albert Einstein College of Medicine, NY
- Department of Cell Biology, Emory University, Atlanta, GA, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
41
|
Xiong GJ, Sheng ZH. Presynaptic perspective: Axonal transport defects in neurodevelopmental disorders. J Cell Biol 2024; 223:e202401145. [PMID: 38568173 PMCID: PMC10988239 DOI: 10.1083/jcb.202401145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Disruption of synapse assembly and maturation leads to a broad spectrum of neurodevelopmental disorders. Presynaptic proteins are largely synthesized in the soma, where they are packaged into precursor vesicles and transported into distal axons to ensure precise assembly and maintenance of presynapses. Due to their morphological features, neurons face challenges in the delivery of presynaptic cargos to nascent boutons. Thus, targeted axonal transport is vital to build functional synapses. A growing number of mutations in genes encoding the transport machinery have been linked to neurodevelopmental disorders. Emerging lines of evidence have started to uncover presynaptic mechanisms underlying axonal transport defects, thus broadening the view of neurodevelopmental disorders beyond postsynaptic mechanisms. In this review, we discuss presynaptic perspectives of neurodevelopmental disorders by focusing on impaired axonal transport and disturbed assembly and maintenance of presynapses. We also discuss potential strategies for restoring axonal transport as an early therapeutic intervention.
Collapse
Affiliation(s)
- Gui-Jing Xiong
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
42
|
De Pace R, Ghosh S, Ryan VH, Sohn M, Jarnik M, Rezvan Sangsari P, Morgan NY, Dale RK, Ward ME, Bonifacino JS. Messenger RNA transport on lysosomal vesicles maintains axonal mitochondrial homeostasis and prevents axonal degeneration. Nat Neurosci 2024; 27:1087-1102. [PMID: 38600167 PMCID: PMC11156585 DOI: 10.1038/s41593-024-01619-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 03/07/2024] [Indexed: 04/12/2024]
Abstract
In neurons, RNA granules are transported along the axon for local translation away from the soma. Recent studies indicate that some of this transport involves hitchhiking of RNA granules on lysosome-related vesicles. In the present study, we leveraged the ability to prevent transport of these vesicles into the axon by knockout of the lysosome-kinesin adaptor BLOC-one-related complex (BORC) to identify a subset of axonal mRNAs that depend on lysosome-related vesicles for transport. We found that BORC knockout causes depletion of a large group of axonal mRNAs mainly encoding ribosomal and mitochondrial/oxidative phosphorylation proteins. This depletion results in mitochondrial defects and eventually leads to axonal degeneration in human induced pluripotent stem cell (iPSC)-derived and mouse neurons. Pathway analyses of the depleted mRNAs revealed a mechanistic connection of BORC deficiency with common neurodegenerative disorders. These results demonstrate that mRNA transport on lysosome-related vesicles is critical for the maintenance of axonal homeostasis and that its failure causes axonal degeneration.
Collapse
Affiliation(s)
- Raffaella De Pace
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Saikat Ghosh
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Veronica H Ryan
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mira Sohn
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michal Jarnik
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Paniz Rezvan Sangsari
- Biomedical Engineering and Physical Science Shared Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Nicole Y Morgan
- Biomedical Engineering and Physical Science Shared Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Ryan K Dale
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Michael E Ward
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Juan S Bonifacino
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
43
|
Dashkova AS, Kovalev VI, Chaplygina AV, Zhdanova DY, Bobkova NV. Unique Properties of Synaptosomes and Prospects for Their Use for the Treatment of Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1031-1044. [PMID: 38981699 DOI: 10.1134/s0006297924060051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative condition affecting millions worldwide. Prevalence of AD correlates with increased life expectancy and aging population in the developed countries. Considering that AD is a multifactorial disease involving various pathological processes such as synaptic dysfunction, neuroinflammation, oxidative stress, and improper protein folding, a comprehensive approach targeting multiple pathways may prove effective in slowing the disease progression. Cellular therapy and its further development in the form of cell vesicle and particularly mitochondrial transplantation represent promising approaches for treating neurodegeneration. The use of synaptosomes, due to uniqueness of their contents, could mark a new stage in the development of comprehensive therapies for neurodegenerative diseases, particularly AD. Synaptosomes contain unique memory mitochondria, which differ not only in size but also in functionality compared to the mitochondria in the neuronal soma. These synaptosomal mitochondria actively participate in cellular communication and signal transmission within synapses. Synaptosomes also contain other elements such as their own protein synthesis machinery, synaptic vesicles with neurotransmitters, synaptic adhesion molecules, and microRNAs - all crucial for synaptic transmission and, consequently, cognitive processes. Complex molecular ensemble ensures maintenance of the synaptic autonomy of mitochondria. Additionally, synaptosomes, with their affinity for neurons, can serve as an optimal platform for targeted drug delivery to nerve cells. This review discusses unique composition of synaptosomes, their capabilities and advantages, as well as limitations of their suggested use as therapeutic agents for treating neurodegenerative pathologies, particularly AD.
Collapse
Affiliation(s)
- Alla S Dashkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vladimir I Kovalev
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Alina V Chaplygina
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Daria Yu Zhdanova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Natalia V Bobkova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
44
|
Hermanova Z, Valihrach L, Kriska J, Maheta M, Tureckova J, Kubista M, Anderova M. The deletion of AQP4 and TRPV4 affects astrocyte swelling/volume recovery in response to ischemia-mimicking pathologies. Front Cell Neurosci 2024; 18:1393751. [PMID: 38818517 PMCID: PMC11138210 DOI: 10.3389/fncel.2024.1393751] [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: 02/29/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Astrocytic Transient receptor potential vanilloid 4 (TRPV4) channels, together with Aquaporin 4 (AQP4), are suspected to be the key players in cellular volume regulation, and therefore may affect the development and severity of cerebral edema during ischemia. In this study, we examined astrocytic swelling/volume recovery in mice with TRPV4 and/or AQP4 deletion in response to in vitro ischemic conditions, to determine how the deletion of these channels can affect the development of cerebral edema. Methods We used three models of ischemia-related pathological conditions: hypoosmotic stress, hyperkalemia, and oxygenglucose deprivation (OGD), and observed their effect on astrocyte volume changes in acute brain slices of Aqp4-/-, Trpv4-/- and double knockouts. In addition, we employed single-cell RT-qPCR to assess the effect of TRPV4 and AQP4 deletion on the expression of other ion channels and transporters involved in the homeostatic functioning of astrocytes. Results Quantification of astrocyte volume changes during OGD revealed that the deletion of AQP4 reduces astrocyte swelling, while simultaneous deletion of both AQP4 and TRPV4 leads to a disruption of astrocyte volume recovery during the subsequent washout. Of note, astrocyte exposure to hypoosmotic stress or hyperkalemia revealed no differences in astrocyte swelling in the absence of AQP4, TRPV4, or both channels. Moreover, under ischemia-mimicking conditions, we identified two distinct subpopulations of astrocytes with low and high volumetric responses (LRA and HRA), and their analyses revealed that mainly HRA are affected by the deletion of AQP4, TRPV4, or both channels. Furthermore, gene expression analysis revealed reduced expression of the ion transporters KCC1 and ClC2 as well as the receptors GABAB and NMDA in Trpv4-/- mice. The deletion of AQP4 instead caused reduced expression of the serine/cysteine peptidase inhibitor Serpina3n. Discussion Thus, we showed that in AQP4 or TRPV4 knockouts, not only the specific function of these channels is affected, but also the expression of other proteins, which may modulate the ischemic cascade and thus influence the final impact of ischemia.
Collapse
Affiliation(s)
- Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lukas Valihrach
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mansi Maheta
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Vestec, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine CAS, Prague, Czechia
| |
Collapse
|
45
|
Fansler MM, Mitschka S, Mayr C. Quantifying 3'UTR length from scRNA-seq data reveals changes independent of gene expression. Nat Commun 2024; 15:4050. [PMID: 38744866 PMCID: PMC11094166 DOI: 10.1038/s41467-024-48254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 04/22/2024] [Indexed: 05/16/2024] Open
Abstract
Although more than half of all genes generate transcripts that differ in 3'UTR length, current analysis pipelines only quantify the amount but not the length of mRNA transcripts. 3'UTR length is determined by 3' end cleavage sites (CS). We map CS in more than 200 primary human and mouse cell types and increase CS annotations relative to the GENCODE database by 40%. Approximately half of all CS are used in few cell types, revealing that most genes only have one or two major 3' ends. We incorporate the CS annotations into a computational pipeline, called scUTRquant, for rapid, accurate, and simultaneous quantification of gene and 3'UTR isoform expression from single-cell RNA sequencing (scRNA-seq) data. When applying scUTRquant to data from 474 cell types and 2134 perturbations, we discover extensive 3'UTR length changes across cell types that are as widespread and coordinately regulated as gene expression changes but affect mostly different genes. Our data indicate that mRNA abundance and mRNA length are two largely independent axes of gene regulation that together determine the amount and spatial organization of protein synthesis.
Collapse
Affiliation(s)
- Mervin M Fansler
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Graduate College, New York, NY, 10021, USA
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sibylle Mitschka
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Christine Mayr
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Graduate College, New York, NY, 10021, USA.
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
46
|
Wang L, Liu X, Zhu C, Wu S, Li Z, Jing L, Zhang Z, Jing Y, Wang Y. Environmental enrichment alleviates hyperalgesia by modulating central sensitization in a nitroglycerin-induced chronic migraine model of mice. J Headache Pain 2024; 25:74. [PMID: 38724948 PMCID: PMC11083806 DOI: 10.1186/s10194-024-01779-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Chronic migraine (CM) is a debilitating neurofunctional disorder primarily affecting females, characterized by central sensitization. Central sensitization refers to the enhanced response to sensory stimulation, which involves changes in neuronal excitability, synaptic plasticity, and neurotransmitter release. Environmental enrichment (EE) can increase the movement, exploration, socialization and other behaviors of mice. EE has shown promising effects in various neurological disorders, but its impact on CM and the underlying mechanism remains poorly understood. Therefore, the purpose of this study was to determine whether EE has the potential to serve as a cost-effective intervention strategy for CM. METHODS A mouse CM model was successfully established by repeated administration of nitroglycerin (NTG). We selected adult female mice around 8 weeks old, exposed them to EE for 2 months, and then induced the CM model. Nociceptive threshold tests were measured using Von Frey filaments and a hot plate. The expression of c-Fos, calcitonin gene-related peptide (CGRP) and inflammatory response were measured using WB and immunofluorescence to evaluate central sensitization. RNA sequencing was used to find differentially expressed genes and signaling pathways. Finally, the expression of the target differential gene was investigated. RESULTS Repeated administration of NTG can induce hyperalgesia in female mice and increase the expression of c-Fos and CGRP in the trigeminal nucleus caudalis (TNC). Early exposure of mice to EE reduced NTG-induced hyperalgesia in CM mice. WB and immunofluorescence revealed that EE inhibited the overexpression of c-Fos and CGRP in the TNC of CM mice and alleviated the inflammatory response of microglia activation. RNA sequencing analysis identified that several central sensitization-related signaling pathways were altered by EE. VGluT1, a key gene involved in behavior, internal stimulus response, and ion channel activity, was found to be downregulated in mice exposed to EE. CONCLUSION EE can significantly ameliorate hyperalgesia in the NTG-induced CM model. The mechanisms may be to modulate central sensitization by reducing the expression of CGRP, attenuating the inflammatory response, and downregulating the expression of VGluT1, etc., suggesting that EE can serve as an effective preventive strategy for CM.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, The Second Hospital of Lanzhou University, Gate, No. 82 Linxia Road, Chengguan District, Lanzhou, 730000, China
| | - Xiaoming Liu
- Institute of Epidemiology and Statistics, School of Public Health, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou, 730000, China
| | - Chenlu Zhu
- Department of Neurology, The Second Hospital of Lanzhou University, Gate, No. 82 Linxia Road, Chengguan District, Lanzhou, 730000, China
| | - Shouyi Wu
- Department of Neurology, The Second Hospital of Lanzhou University, Gate, No. 82 Linxia Road, Chengguan District, Lanzhou, 730000, China
| | - Zhilei Li
- Department of Neurology, The Second Hospital of Lanzhou University, Gate, No. 82 Linxia Road, Chengguan District, Lanzhou, 730000, China
| | - Lipeng Jing
- Institute of Epidemiology and Statistics, School of Public Health, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou, 730000, China
| | - Zhenchang Zhang
- Department of Neurology, The Second Hospital of Lanzhou University, Gate, No. 82 Linxia Road, Chengguan District, Lanzhou, 730000, China
| | - Yuhong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, No. 222 Tianshui South Road, Chengguan District, Lanzhou, 730000, China.
| | - Yonggang Wang
- Department of Neurology, The Second Hospital of Lanzhou University, Gate, No. 82 Linxia Road, Chengguan District, Lanzhou, 730000, China.
- Headache Center, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
47
|
Ramnauth AD, Tippani M, Divecha HR, Papariello AR, Miller RA, Nelson ED, Pattie EA, Kleinman JE, Maynard KR, Collado-Torres L, Hyde TM, Martinowich K, Hicks SC, Page SC. Spatiotemporal analysis of gene expression in the human dentate gyrus reveals age-associated changes in cellular maturation and neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.20.567883. [PMID: 38045413 PMCID: PMC10690172 DOI: 10.1101/2023.11.20.567883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The dentate gyrus of the hippocampus is important for many cognitive functions, including learning, memory, and mood. Here, we investigated age-associated changes in transcriptome-wide spatial gene expression in the human dentate gyrus across the lifespan. Genes associated with neurogenesis and the extracellular matrix were enriched in infants, while gene markers of inhibitory neurons and cell proliferation showed increases and decreases in post-infancy, respectively. While we did not find evidence for neural proliferation post-infancy, we did identify molecular signatures supporting protracted maturation of granule cells. We also identified a wide-spread hippocampal aging signature and an age-associated increase in genes related to neuroinflammation. Our findings suggest major changes to the putative neurogenic niche after infancy and identify molecular foci of brain aging in glial and neuropil enriched tissue.
Collapse
|
48
|
Hayden AN, Brandel KL, Merlau PR, Vijayakumar P, Leptich EJ, Pietryk EW, Gaytan ES, Ni CW, Chao HT, Rosenfeld JA, Arey RN. Behavioral screening of conserved RNA-binding proteins reveals CEY-1/YBX RNA-binding protein dysfunction leads to impairments in memory and cognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574402. [PMID: 38260399 PMCID: PMC10802296 DOI: 10.1101/2024.01.05.574402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to increase memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.
Collapse
Affiliation(s)
- Ashley N Hayden
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Katie L Brandel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Paul R Merlau
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | | | - Emily J Leptich
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Edward W Pietryk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
| | - Elizabeth S Gaytan
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Postbaccalaureate Research Education Program, Baylor College of Medicine, Houston, TX, 77030
| | - Connie W Ni
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Neuroscience, Rice University, Houston, TX 77005
| | - Hsiao-Tuan Chao
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, 77030
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, TX, 77030
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Baylor Genetics Laboratories, Houston, TX 77021
| | - Rachel N Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
49
|
Rey F, Esposito L, Maghraby E, Mauri A, Berardo C, Bonaventura E, Tonduti D, Carelli S, Cereda C. Role of epigenetics and alterations in RNA metabolism in leukodystrophies. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1854. [PMID: 38831585 DOI: 10.1002/wrna.1854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 06/05/2024]
Abstract
Leukodystrophies are a class of rare heterogeneous disorders which affect the white matter of the brain, ultimately leading to a disruption in brain development and a damaging effect on cognitive, motor and social-communicative development. These disorders present a great clinical heterogeneity, along with a phenotypic overlap and this could be partially due to contributions from environmental stimuli. It is in this context that there is a great need to investigate what other factors may contribute to both disease insurgence and phenotypical heterogeneity, and novel evidence are raising the attention toward the study of epigenetics and transcription mechanisms that can influence the disease phenotype beyond genetics. Modulation in the epigenetics machinery including histone modifications, DNA methylation and non-coding RNAs dysregulation, could be crucial players in the development of these disorders, and moreover an aberrant RNA maturation process has been linked to leukodystrophies. Here, we provide an overview of these mechanisms hoping to supply a closer step toward the analysis of leukodystrophies not only as genetically determined but also with an added level of complexity where epigenetic dysregulation is of key relevance. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNA RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Federica Rey
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Sciences, University of Milano, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
| | - Letizia Esposito
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Sciences, University of Milano, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
| | - Erika Maghraby
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
- Department of Biology and Biotechnology "L. Spallanzani" (DBB), University of Pavia, Pavia, Italy
| | - Alessia Mauri
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Sciences, University of Milano, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
| | - Clarissa Berardo
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Sciences, University of Milano, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
| | - Eleonora Bonaventura
- Unit of Pediatric Neurology, COALA Center for Diagnosis and Treatment of Leukodystrophies, V. Buzzi Children's Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Davide Tonduti
- Unit of Pediatric Neurology, COALA Center for Diagnosis and Treatment of Leukodystrophies, V. Buzzi Children's Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Stephana Carelli
- Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi," Department of Biomedical and Clinical Sciences, University of Milano, Milan, Italy
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
| | - Cristina Cereda
- Center of Functional Genomics and Rare Diseases, Department of Pediatrics, Buzzi Children's Hospital, Milan, Italy
| |
Collapse
|
50
|
Park I, Kim HJ, Shin J, Jung YJ, Lee D, Lim J, Park JM, Park JW, Kim J. AFM Imaging Reveals MicroRNA-132 to be a Positive Regulator of Synaptic Functions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306630. [PMID: 38493494 PMCID: PMC11077659 DOI: 10.1002/advs.202306630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/17/2024] [Indexed: 03/19/2024]
Abstract
The modification of synaptic and neural connections in adults, including the formation and removal of synapses, depends on activity-dependent synaptic and structural plasticity. MicroRNAs (miRNAs) play crucial roles in regulating these changes by targeting specific genes and regulating their expression. The fact that somatic and dendritic activity in neurons often occurs asynchronously highlights the need for spatial and dynamic regulation of protein synthesis in specific milieu and cellular loci. MicroRNAs, which can show distinct patterns of enrichment, help to establish the localized distribution of plasticity-related proteins. The recent study using atomic force microscopy (AFM)-based nanoscale imaging reveals that the abundance of miRNA(miR)-134 is inversely correlated with the functional activity of dendritic spine structures. However, the miRNAs that are selectively upregulated in potentiated synapses, and which can thereby support prospective changes in synaptic efficacy, remain largely unknown. Using AFM force imaging, significant increases in miR-132 in the dendritic regions abutting functionally-active spines is discovered. This study provides evidence for miR-132 as a novel positive miRNA regulator residing in dendritic shafts, and also suggests that activity-dependent miRNAs localized in distinct sub-compartments of neurons play bi-directional roles in controlling synaptic transmission and synaptic plasticity.
Collapse
Affiliation(s)
- Ikbum Park
- Technical Support Center for Chemical IndustryKorea Research Institute of Chemical Technology (KRICT)Ulsan44412Republic of Korea
| | - Hyun Jin Kim
- Department of Life SciencesPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Juyoung Shin
- Department of Life SciencesPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Yu Jin Jung
- Center for Specialty ChemicalsKorea Research Institute of Chemical Technology (KRICT)Ulsan44412Republic of Korea
| | - Donggyu Lee
- Division of Electronics and Information SystemDaegu Gyeongbuk Institute of Science and Technology (DGIST)Daegu42988Republic of Korea
| | - Ji‐seon Lim
- Department of ChemistryPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Jong Mok Park
- Technical Support Center for Chemical IndustryKorea Research Institute of Chemical Technology (KRICT)Ulsan44412Republic of Korea
| | - Joon Won Park
- Department of ChemistryPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
| | - Joung‐Hun Kim
- Department of Life SciencesPohang University of Science and Technology (POSTECH)Pohang37673Republic of Korea
- Institute of Convergence ScienceYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|