1
|
Farkas D, Dobránszki J. Vegetal memory through the lens of transcriptomic changes - recent progress and future practical prospects for exploiting plant transcriptional memory. PLANT SIGNALING & BEHAVIOR 2024; 19:2383515. [PMID: 39077764 PMCID: PMC11290777 DOI: 10.1080/15592324.2024.2383515] [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: 06/11/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/31/2024]
Abstract
Plant memory plays an important role in the efficient and rapid acclimation to a swiftly changing environment. In addition, since plant memory can be inherited, it is also of adaptive and evolutionary importance. The ability of a plant to store, retain, retrieve and delete information on acquired experience is based on cellular, biochemical and molecular networks in the plants. This review offers an up-to-date overview on the formation, types, checkpoints of plant memory based on our current knowledge and focusing on its transcriptional aspects, the transcriptional memory. Roles of long and small non-coding RNAs are summarized in the regulation, formation and the cooperation between the different layers of the plant memory, i.e. in the establishment of epigenetic changes associated with memory formation in plants. The RNA interference mechanisms at the RNA and DNA level and the interplays between them are also presented. Furthermore, this review gives an insight of how exploitation of plant transcriptional memory may provide new opportunities for elaborating promising cost-efficient, and effective strategies to cope with the ever-changing environmental perturbations, caused by climate change. The potentials of plant memory-based methods, such as crop priming, cross acclimatization, memory modification by miRNAs and associative use of plant memory, in the future's agriculture are also discussed.
Collapse
Affiliation(s)
- Dóra Farkas
- Centre for Agricultural Genomics and Biotechnology, Faculty of the Agricultural and Food Science and Environmental Management, University of Debrecen, Nyíregyháza, Hungary
| | - Judit Dobránszki
- Centre for Agricultural Genomics and Biotechnology, Faculty of the Agricultural and Food Science and Environmental Management, University of Debrecen, Nyíregyháza, Hungary
| |
Collapse
|
2
|
Dos Santos GRO, Cararo-Lopes MM, Possebom IR, de Sá Lima L, Scavone C, Kawamoto EM. Sex-dependent changes in AMPAR expression and Na, K-ATPase activity in the cerebellum and hippocampus of α-Klotho-Hypomorphic mice. Neuropharmacology 2024; 258:110097. [PMID: 39094831 DOI: 10.1016/j.neuropharm.2024.110097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Aging is characterized by a functional decline in several physiological systems. α-Klotho-hypomorphic mice (Kl-/-) exhibit accelerated aging and cognitive decline. We evaluated whether male and female α-Klotho-hypomorphic mice show changes in the expression of synaptic proteins, N-methyl-d-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, postsynaptic density protein 95 (PSD-95), synaptophysin and synapsin, and the activity of Na+, K+-ATPase (NaK) isoforms in the cerebellum and hippocampus. In this study, we demonstrated that in the cerebellum, Kl-/- male mice have reduced expression of GluA1 (AMPA) compared to wild-type (Kl+/+) males and Kl-/- females. Also, Kl-/- male and female mice show reduced ɑ2/ɑ3-NaK and Mg2+-ATPase activities in the cerebellum, respectively, and sex-based differences in NaK and Mg2+-ATPase activities in both the regions. Our findings suggest that α-Klotho could influence the expression of AMPAR and the activity of NaK isoforms in the cerebellum in a sex-dependent manner, and these changes may contribute, in part, to cognitive decline.
Collapse
Affiliation(s)
| | - Marina Minto Cararo-Lopes
- Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, 79070-900, Campo Grande, MS, Brazil
| | - Isabela Ribeiro Possebom
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Larissa de Sá Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Elisa Mitiko Kawamoto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil.
| |
Collapse
|
3
|
Machado JPD, de Almeida V, Zuardi AW, Hallak JEC, Crippa JA, Vieira AS. Cannabidiol modulates hippocampal genes involved in mitochondrial function, ribosome biogenesis, synapse organization, and chromatin modifications. Acta Neuropsychiatr 2024; 36:330-336. [PMID: 38528655 DOI: 10.1017/neu.2024.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
BACKGROUND Cannabidiol (CBD) is one of the main cannabinoids present in Cannabis sativa female flowers. Previous investigation has already provided insights into the CBD molecular mechanism; however, there is no transcriptome data for CBD effects on hippocampal subfields. Here, we investigate transcriptomic changes in dorsal and ventral CA1 of adult mice hippocampus after 100 mg/kg of CBD administration (i.p.) for one or seven consecutive days. METHODS C57BL/6JUnib mice were treated with either vehicle or CBD for 1 or 7 days. The collected brains were sectioned, and the hippocampal sub-regions were laser microdissected for RNA-Seq analysis. RESULTS The transcriptome analysis following 7 days of CBD administration indicates the differential expression of 1559 genes in dCA1 and 2924 genes in vCA1. Furthermore, GO/KEGG analysis identified 88 significantly enriched biological process and 26 significantly enriched pathways for dCBD7, whereas vCBD7 revealed 128 enriched BPs and 24 pathways. CONCLUSION This dataset indicates a widespread decrease of electron transport chain and ribosome biogenesis transcripts in CA1, while chromatin modifications and synapse organization transcripts were increased following CBD administration for 7 days.
Collapse
Affiliation(s)
- João P D Machado
- Laboratory of Electrophysiology, Neurobiology and Behaviour, Dept Functional and Structural Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Valéria de Almeida
- Laboratory of Neuroproteomics,, Dept Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinsas, São Paulo, Brazil
| | - Antonio W Zuardi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- National Institute for Science and Technology - Translational Medicine, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jaime E C Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- National Institute for Science and Technology - Translational Medicine, Rio de Janeiro, Rio de Janeiro, Brazil
| | - José A Crippa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- National Institute for Science and Technology - Translational Medicine, Rio de Janeiro, Rio de Janeiro, Brazil
| | - André S Vieira
- Laboratory of Electrophysiology, Neurobiology and Behaviour, Dept Functional and Structural Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| |
Collapse
|
4
|
Zuniga G, Katsumura S, De Mange J, Ramirez P, Atrian F, Morita M, Frost B. Pathogenic tau induces an adaptive elevation in mRNA translation rate at early stages of disease. Aging Cell 2024; 23:e14245. [PMID: 38932463 PMCID: PMC11464109 DOI: 10.1111/acel.14245] [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: 03/06/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Alterations in the rate and accuracy of messenger RNA (mRNA) translation are associated with aging and several neurodegenerative disorders, including Alzheimer's disease and related tauopathies. We previously reported that error-containing RNA that are normally cleared via nonsense-mediated mRNA decay (NMD), a key RNA surveillance mechanism, are translated in the adult brain of a Drosophila model of tauopathy. In the current study, we find that newly-synthesized peptides and translation machinery accumulate within nuclear envelope invaginations that occur as a consequence of tau pathology, and that the rate of mRNA translation is globally elevated in early stages of disease in adult brains of Drosophila models of tauopathy. Polysome profiling from adult heads of tau transgenic Drosophila reveals the preferential translation of specific mRNA that have been previously linked to neurodegeneration. Unexpectedly, we find that panneuronal elevation of NMD further elevates the global translation rate in tau transgenic Drosophila, as does treatment with rapamycin. As NMD activation and rapamycin both suppress tau-induced neurodegeneration, their shared effect on translation suggests that elevated rates of mRNA translation are an early adaptive mechanism to limit neurodegeneration. Our work provides compelling evidence that tau-induced deficits in NMD reshape the tau translatome by increasing translation of RNA that are normally repressed in healthy cells.
Collapse
Affiliation(s)
- Gabrielle Zuniga
- Barshop Institute for Longevity and Aging StudiesSan AntonioTexasUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesSan AntonioTexasUSA
- Department of Cell Systems and AnatomyUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Sakie Katsumura
- Barshop Institute for Longevity and Aging StudiesSan AntonioTexasUSA
- Department of Molecular MedicineUniversity of Texas Health San AntonioSan AntonioTexasUSA
- Premium Research Institute for Human Metaverse Medicine (WPI‐PRIMe)Osaka UniversitySuitaOsakaJapan
| | - Jasmine De Mange
- Barshop Institute for Longevity and Aging StudiesSan AntonioTexasUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesSan AntonioTexasUSA
- Department of Cell Systems and AnatomyUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Paulino Ramirez
- Barshop Institute for Longevity and Aging StudiesSan AntonioTexasUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesSan AntonioTexasUSA
- Department of Cell Systems and AnatomyUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Farzaneh Atrian
- Barshop Institute for Longevity and Aging StudiesSan AntonioTexasUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesSan AntonioTexasUSA
- Department of Cell Systems and AnatomyUniversity of Texas Health San AntonioSan AntonioTexasUSA
| | - Masahiro Morita
- Barshop Institute for Longevity and Aging StudiesSan AntonioTexasUSA
- Department of Molecular MedicineUniversity of Texas Health San AntonioSan AntonioTexasUSA
- Premium Research Institute for Human Metaverse Medicine (WPI‐PRIMe)Osaka UniversitySuitaOsakaJapan
| | - Bess Frost
- Barshop Institute for Longevity and Aging StudiesSan AntonioTexasUSA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesSan AntonioTexasUSA
- Department of Cell Systems and AnatomyUniversity of Texas Health San AntonioSan AntonioTexasUSA
| |
Collapse
|
5
|
Kasegawa C, Itaguchi Y, Yamawaki Y, Miki M, Hayashi M, Miyazaki M. Effects of within-day intervals on adaptation to visually induced motion sickness in a virtual-reality motorcycling simulator. Sci Rep 2024; 14:21302. [PMID: 39307847 PMCID: PMC11417106 DOI: 10.1038/s41598-024-71526-9] [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: 10/11/2023] [Accepted: 08/28/2024] [Indexed: 09/26/2024] Open
Abstract
This study investigated the effects of the time interval between virtual reality (VR) sessions on visually induced motion sickness (VIMS) reduction to better understand adaptation to and recovery from a nauseating VR experience. The participants experienced two 6-min VR sessions of a first-person motorcycle ride through a head-mounted display with (1) a 6-min interval, (2) an interval until the VIMS score reached zero, and (3) a 60-min interval. The results showed that for each condition, VIMS in the second session was aggravated, unchanged, or attenuated, respectively, indicating that additional resting time was necessary for VIMS adaptation. This study suggests that a certain type of multisensory learning attenuates VIMS symptoms within a relatively short time, requiring at least 20 min of additional resting time after subjective recovery from VIMS symptoms. This finding has important implications for reducing the time interval between repeated challenges when adapting to nauseating stimuli during VR experiences.
Collapse
Affiliation(s)
- Chihiro Kasegawa
- Department of Informatics, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, 432-8011, Japan
| | | | - Yumi Yamawaki
- Department of Informatics, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, 432-8011, Japan
| | | | - Masami Hayashi
- Department of Informatics, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, 432-8011, Japan
| | - Makoto Miyazaki
- Department of Informatics, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, 432-8011, Japan.
- Faculty of Informatics, Shizuoka University, Hamamatsu, 432-8011, Japan.
| |
Collapse
|
6
|
Welle TM, Rajgor D, Kareemo DJ, Garcia JD, Zych SM, Wolfe SE, Gookin SE, Martinez TP, Dell'Acqua ML, Ford CP, Kennedy MJ, Smith KR. miRNA-mediated control of gephyrin synthesis drives sustained inhibitory synaptic plasticity. EMBO Rep 2024:10.1038/s44319-024-00253-z. [PMID: 39294503 DOI: 10.1038/s44319-024-00253-z] [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: 12/15/2023] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024] Open
Abstract
Activity-dependent protein synthesis is crucial for long-lasting forms of synaptic plasticity. However, our understanding of translational mechanisms controlling GABAergic synapses is limited. One distinct form of inhibitory long-term potentiation (iLTP) enhances postsynaptic clusters of GABAARs and the primary inhibitory scaffold, gephyrin, to promote sustained synaptic strengthening. While we previously found that persistent iLTP requires mRNA translation, the mechanisms controlling plasticity-induced gephyrin translation remain unknown. We identify miR153 as a novel regulator of Gphn mRNA translation which controls gephyrin protein levels and synaptic clustering, ultimately impacting inhibitory synaptic structure and function. iLTP induction downregulates miR153, reversing its translational suppression of Gphn mRNA and promoting de novo gephyrin protein synthesis and synaptic clustering during iLTP. Finally, we find that reduced miR153 expression during iLTP is driven by an excitation-transcription coupling pathway involving calcineurin, NFAT and HDACs, which also controls the miRNA-dependent upregulation of GABAARs. Together, we delineate a miRNA-dependent post-transcriptional mechanism that controls the expression of the key synaptic scaffold, gephyrin, and may converge with parallel miRNA pathways to coordinate gene upregulation to maintain inhibitory synaptic plasticity.
Collapse
Affiliation(s)
- Theresa M Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Dipen Rajgor
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Dean J Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Joshua D Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sarah M Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sarah E Wolfe
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Sara E Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Tyler P Martinez
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO, 80045, USA.
| |
Collapse
|
7
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2024:10.1038/s41380-024-02725-z. [PMID: 39223276 DOI: 10.1038/s41380-024-02725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
8
|
N. Costa M, Goto-Silva L, M. Nascimento J, Domith I, Karmirian K, Feilding A, Trindade P, Martins-de-Souza D, K. Rehen S. LSD Modulates Proteins Involved in Cell Proteostasis, Energy Metabolism and Neuroplasticity in Human Cerebral Organoids. ACS OMEGA 2024; 9:36553-36568. [PMID: 39220485 PMCID: PMC11360045 DOI: 10.1021/acsomega.4c04712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Proteomic analysis of human cerebral organoids may reveal how psychedelics regulate biological processes, shedding light on drug-induced changes in the brain. This study elucidates the proteomic alterations induced by lysergic acid diethylamide (LSD) in human cerebral organoids. By employing high-resolution mass spectrometry-based proteomics, we quantitatively analyzed the differential abundance of proteins in cerebral organoids exposed to LSD. Our findings indicate changes in proteostasis, energy metabolism, and neuroplasticity-related pathways. Specifically, LSD exposure led to alterations in protein synthesis, folding, autophagy, and proteasomal degradation, suggesting a complex interplay in the regulation of neural cell function. Additionally, we observed modulation in glycolysis and oxidative phosphorylation, crucial for cellular energy management and synaptic function. In support of the proteomic data, complementary experiments demonstrated LSD's potential to enhance neurite outgrowth in vitro, confirming its impact on neuroplasticity. Collectively, our results provide a comprehensive insight into the molecular mechanisms through which LSD may affect neuroplasticity and potentially contribute to therapeutic effects for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Marcelo N. Costa
- D’Or
Institute for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio de Janeiro 22281-100, RJ, Brazil
- Department
of Genetics, Institute of Biology, Federal
University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373 - Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| | - Livia Goto-Silva
- D’Or
Institute for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio de Janeiro 22281-100, RJ, Brazil
| | - Juliana M. Nascimento
- D’Or
Institute for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio de Janeiro 22281-100, RJ, Brazil
- Department
of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas, Rua Monteiro Lobato, 255 - Cidade
Universitária Zeferino Vaz, Campinas 13083-862, SP, Brazil
| | - Ivan Domith
- D’Or
Institute for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio de Janeiro 22281-100, RJ, Brazil
- Pioneer
Science Initiative, D’Or Institute
for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio
de Janeiro22281-100, RJ, Brazil
| | - Karina Karmirian
- D’Or
Institute for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio de Janeiro 22281-100, RJ, Brazil
| | - Amanda Feilding
- Beckley
Foundation, Beckley Park, Oxford OX3
9SY, United Kingdom
| | - Pablo Trindade
- Department
of Clinical and Toxicological Analysis (DACT), College of Pharmacy, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373
- Cidade Universitária, Rio de Janeiro 21941-853, RJ, Brazil
| | - Daniel Martins-de-Souza
- D’Or
Institute for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio de Janeiro 22281-100, RJ, Brazil
- Laboratory
of Neuroproteomics, Department of Biochemistry and Tissue Biology,
Institute of Biology, State University of
Campinas, Rua Monteiro
Lobato, 255 - Cidade Universitária Zeferino Vaz, Campinas 13083-862, SP, Brazil
- Experimental
Medicine Research Cluster (EMRC), State
University of Campinas, Rua Monteiro Lobato, 255 - Cidade Universitária Zeferino Vaz, Campinas 13083-862, SP, Brazil
| | - Stevens K. Rehen
- D’Or
Institute for Research and Education, Rua Diniz Cordeiro, 30−Botafogo, Rio de Janeiro 22281-100, RJ, Brazil
- Department
of Genetics, Institute of Biology, Federal
University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373 - Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
9
|
Hooshmandi M, Wong C, Lister KC, Brown N, Cai W, Ho-Tieng D, Stecum P, Backman T, Kostantin E, Khoutorsky A. Protocol for measuring protein synthesis in specific cell types in the mouse brain using in vivo non-canonical amino acid tagging. STAR Protoc 2024; 5:102775. [PMID: 38085640 PMCID: PMC10783633 DOI: 10.1016/j.xpro.2023.102775] [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] [Received: 08/07/2023] [Revised: 10/09/2023] [Accepted: 11/29/2023] [Indexed: 01/14/2024] Open
Abstract
The fluorescent non-canonical amino acid tagging (FUNCAT) technique has been used to visualize newly synthesized proteins in cell lines and tissues. Here, we present a protocol for measuring protein synthesis in specific cell types in the mouse brain using in vivo FUNCAT. We describe steps for metabolically labeling newly synthesized proteins with azidohomoalanine, which introduces an azide group into the polypeptide. We then detail procedures for binding a fluorophore-conjugated alkyne to the azide group to allow its visualization. For complete details on the use and execution of this protocol, please refer to tom Dieck et al. (2012)1 and Hooshmandi et al. (2023).2.
Collapse
Affiliation(s)
- Mehdi Hooshmandi
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada.
| | - Calvin Wong
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Kevin C Lister
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Nicole Brown
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Weihua Cai
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - David Ho-Tieng
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Patricia Stecum
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Thomas Backman
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - Elie Kostantin
- Clinical Department of Laboratory Medicine, Cite-de-la-Sante Hospital, Optilab LLL and University of Montreal, Montreal, QC, Canada
| | - Arkady Khoutorsky
- Department of Anesthesia and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada.
| |
Collapse
|
10
|
Garcia-Pardo J, Ventura S. Cryo-EM structures of functional and pathological amyloid ribonucleoprotein assemblies. Trends Biochem Sci 2024; 49:119-133. [PMID: 37926650 DOI: 10.1016/j.tibs.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023]
Abstract
Amyloids are implicated in neurodegenerative and systemic diseases, yet they serve important functional roles in numerous organisms. Heterogeneous nuclear ribonucleoproteins (hnRNPs) represent a large family of RNA-binding proteins (RBPs) that control central events of RNA biogenesis in normal and diseased cellular conditions. Many of these proteins contain prion-like sequences of low complexity, which not only assemble into functional fibrils in response to cellular cues but can also lead to disease when missense mutations arise in their sequences. Recent advances in cryo-electron microscopy (cryo-EM) have provided unprecedented high-resolution structural insights into diverse amyloid assemblies formed by hnRNPs and structurally related RBPs, including TAR DNA-binding protein 43 (TDP-43), Fused in Sarcoma (FUS), Orb2, hnRNPA1, hnRNPA2, and hnRNPDL-2. This review provides a comprehensive overview of these structures and explores their functional and pathological implications.
Collapse
Affiliation(s)
- Javier Garcia-Pardo
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain.
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193, Bellaterra, Barcelona, Spain.
| |
Collapse
|
11
|
Welle TM, Rajgor D, Garcia JD, Kareemo D, Zych SM, Gookin SE, Martinez TP, Dell’Acqua ML, Ford CP, Kennedy MJ, Smith KR. miRNA-mediated control of gephyrin synthesis drives sustained inhibitory synaptic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.570420. [PMID: 38168421 PMCID: PMC10760056 DOI: 10.1101/2023.12.12.570420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Activity-dependent protein synthesis is crucial for many long-lasting forms of synaptic plasticity. However, our understanding of the translational mechanisms controlling inhibitory synapses is limited. One distinct form of inhibitory long-term potentiation (iLTP) enhances postsynaptic clusters of GABAARs and the primary inhibitory scaffold, gephyrin, to promote sustained synaptic strengthening. While we previously found that persistent iLTP requires mRNA translation, the precise mechanisms controlling gephyrin translation during this process remain unknown. Here, we identify miR153 as a novel regulator of Gphn mRNA translation which controls gephyrin protein levels and synaptic clustering, ultimately impacting GABAergic synaptic structure and function. We find that iLTP induction downregulates miR153, reversing its translational suppression of Gphn mRNA and allowing for increased de novo gephyrin protein synthesis and synaptic clustering during iLTP. Finally, we find that reduced miR153 expression during iLTP is driven by an excitation-transcription coupling pathway involving calcineurin, NFAT and HDACs, which also controls the miRNA-dependent upregulation of GABAARs. Overall, this work delineates a miRNA-dependent post-transcriptional mechanism that controls the expression of the key synaptic scaffold, gephyrin, and may converge with parallel miRNA pathways to coordinate gene upregulation to maintain inhibitory synaptic plasticity.
Collapse
Affiliation(s)
- Theresa M. Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
- T.M.W and D.R. contributed equally to this work
| | - Dipen Rajgor
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
- T.M.W and D.R. contributed equally to this work
| | - Joshua D. Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Dean Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Sarah M. Zych
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Sara E. Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Tyler P. Martinez
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Mark L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Matthew J. Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| | - Katharine R. Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045
| |
Collapse
|
12
|
Bosetto Fiebrantz AK, Felski Leite L, Dal Pisol Schwab E, Sartori Bonini J, da Silva WC. On the participation of adenosinergic receptors in the reconsolidation of spatial long-term memory in male rats. Learn Mem 2023; 30:260-270. [PMID: 37802547 PMCID: PMC10561635 DOI: 10.1101/lm.053785.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/09/2023] [Indexed: 10/10/2023]
Abstract
To date, there is insufficient evidence to explain the role of adenosinergic receptors in the reconsolidation of long-term spatial memory. In this work, the role of the adenosinergic receptor family (A1, A2A, A2B, and A3) in this process has been elucidated. It was demonstrated that when infused bilaterally into the hippocampal CA1 region immediately after an early nonreinforced test session performed 24 h posttraining in the Morris water maze task, adenosine can cause anterograde amnesia for recent and late long-term spatial memory. This effect on spatial memory reconsolidation was blocked by A1 or A3 receptor antagonists and mimicked by A1 plus A3 receptor agonists, showing that this effect occurs through A1 and A3 receptors simultaneously. The A3 receptor alone participates only in the reconsolidation of late long-term spatial memory. When the memory to be reconsolidated was delayed (reactivation 5 d posttraining), the amnesic effect of adenosine became transient and did not occur in a test performed 5 d after the reactivation of the mnemonic trace. Finally, it has been shown that the amnesic effect of adenosine on spatial memory reconsolidation depends on the occurrence of protein degradation and that the amnesic effect of inhibition of protein synthesis on spatial memory reconsolidation is dependent on the activation of A3 receptors.
Collapse
Affiliation(s)
- Anne Karine Bosetto Fiebrantz
- Laboratório de Neuropsicofarmacologia, Departamento de Farmácia, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná 85040-167, Brasil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90035-003, Brasil
| | - Luana Felski Leite
- Laboratório de Neuropsicofarmacologia, Departamento de Farmácia, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná 85040-167, Brasil
| | - Eduarda Dal Pisol Schwab
- Laboratório de Neuropsicofarmacologia, Departamento de Farmácia, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná 85040-167, Brasil
| | - Juliana Sartori Bonini
- Laboratório de Neurociências e Comportamento, Departamento de Farmácia, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná 85040-167, Brasil
| | - Weber Cláudio da Silva
- Laboratório de Neuropsicofarmacologia, Departamento de Farmácia, Universidade Estadual do Centro-Oeste, Guarapuava, Paraná 85040-167, Brasil
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul 90035-003, Brasil
| |
Collapse
|
13
|
Keine C, Radulovic T, Al-Yaari M, Young SM. Confocal Imaging and 3D Reconstruction to Determine How Genetic Perturbations Impact Presynaptic Morphology at the Mouse Calyx of Held. Bio Protoc 2023; 13:e4799. [PMID: 37849785 PMCID: PMC10577601 DOI: 10.21769/bioprotoc.4799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 10/19/2023] Open
Abstract
Neurons communicate via synapses-specialized structures that consist of a presynaptic terminal of one neuron and a postsynaptic terminal of another. As knowledge is emerging that mutations in molecules that regulate synaptic function underpin many neurological disorders, it is crucial to elucidate the molecular mechanisms regulating synaptic function to understand synaptic strength, plasticity, modulation, and pathology, which ultimately impact neuronal circuit output and behavior. The presynaptic calyx of Held is a large glutamatergic presynaptic terminal in the auditory brainstem, which due to its accessibility and the possibility to selectively perform molecular perturbations on it, is an ideal model to study the role of presynaptic proteins in regulating synaptic function. In this protocol, we describe the use of confocal imaging and three-dimensional reconstruction of the calyx of Held to assess alterations in gross morphology following molecular perturbation. Using viral-vector delivery to perform molecular perturbations at distinct developmental time points, we provide a fast and cost-effective method to investigate how presynaptic proteins regulate gross morphology such as surface area and synapse volume throughout the lifetime of a neuronal circuit. Key features Confocal imaging and 3D reconstruction of presynaptic terminals. Used with a virus-mediated expression of mEGFP to achieve efficient, cell-type specific labeling of the presynaptic compartment. Protocol was developed with the calyx of Held but is suitable for pre- and postsynaptic compartments of various neurons across multiple mammalian and invertebrate species.
Collapse
Affiliation(s)
- Christian Keine
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of
Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg,
Germany
| | - Tamara Radulovic
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of
Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg,
Germany
| | - Mohammed Al-Yaari
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
- Department of Otolaryngology, Iowa Neuroscience
Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
14
|
Cohen T, Shomron N. Can RNA Affect Memory Modulation? Implications for PTSD Understanding and Treatment. Int J Mol Sci 2023; 24:12908. [PMID: 37629089 PMCID: PMC10454422 DOI: 10.3390/ijms241612908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/13/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Memories are a central aspect of our lives, but the mechanisms underlying their formation, consolidation, retrieval, and extinction remain poorly understood. In this review, we explore the molecular mechanisms of memory modulation and investigate the effects of RNA on these processes. Specifically, we examine the effects of time and location on gene expression alterations. We then discuss the potential for harnessing these alterations to modulate memories, particularly fear memories, to alleviate post-traumatic stress disorder (PTSD) symptoms. The current state of research suggests that transcriptional changes play a major role in memory modulation and targeting them through microRNAs may hold promise as a novel approach for treating memory-related disorders such as PTSD.
Collapse
Affiliation(s)
- Tehila Cohen
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Edmond J Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Tel Aviv University Innovation Labs (TILabs), Tel Aviv 6997801, Israel
| |
Collapse
|
15
|
Ma T. Roles of eukaryotic elongation factor 2 kinase (eEF2K) in neuronal plasticity, cognition, and Alzheimer disease. J Neurochem 2023; 166:47-57. [PMID: 34796967 PMCID: PMC9117558 DOI: 10.1111/jnc.15541] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 11/15/2021] [Indexed: 01/04/2023]
Abstract
Understanding the molecular signaling mechanisms underlying cognition and neuronal plasticity would provide insights into the pathogenesis of neuronal disorders characterized by cognitive syndromes such as Alzheimer disease (AD). Phosphorylation of the mRNA translational factor eukaryotic elongation factor 2 (eEF2) by its specific kinase eEF2K is critically involved in protein synthesis regulation. In this review, we discussed recent studies on the roles of eEF2K/eEF2 signaling in the context of regulation/dysregulation of cognitive function and synaptic plasticity. We specifically focus on the discussion of recent evidence indicating suppression of eEF2K signaling as a potential novel therapeutic avenue for AD and related dementias (ADRDs).
Collapse
Affiliation(s)
- Tao Ma
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine
| |
Collapse
|
16
|
Kuznetsov IA, Kuznetsov AV. Dynein Dysfunction Prevents Maintenance of High Concentrations of Slow Axonal Transport Cargos at the Axon Terminal: A Computational Study. J Biomech Eng 2023; 145:071001. [PMID: 36795013 PMCID: PMC10158974 DOI: 10.1115/1.4056915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
Here, we report computational studies of bidirectional transport in an axon, specifically focusing on predictions when the retrograde motor becomes dysfunctional. We are motivated by reports that mutations in dynein-encoding genes can cause diseases associated with peripheral motor and sensory neurons, such as type 2O Charcot-Marie-Tooth disease. We use two different models to simulate bidirectional transport in an axon: an anterograde-retrograde model, which neglects passive transport by diffusion in the cytosol, and a full slow transport model, which includes passive transport by diffusion in the cytosol. As dynein is a retrograde motor, its dysfunction should not directly influence anterograde transport. However, our modeling results unexpectedly predict that slow axonal transport fails to transport cargos against their concentration gradient without dynein. The reason is the lack of a physical mechanism for the reverse information flow from the axon terminal, which is required so that the cargo concentration at the terminal could influence the cargo concentration distribution in the axon. Mathematically speaking, to achieve a prescribed concentration at the terminal, equations governing cargo transport must allow for the imposition of a boundary condition postulating the cargo concentration at the terminal. Perturbation analysis for the case when the retrograde motor velocity becomes close to zero predicts uniform cargo distributions along the axon. The obtained results explain why slow axonal transport must be bidirectional to allow for the maintenance of concentration gradients along the axon length. Our result is limited to small cargo diffusivity, which is a reasonable assumption for many slow axonal transport cargos (such as cytosolic and cytoskeletal proteins, neurofilaments, actin, and microtubules) which are transported as large multiprotein complexes or polymers.
Collapse
Affiliation(s)
- Ivan A. Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Andrey V. Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910
| |
Collapse
|
17
|
Andres-Alonso M, Borgmeyer M, Mirzapourdelavar H, Lormann J, Klein K, Schweizer M, Hoffmeister-Ullerich S, Oelschlegel AM, Dityatev A, Kreutz MR. Golgi satellites are essential for polysialylation of NCAM and expression of LTP at distal synapses. Cell Rep 2023; 42:112692. [PMID: 37355986 DOI: 10.1016/j.celrep.2023.112692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/28/2023] [Accepted: 06/08/2023] [Indexed: 06/27/2023] Open
Abstract
The complex cytoarchitecture of neurons poses significant challenges for the maturation of synaptic membrane proteins. It is currently unclear whether locally secreted synaptic proteins bypass the Golgi or whether they traffic through Golgi satellites (GSs). Here, we create a transgenic GS reporter mouse line and show that GSs are widely distributed along dendrites and are capable of mature glycosylation, in particular sialylation. We find that polysialylation of locally secreted NCAM takes place at GSs. Accordingly, in mice lacking a component of trans-Golgi network-to-plasma membrane trafficking, we find fewer GSs and significantly reduced PSA-NCAM levels in distal dendrites of CA1 neurons that receive input from the temporoammonic pathway. Induction of long-term potentiation at those, but not more proximal, synapses is severely impaired. We conclude that GSs serve the need for local mature glycosylation of synaptic membrane proteins in distal dendrites and thereby contribute to rapid changes in synaptic strength.
Collapse
Affiliation(s)
- Maria Andres-Alonso
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany.
| | - Maximilian Borgmeyer
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | | | - Jakob Lormann
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Kim Klein
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michaela Schweizer
- Core Facility Morphology und Electron Microscopy, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sabine Hoffmeister-Ullerich
- Core Facility Bioanalytik, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anja M Oelschlegel
- RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; Medical Faculty, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Michael R Kreutz
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
18
|
Rubio FJ, Olivares DE, Dunn C, Zhang S, Hilaire EM, Henry A, Mejias-Aponte C, Nogueras-Ortiz CJ, Selvam PV, Cruz FC, Madangopal R, Morales M, Hope BT. Flow Cytometry of Synaptoneurosomes (FCS) Reveals Increased Ribosomal S6 and Calcineurin Proteins in Activated Medial Prefrontal Cortex to Nucleus Accumbens Synapses. J Neurosci 2023; 43:4217-4233. [PMID: 37160369 PMCID: PMC10255002 DOI: 10.1523/jneurosci.0927-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 05/11/2023] Open
Abstract
Learning and behavior activate cue-specific patterns of sparsely distributed cells and synapses called ensembles that undergo memory-encoding engram alterations. While Fos is often used to label selectively activated cell bodies and identify neuronal ensembles, there is no comparable endogenous marker to label activated synapses and identify synaptic ensembles. For the purpose of identifying candidate synaptic activity markers, we optimized a flow cytometry of synaptoneurosome (FCS) procedure for assessing protein alterations in activated synapses from male and female rats. After injecting yellow fluorescent protein (YFP)-expressing adeno-associated virus into medial prefrontal cortex (mPFC) to label terminals in nucleus accumbens (NAc) of rats, we injected 20 mg/kg cocaine in a novel context (cocaine+novelty) to activate synapses, and prepared NAc synaptoneurosomes 0-60 min following injections. For FCS, we used commercially available antibodies to label presynaptic and postsynaptic markers synaptophysin and PSD-95 as well as candidate markers of synaptic activity [activity-regulated cytoskeleton protein (Arc), CaMKII and phospho-CaMKII, ribosomal protein S6 (S6) and phospho-S6, and calcineurin and phospho-calcineurin] in YFP-labeled synaptoneurosomes. Cocaine+novelty increased the percentage of S6-positive synaptoneurosomes at 5-60 min and calcineurin-positive synaptoneurosomes at 5-10 min. Electron microscopy verified that S6 and calcineurin levels in synaptoneurosomes were increased 10 min after cocaine+novelty. Pretreatment with the anesthetic chloral hydrate blocked cocaine+novelty-induced S6 and calcineurin increases in synaptoneurosomes, and novel context exposure alone (without cocaine) increased S6, both of which indicate that these increases were due to neural activity per se. Overall, FCS can be used to study protein alterations in activated synapses coming from specifically labeled mPFC projections to NAc.SIGNIFICANCE STATEMENT Memories are formed during learning and are stored in the brain by long-lasting molecular and cellular alterations called engrams formed within specific patterns of cue-activated neurons called neuronal ensembles. While Fos has been used to identify activated ensemble neurons and the engrams within them, we have not had a similar marker for activated synapses that can be used to identify synaptic engrams. Here we developed a procedure for high-throughput in-line analysis of flow cytometry of synaptoneurosome (FCS) and found that ribosomal S6 protein and calcineurin were increased in activated mPFC-NAc synapses. FCS can be used to study protein alterations in activated synapses within specifically labeled circuits.
Collapse
Affiliation(s)
- F Javier Rubio
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Daniel E Olivares
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Christopher Dunn
- Flow Cytometry Unit, Intramural Research Program/National Institute on Aging/National Institutes of Health, Baltimore, Maryland 21224
| | - Shiliang Zhang
- Confocal and Electron Microscopy Core, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Elias M Hilaire
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Akeem Henry
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Carlos Mejias-Aponte
- Confocal and Electron Microscopy Core, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Carlos J Nogueras-Ortiz
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, Baltimore, Maryland 21224
| | - Pooja V Selvam
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Fabio C Cruz
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, CEP 04023-062, São Paulo, Brazil
| | - Rajtarun Madangopal
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Marisela Morales
- Neuronal Networks Section, Integrative Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| | - Bruce T Hope
- Neuronal Ensembles in Addiction Section, Behavioral Neuroscience Research Branch, Intramural Research Program/National Institute on Drug Abuse/National Institutes of Health, Baltimore, Maryland 21224
| |
Collapse
|
19
|
Pavon MV, Navakkode S, Wong LW, Sajikumar S. Inhibition of Nogo-A rescues synaptic plasticity and associativity in APP/PS1 animal model of Alzheimer's disease. Semin Cell Dev Biol 2023; 139:111-120. [PMID: 35431138 DOI: 10.1016/j.semcdb.2022.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and cognitive decline. Synaptic impairment is one of the first events to occur in the progression of this disease. Synaptic plasticity and cellular association of various plastic events have been shown to be affected in AD models. Nogo-A, a well-known axonal growth inhibitor with a recently discovered role as a plasticity suppressor, and its main receptor Nogo-66 receptor 1 (NGR1) have been found to be overexpressed in the hippocampus of Alzheimer's patients. However, the role of Nogo-A and its receptor in the pathology of AD is still widely unknown. In this work we set out to investigate whether Nogo-A is working as a plasticity suppressor in AD. Our results show that inhibition of the Nogo-A pathway via the Nogo-R antibody in an Alzheimer's mouse model, APP/PS1, leads to the restoration of both synaptic plasticity and associativity in a protein synthesis and NMDR-dependent manner. We also show that inhibition of the p75NTR pathway, which is strongly associated with NGR1, restores synaptic plasticity as well. Mechanistically, we propose that the restoration of synaptic plasticity in APP/PS1 via inhibition of the Nogo-A pathway is due to the modulation of the RhoA-ROCK2 pathway and increase in plasticity related proteins. Our study identifies Nogo-A as a plasticity suppressor in AD models hence targeting Nogo-A could be a promising strategy to understanding AD pathology.
Collapse
Affiliation(s)
- Maria Vazquez Pavon
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sheeja Navakkode
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore.
| |
Collapse
|
20
|
Inhibition of hippocampal palmitoyl acyltransferase activity impairs spatial learning and memory consolidation. Neurobiol Learn Mem 2023; 200:107733. [PMID: 36804592 DOI: 10.1016/j.nlm.2023.107733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/12/2023] [Accepted: 02/15/2023] [Indexed: 02/21/2023]
Abstract
Protein palmitoylation regulates trafficking, mobilization, localization, interaction, and distribution of proteins through the palmitoyl acyltransferases (PATs) enzymes. Protein palmitoylation controls rapid and dynamic changes of the synaptic architecture that modifies the efficiency and strength of synaptic connections, a fundamental mechanism to generate stable and long-lasting memory traces. Although protein palmitoylation in functional synaptic plasticity has been widely described, its role in learning and memory processes is poorly understood. In this work, we found that PATs inhibition into the hippocampus before and after the training of Morris water maze (MWM) and object location memory (OLM) impaired spatial learning. However, we demonstrated that PATs inhibition during the retrieval does not affect the expression of spatial memory in both MWM and OLM. Accordingly, long-term potentiation induction is impaired by inhibiting PATs into the hippocampus before high-frequency electrical stimulation but not after. These findings suggest that PATs activity is necessary to modify neural plasticity, a mechanism required for memory acquisition and consolidation. Like phosphorylation, active palmitoylation is required to regulate the function of already existing proteins that change synaptic strength in the hippocampus to acquire and later consolidate spatial memories.
Collapse
|
21
|
Cohen LD, Ziv T, Ziv NE. Synapse integrity and function: Dependence on protein synthesis and identification of potential failure points. Front Mol Neurosci 2022; 15:1038614. [PMID: 36583084 PMCID: PMC9792512 DOI: 10.3389/fnmol.2022.1038614] [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: 09/07/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022] Open
Abstract
Synaptic integrity and function depend on myriad proteins - labile molecules with finite lifetimes that need to be continually replaced with freshly synthesized copies. Here we describe experiments designed to expose synaptic (and neuronal) properties and functions that are particularly sensitive to disruptions in protein supply, identify proteins lost early upon such disruptions, and uncover potential, yet currently underappreciated failure points. We report here that acute suppressions of protein synthesis are followed within hours by reductions in spontaneous network activity levels, impaired oxidative phosphorylation and mitochondrial function, and, importantly, destabilization and loss of both excitatory and inhibitory postsynaptic specializations. Conversely, gross impairments in presynaptic vesicle recycling occur over longer time scales (days), as does overt cell death. Proteomic analysis identified groups of potentially essential 'early-lost' proteins including regulators of synapse stability, proteins related to bioenergetics, fatty acid and lipid metabolism, and, unexpectedly, numerous proteins involved in Alzheimer's disease pathology and amyloid beta processing. Collectively, these findings point to neuronal excitability, energy supply and synaptic stability as early-occurring failure points under conditions of compromised supply of newly synthesized protein copies.
Collapse
Affiliation(s)
- Laurie D. Cohen
- Technion Faculty of Medicine, Rappaport Institute and Network Biology Research Laboratories, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion, Haifa, Israel
| | - Noam E. Ziv
- Technion Faculty of Medicine, Rappaport Institute and Network Biology Research Laboratories, Haifa, Israel,*Correspondence: Noam E. Ziv,
| |
Collapse
|
22
|
Alhowail A. Mechanisms Underlying Cognitive Impairment Induced by Prenatal Alcohol Exposure. Brain Sci 2022; 12:brainsci12121667. [PMID: 36552126 PMCID: PMC9775935 DOI: 10.3390/brainsci12121667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/07/2022] Open
Abstract
Alcohol is one of the most commonly used illicit substances among pregnant women. Clinical and experimental studies have revealed that prenatal alcohol exposure affects fetal brain development and ultimately results in the persistent impairment of the offspring's cognitive functions. Despite this, the rate of alcohol use among pregnant women has been progressively increasing. Various aspects of human and animal behavior, including learning and memory, are dependent on complex interactions between multiple mechanisms, such as receptor function, mitochondrial function, and protein kinase activation, which are especially vulnerable to alterations during the developmental period. Thus, the exploration of the mechanisms that are altered in response to prenatal alcohol exposure is necessary to develop an understanding of how homeostatic imbalance and various long-term neurobehavioral impairments manifest following alcohol abuse during pregnancy. There is evidence that prenatal alcohol exposure results in vast alterations in mechanisms such as long-term potentiation, mitochondrial function, and protein kinase activation in the brain of offspring. However, to the best of our knowledge, there are very few recent reviews that focus on the cognitive effects of prenatal alcohol exposure and the associated mechanisms. Therefore, in this review, we aim to provide a comprehensive summary of the recently reported alterations to various mechanisms following alcohol exposure during pregnancy, and to draw potential associations with behavioral changes in affected offspring.
Collapse
Affiliation(s)
- Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Al Qassim 51452, Saudi Arabia
| |
Collapse
|
23
|
Protein synthesis inhibition and loss of homeostatic functions in astrocytes from an Alzheimer's disease mouse model: a role for ER-mitochondria interaction. Cell Death Dis 2022; 13:878. [PMID: 36257957 PMCID: PMC9579125 DOI: 10.1038/s41419-022-05324-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Deregulation of protein synthesis and ER stress/unfolded protein response (ER stress/UPR) have been reported in astrocytes. However, the relationships between protein synthesis deregulation and ER stress/UPR, as well as their role in the altered homeostatic support of Alzheimer's disease (AD) astrocytes remain poorly understood. Previously, we reported that in astrocytic cell lines from 3xTg-AD mice (3Tg-iAstro) protein synthesis was impaired and ER-mitochondria distance was reduced. Here we show that impaired protein synthesis in 3Tg-iAstro is associated with an increase of p-eIF2α and downregulation of GADD34. Although mRNA levels of ER stress/UPR markers were increased two-three-fold, we found neither activation of PERK nor downstream induction of ATF4 protein. Strikingly, the overexpression of a synthetic ER-mitochondrial linker (EML) resulted in a reduced protein synthesis and augmented p-eIF2α without any effect on ER stress/UPR marker genes. In vivo, in hippocampi of 3xTg-AD mice, reduced protein synthesis, increased p-eIF2α and downregulated GADD34 protein were found, while no increase of p-PERK or ATF4 proteins was observed, suggesting that in AD astrocytes, both in vitro and in vivo, phosphorylation of eIF2α and impairment of protein synthesis are PERK-independent. Next, we investigated the ability of 3xTg-AD astrocytes to support metabolism and function of other cells of the central nervous system. Astrocyte-conditioned medium (ACM) from 3Tg-iAstro cells significantly reduced protein synthesis rate in primary hippocampal neurons. When added as a part of pericyte/endothelial cell (EC)/astrocyte 3D co-culture, 3Tg-iAstro, but not WT-iAstro, severely impaired formation and ramification of tubules, the effect, replicated by EML overexpression in WT-iAstro cells. Finally, a chemical chaperone 4-phenylbutyric acid (4-PBA) rescued protein synthesis, p-eIF2α levels in 3Tg-iAstro cells and tubulogenesis in pericyte/EC/3Tg-iAstro co-culture. Collectively, our results suggest that a PERK-independent, p-eIF2α-associated impairment of protein synthesis compromises astrocytic homeostatic functions, and this may be caused by the altered ER-mitochondria interaction.
Collapse
|
24
|
Vasilopoulou F, Bellver-Sanchis A, Companys-Alemany J, Jarne-Ferrer J, Irisarri A, Palomera-Ávalos V, Gonzalez-Castillo C, Ortuño-Sahagún D, Sanfeliu C, Pallàs M, Griñán-Ferré C. Cognitive Decline and BPSD Are Concomitant with Autophagic and Synaptic Deficits Associated with G9a Alterations in Aged SAMP8 Mice. Cells 2022; 11:cells11162603. [PMID: 36010679 PMCID: PMC9406492 DOI: 10.3390/cells11162603] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/14/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
Behavioural and psychological symptoms of dementia (BPSD) are presented in 95% of Alzheimer’s Disease (AD) patients and are also associated with neurotrophin deficits. The molecular mechanisms leading to age-related diseases are still unclear; however, emerging evidence has suggested that epigenetic modulation is a key pathophysiological basis of ageing and neurodegeneration. In particular, it has been suggested that G9a methyltransferase and its repressive histone mark (H3K9me2) are important in shaping learning and memory by modulating autophagic activity and synaptic plasticity. This work deepens our understanding of the epigenetic mechanisms underlying the loss of cognitive function and BPSD in AD. For this purpose, several tasks were performed to evaluate the parameters of sociability (three-chamber test), aggressiveness (resident intruder), anxiety (elevated plus maze and open field) and memory (novel object recognition test) in mice, followed by the evaluation of epigenetic, autophagy and synaptic plasticity markers at the molecular level. The behavioural alterations presented by senescence-accelerated mice prone 8 (SAMP8) of 12 months of age compared with their senescence-accelerated mouse resistant mice (SAMR1), the healthy control strain was accompanied by age-related cognitive deficits and alterations in epigenetic markers. Increased levels of G9a are concomitant to the dysregulation of the JNK pathway in aged SAMP8, driving a failure in autophagosome formation. Furthermore, lower expression of the genes involved in the memory-consolidation process modulated by ERK was observed in the aged male SAMP8 model, suggesting the implication of G9a. In any case, two of the most important neurotrophins, namely brain-derived neurotrophic factor (Bdnf) and neurotrophin-3 (NT3), were found to be reduced, along with a decrease in the levels of dendritic branching and spine density presented by SAMP8 mice. Thus, the present study characterizes and provides information regarding the non-cognitive and cognitive states, as well as molecular alterations, in aged SAMP8, demonstrating the AD-like symptoms presented by this model. In any case, our results indicate that higher levels of G9a are associated with autophagic deficits and alterations in synaptic plasticity, which could further explain the BPSD and cognitive decline exhibited by the model.
Collapse
Affiliation(s)
- Foteini Vasilopoulou
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
| | - Aina Bellver-Sanchis
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
| | - Júlia Companys-Alemany
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
| | - Júlia Jarne-Ferrer
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
| | - Alba Irisarri
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
| | - Verónica Palomera-Ávalos
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
| | | | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunología Molecular, Instituto de Investigación de Ciencias Biomédicas (IICB) CUCS, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Coral Sanfeliu
- Institut d’Investigacions Biomèdiques de Barcelona (IIBB), CSIC and Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
| | - Christian Griñán-Ferré
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona, Avda. Joan XXIII, 27, 08028 Barcelona, Spain
- Correspondence:
| |
Collapse
|
25
|
Ostroff LE, Cain CK. Persistent up-regulation of polyribosomes at synapses during long-term memory, reconsolidation, and extinction of associative memory. LEARNING & MEMORY (COLD SPRING HARBOR, N.Y.) 2022; 29:192-202. [PMID: 35882501 PMCID: PMC9374273 DOI: 10.1101/lm.053577.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Local protein synthesis at synapses can provide a rapid supply of proteins to support synaptic changes during consolidation of new memories, but its role in the maintenance or updating of established memories is unknown. Consolidation requires new protein synthesis in the period immediately following learning, whereas established memories are resistant to protein synthesis inhibitors. We have previously reported that polyribosomes are up-regulated in the lateral amygdala (LA) during consolidation of aversive-cued Pavlovian conditioning. In this study, we used serial section electron microscopy reconstructions to determine whether the distribution of dendritic polyribosomes returns to baseline during the long-term memory phase. Relative to control groups, long-term memory was associated with up-regulation of polyribosomes throughout dendrites, including in dendritic spines of all sizes. Retrieval of a consolidated memory by presentation of a small number of cues induces a new, transient requirement for protein synthesis to maintain the memory, while presentation of a large number of cues results in extinction learning, forming a new memory. One hour after retrieval or extinction training, the distribution of dendritic polyribosomes was similar except in the smallest spines, which had more polyribosomes in the extinction group. Our results demonstrate that the effects of learning on dendritic polyribosomes are not restricted to the transient translation-dependent phase of memory formation. Cued Pavlovian conditioning induces persistent synapse strengthening in the LA that is not reversed by retrieval or extinction, and dendritic polyribosomes may therefore correlate generally with synapse strength as opposed to recent activity or transient translational processes.
Collapse
Affiliation(s)
- Linnaea E Ostroff
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA.,Connecticut Institute for the Brain and Cognitive Science, University of Connecticut, Storrs, Connecticut 06269, USA.,Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Christopher K Cain
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York 10962, USA.,Child and Adolescent Psychiatry, New York University Langone Health, New York, New York 10016, USA
| |
Collapse
|
26
|
Youssef MMM, Hamada HT, Lai ESK, Kiyama Y, El-Tabbal M, Kiyonari H, Nakano K, Kuhn B, Yamamoto T. TOB is an effector of the hippocampus-mediated acute stress response. Transl Psychiatry 2022; 12:302. [PMID: 35906220 PMCID: PMC9338090 DOI: 10.1038/s41398-022-02078-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Stress affects behavior and involves critical dynamic changes at multiple levels ranging from molecular pathways to neural circuits and behavior. Abnormalities at any of these levels lead to decreased stress resilience and pathological behavior. However, temporal modulation of molecular pathways underlying stress response remains poorly understood. Transducer of ErbB2.1, known as TOB, is involved in different physiological functions, including cellular stress and immediate response to stimulation. In this study, we investigated the role of TOB in psychological stress machinery at molecular, neural circuit, and behavioral levels. Interestingly, TOB protein levels increased after mice were exposed to acute stress. At the neural circuit level, functional magnetic resonance imaging (fMRI) suggested that intra-hippocampal and hippocampal-prefrontal connectivity were dysregulated in Tob knockout (Tob-KO) mice. Electrophysiological recordings in hippocampal slices showed increased postsynaptic AMPAR-mediated neurotransmission, accompanied by decreased GABA neurotransmission and subsequently altered Excitatory/Inhibitory balance after Tob deletion. At the behavioral level, Tob-KO mice show abnormal, hippocampus-dependent, contextual fear conditioning and extinction, and depression-like behaviors. On the other hand, increased anxiety observed in Tob-KO mice is hippocampus-independent. At the molecular level, we observed changes in factors involved in stress response like decreased stress-induced LCN2 expression and ERK phosphorylation, as well as increased MKP-1 expression. This study introduces TOB as an important modulator in the hippocampal stress signaling machinery. In summary, we reveal a molecular pathway and neural circuit mechanism by which Tob deletion contributes to expression of pathological stress-related behavior.
Collapse
Affiliation(s)
- Mohieldin M M Youssef
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| | - Hiro Taiyo Hamada
- Neural Computation Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Esther Suk King Lai
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Yuji Kiyama
- Laboratory of Biochemistry and Molecular Biology, Graduate school of medical and dental sciences, Kagoshima University, Kagoshima, Japan
| | - Mohamed El-Tabbal
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Kohei Nakano
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Bernd Kuhn
- Optical Neuroimaging Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
27
|
Gil-Marti B, Barredo CG, Pina-Flores S, Trejo JL, Turiegano E, Martin FA. The elusive transcriptional memory trace. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac008. [PMID: 38596710 PMCID: PMC10913820 DOI: 10.1093/oons/kvac008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/19/2022] [Accepted: 05/07/2022] [Indexed: 04/11/2024]
Abstract
Memory is the brain faculty to store and remember information. It is a sequential process in which four different phases can be distinguished: encoding or learning, consolidation, storage and reactivation. Since the discovery of the first Drosophila gene essential for memory formation in 1976, our knowledge of its mechanisms has progressed greatly. The current view considers the existence of engrams, ensembles of neuronal populations whose activity is temporally coordinated and represents the minimal correlate of experience in brain circuits. In order to form and maintain the engram, protein synthesis and, probably, specific transcriptional program(s) is required. The immediate early gene response during learning process has been extensively studied. However, a detailed description of the transcriptional response for later memory phases was technically challenging. Recent advances in transcriptomics have allowed us to tackle this biological problem. This review summarizes recent findings in this field, and discusses whether or not it is possible to identify a transcriptional trace for memory.
Collapse
Affiliation(s)
- Beatriz Gil-Marti
- Molecular Physiology of Behavior Laboratory, Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Spanish National Research Council (CSIC), 28002 Madrid, Spain
- Department of Biology, Autonomous University of Madrid, 28049 Madrid, Spain
| | - Celia G Barredo
- Molecular Physiology of Behavior Laboratory, Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Spanish National Research Council (CSIC), 28002 Madrid, Spain
| | - Sara Pina-Flores
- Molecular Physiology of Behavior Laboratory, Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Spanish National Research Council (CSIC), 28002 Madrid, Spain
| | - Jose Luis Trejo
- Neurogenesis of the Adult Animal Laboratory. Department of Translational Neuroscience, Cajal Institute, Spanish National Research Council (CSIC), 28049, Madrid, Spain
| | - Enrique Turiegano
- Department of Biology, Autonomous University of Madrid, 28049 Madrid, Spain
| | - Francisco A Martin
- Molecular Physiology of Behavior Laboratory, Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Spanish National Research Council (CSIC), 28002 Madrid, Spain
| |
Collapse
|
28
|
Lim JS, Kim HJ, Park I, Woo S, Kim JH, Park JW. Force Mapping Reveals the Spatial Distribution of Individual Proteins in a Neuron. NANO LETTERS 2022; 22:3865-3871. [PMID: 35549313 DOI: 10.1021/acs.nanolett.1c04395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Conventional methods for studying the spatial distribution and expression level of proteins within neurons have primarily relied on immunolabeling and/or signal amplification. Here, we present an atomic force microscopy (AFM)-based nanoscale force mapping method, where Anti-LIMK1-tethered AFM probes were used to visualize individual LIMK1 proteins in cultured neurons directly through force measurements. We observed that the number density of LIMK1 decreased in neuronal somas after the cells were depolarized. We also elucidated the spatial distribution of LIMK1 in single spine areas and found that the protein predominantly locates at heads of spines rather than dendritic shafts. The study demonstrates that our method enables unveiling of the abundance and spatial distribution of a protein of interest in neurons without signal amplification or labeling. We expected that this approach should facilitate the studies of protein expression phenomena in depth in a wide range of biological systems.
Collapse
Affiliation(s)
- Ji-Seon Lim
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Hyun Jin Kim
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Ikbum Park
- Analysis and Assessment Research Center, Research Institute of Industrial Science and Technology, 67 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Sungwook Woo
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Sciences, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Joon Won Park
- Department of Chemistry, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
- Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
29
|
Kuznetsov IA, Kuznetsov AV. Bidirectional, unlike unidirectional transport, allows transporting axonal cargos against their concentration gradient. J Theor Biol 2022; 546:111161. [DOI: 10.1016/j.jtbi.2022.111161] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/04/2022] [Accepted: 05/06/2022] [Indexed: 11/25/2022]
|
30
|
Grochowska KM, Andres‐Alonso M, Karpova A, Kreutz MR. The needs of a synapse—How local organelles serve synaptic proteostasis. EMBO J 2022; 41:e110057. [PMID: 35285533 PMCID: PMC8982616 DOI: 10.15252/embj.2021110057] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/24/2021] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Synaptic function crucially relies on the constant supply and removal of neuronal membranes. The morphological complexity of neurons poses a significant challenge for neuronal protein transport since the machineries for protein synthesis and degradation are mainly localized in the cell soma. In response to this unique challenge, local micro‐secretory systems have evolved that are adapted to the requirements of neuronal membrane protein proteostasis. However, our knowledge of how neuronal proteins are synthesized, trafficked to membranes, and eventually replaced and degraded remains scarce. Here, we review recent insights into membrane trafficking at synaptic sites and into the contribution of local organelles and micro‐secretory pathways to synaptic function. We describe the role of endoplasmic reticulum specializations in neurons, Golgi‐related organelles, and protein complexes like retromer in the synthesis and trafficking of synaptic transmembrane proteins. We discuss the contribution of autophagy and of proteasome‐mediated and endo‐lysosomal degradation to presynaptic proteostasis and synaptic function, as well as nondegradative roles of autophagosomes and lysosomes in signaling and synapse remodeling. We conclude that the complexity of neuronal cyto‐architecture necessitates long‐distance protein transport that combines degradation with signaling functions.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Maria Andres‐Alonso
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
| | - Anna Karpova
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
| | - Michael R Kreutz
- Leibniz Group “Dendritic Organelles and Synaptic Function” Center for Molecular Neurobiology ZMNH University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Research Group Neuroplasticity Leibniz Institute for Neurobiology Magdeburg Germany
- Center for Behavioral Brain Sciences Otto von Guericke University Magdeburg Germany
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
| |
Collapse
|
31
|
Yeh KC, Hung CF, Lee HL, Hsieh TY, Wang SJ. Soybean Meal Extract Preserves Memory Ability by Increasing Presynaptic Function and Modulating Gut Microbiota in Rats. Mol Neurobiol 2022; 59:1649-1664. [PMID: 35001354 DOI: 10.1007/s12035-021-02669-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022]
Abstract
Age-related degenerative brain diseases frequently manifest as memory deficits. Dietary interventions or nutraceuticals may provide efficacious treatments through prevention and cure. Soybean meal, a byproduct of soy oil refining, has health benefits, but its effect on memory function is unknown. Therefore, we evaluated the effect of the oral administration of soybean meal extract (SME) for 2 weeks on memory function using the Morris water maze (MWM) test in healthy rats and investigated the possible underlying mechanisms. First, analysis of the composition revealed that SME is rich in isoflavones; SME did not exhibit hepatotoxicity or renal toxicity at the different doses tested. The MWM results revealed that the escape latency and movement distance of rats were significantly shorter in the SME group than in the control group, indicating that SME can help in memory preservation. In addition, SME increased the levels of presynaptic proteins such as synaptophysin, synaptobrevin, synaptotagmin, syntaxin, synapsin I, and 25-kDa synaptosome-associated protein as well as protein kinases and their phosphorylated expression, including extracellular signal-regulated kinases 1 and 2 (ERK1/2), protein kinase C (PKC), and Ca2+/calmodulin-dependent protein kinase II (CaMKII) in the hippocampal nerve terminals (synaptosomes). Transmission electron microscopy also indicated that SME increased the number of synaptic vesicles in hippocampal synaptosomes. Furthermore, SME rats exhibited altered microbiota composition compared with control rats. Therefore, our data suggest that SME can increase presynaptic function and modulate gut microbiota, thus aiding in memory preservation in rats.
Collapse
Affiliation(s)
- Kun-Chieh Yeh
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan
- Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- Department of Surgery, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Hui-Ling Lee
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Ting-Yang Hsieh
- P.H.D. Program in Neutrition & Food Science, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Su-Jane Wang
- School of Medicine, Fu Jen Catholic University, No.510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan.
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.
| |
Collapse
|
32
|
Kuznetsov IA, Kuznetsov AV. Can the lack of fibrillar form of alpha-synuclein in Lewy bodies be explained by its catalytic activity? Math Biosci 2022; 344:108754. [PMID: 34890628 PMCID: PMC8882444 DOI: 10.1016/j.mbs.2021.108754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/03/2023]
Abstract
Finding the causative pathophysiological mechanisms for Parkinson's disease (PD) is important for developing therapeutic interventions. Until recently, it was believed that Lewy bodies (LBs), the hallmark of PD, are mostly composed of alpha-synuclein (α-syn) fibrils. Recent results (Shahmoradian et al. (2019)) demonstrated that the fibrillar form of α-syn is lacking from LBs. Here we propose that this surprising observation can be explained by the catalytic activity of the fibrillar form of α-syn. We assumed that α-syn fibrils catalyze the formation of LBs, but do not become part of them. We developed a mathematical model based on this hypothesis. By using the developed model, we investigated the consequences of this hypothesis. In particular, the model suggests that the long incubation time of PD can be explained by a two-step aggregation process that leads to its development: (i) aggregation of monomeric α-syn into α-syn oligomers and fibrils and (ii) clustering of membrane-bound organelles, which may cause disruption of axonal trafficking and lead to neuron starvation and death. The model shows that decreasing the rate of destruction of α-syn aggregates in somatic lysosomes accelerates the formation of LBs. Another consequence of the model is the prediction that removing α-syn aggregates from the brain after the aggregation of membrane-bound organelles into LBs has started may not stop the progression of PD because LB formation is an autocatalytic process; hence, the formation of LBs will be catalyzed by aggregates of membrane-bound organelles even in the absence of α-syn aggregates. The performed sensitivity study made it possible to establish the hierarchy of model parameters with respect to their effect on the formation of vesicle aggregates in the soma.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910, USA.
| |
Collapse
|
33
|
Khlebodarova TM. The molecular view of mechanical stress of brain cells, local translation, and neurodegenerative diseases. Vavilovskii Zhurnal Genet Selektsii 2021; 25:92-100. [PMID: 34901706 PMCID: PMC8629365 DOI: 10.18699/vj21.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/03/2022] Open
Abstract
The assumption that chronic mechanical stress in brain cells stemming from intracranial hypertension,
arterial hypertension, or mechanical injury is a risk factor for neurodegenerative diseases was put forward in the
1990s and has since been supported. However, the molecular mechanisms that underlie the way from cell exposure to mechanical stress to disturbances in synaptic plasticity followed by changes in behavior, cognition, and
memory are still poorly understood. Here we review (1) the current knowledge of molecular mechanisms regulating local translation and the actin cytoskeleton state at an activated synapse, where they play a key role in the
formation of various sorts of synaptic plasticity and long-term memory, and (2) possible pathways of mechanical
stress intervention. The roles of the mTOR (mammalian target of rapamycin) signaling pathway; the RNA-binding
FMRP protein; the CYFIP1 protein, interacting with FMRP; the family of small GTPases; and the WAVE regulatory
complex in the regulation of translation initiation and actin cytoskeleton rearrangements in dendritic spines of the
activated synapse are discussed. Evidence is provided that chronic mechanical stress may result in aberrant activation of mTOR signaling and the WAVE regulatory complex via the YAP/TAZ system, the key sensor of mechanical
signals, and influence the associated pathways regulating the formation of F actin filaments and the dendritic spine
structure. These consequences may be a risk factor for various neurological conditions, including autistic spectrum
disorders and epileptic encephalopathy. In further consideration of the role of the local translation system in the
development of neuropsychic and neurodegenerative diseases, an original hypothesis was put forward that one
of the possible causes of synaptopathies is impaired proteome stability associated with mTOR hyperactivity and
formation of complex dynamic modes of de novo protein synthesis in response to synapse-stimulating factors,
including chronic mechanical stress.
Collapse
Affiliation(s)
- T M Khlebodarova
- Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Kurchatov Genomic Center of the Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
34
|
Wang Z, Yu W, Liu L, Niu J, Zhang X, Nan F, Xu L, Jiang B, Ke D, Zhu W, Tian Z, Wang Y, Wang B. Human Cytomegalovirus Immediate Early Protein 2 Protein Causes Cognitive Disorder by Damaging Synaptic Plasticity in Human Cytomegalovirus-UL122-Tg Mice. Front Aging Neurosci 2021; 13:720582. [PMID: 34790111 PMCID: PMC8591137 DOI: 10.3389/fnagi.2021.720582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/07/2021] [Indexed: 11/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection is very common in the human population all around the world. Although the majority of HCMV infections are asymptomatic, they can cause neurologic deficits. Previous studies have shown that immediate early protein 2 (IE2, also known as UL122) of HCMV is related with the cognitive disorder mechanism. Due to species isolation, a HCMV-infected animal model could not be established which meant a study into the long-term effects of IE2 on neural development could not be carried out. By establishing HCMV-UL122-Tg mice (UL122 mice), we explored the cognitive behavior and complexity of neuron changes in this transgenic UL122 mice that could consistently express IE2 protein at different ages (confirmed in both 6- and 12-month-old UL122 mice). In the Morris water maze, cognitive impairment was more pronounced in 12-month-old UL122 mice than in 6-month-old ones. At the same time, a decrease of the density of dendritic spines and branches in the hippocampal neurons of 12-month-old mice was observed. Moreover, long-term potentiation was showed to be impaired in 12-month-old UL122 mice. The expressions of several synaptic plasticity-regulated molecules were reduced in 12-month-old UL122 mice, including scaffold proteins postsynaptic density protein 95 (PSD95) and microtubule-associated protein 2 (MAP2). Binding the expression of IE2 was increased in 12-month-old mice compared with 6-month-old mice, and results of statistical analysis suggested that the cognitive damage was not caused by natural animal aging, which might exclude the effect of natural aging on cognitive impairment. All these results suggested that IE2 acted as a pathogenic regulator in damaging synaptic plasticity by downregulating the expression of plasticity-related proteins (PRPs), and this damage increased with aging.
Collapse
Affiliation(s)
- Zhifei Wang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenwen Yu
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Lili Liu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Junyun Niu
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xianjuan Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fulong Nan
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lili Xu
- Qingdao Women and Childrens Hospital, Qingdao University, Qingdao, China
| | - Bin Jiang
- Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Dingxin Ke
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenhua Zhu
- Qingdao Fuwai Cardiovascular Hospital, Qingdao, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yashuo Wang
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
35
|
Sanchez-Mirasierra I, Hernandez-Diaz S, Ghimire S, Montecinos-Oliva C, Soukup SF. Macros to Quantify Exosome Release and Autophagy at the Neuromuscular Junction of Drosophila Melanogaster. Front Cell Dev Biol 2021; 9:773861. [PMID: 34869373 PMCID: PMC8634598 DOI: 10.3389/fcell.2021.773861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/21/2021] [Indexed: 01/18/2023] Open
Abstract
Automatic quantification of image parameters is a powerful and necessary tool to explore and analyze crucial cell biological processes. This article describes two ImageJ/Fiji automated macros to approach the analysis of synaptic autophagy and exosome release from 2D confocal images. Emerging studies point out that exosome biogenesis and autophagy share molecular and organelle components. Indeed, the crosstalk between these two processes may be relevant for brain physiology, neuronal development, and the onset/progression of neurodegenerative disorders. In this context, we describe here the macros "Autophagoquant" and "Exoquant" to assess the quantification of autophagosomes and exosomes at the neuronal presynapse of the Neuromuscular Junction (NMJ) in Drosophila melanogaster using confocal microscopy images. The Drosophila NMJ is a valuable model for the study of synapse biology, autophagy, and exosome release. By use of Autophagoquant and Exoquant, researchers can have an unbiased, standardized, and rapid tool to analyze autophagy and exosomal release in Drosophila NMJ. Code available at: https://github.com/IreneSaMi/Exoquant-Autophagoquant.
Collapse
Affiliation(s)
| | | | | | | | - Sandra-Fausia Soukup
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
36
|
Fu CH, Han XY, Tong L, Nie PY, Hu YD, Ji LL. miR-142 downregulation alleviates the impairment of spatial learning and memory, reduces the level of apoptosis, and upregulates the expression of pCaMKII and BAI3 in the hippocampus of APP/PS1 transgenic mice. Behav Brain Res 2021; 414:113485. [PMID: 34302879 DOI: 10.1016/j.bbr.2021.113485] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/20/2021] [Accepted: 07/17/2021] [Indexed: 01/20/2023]
Abstract
MicroRNA-142-5p (miR-142-5p) has been found to be dysregulated in several neurodegenerative disorders. However, little is known about the involvement of miR-142-5p in Alzheimer's disease (AD). Brain angiogenesis inhibitor 3 (BAI3), which belongs to the adhesion-G protein-coupled receptor subgroup, contributes to a variety of neuropsychiatric disorders. Despite its very high expression in neurons, the role of BAI3 in AD remains elusive, and its mechanism at the cellular and molecular levels needs to be further elucidated. The current study sought to investigate whether miR-142-5p influenced BAI3 expression and neuronal synaptotoxicity induced by Aβ, both in APP/PS1 transgenic mice and a cellular model of Alzheimer's disease. Altered expression of miR-142-5p was found in the hippocampus of AD mice. Inhibition of miR-142 could upregulate BAI3 expression, enhance neuronal viability and prevent neurons from undergoing apoptosis. In addition, the reduction of phosphorylation of Synapsin I and calcium/calmodulin-dependent protein kinase II (CaMKII), as well as the expression of PSD-95 in the hippocampus of APP/PS1 transgenic mice, were significantly restored by inhibiting miR-142. Meanwhile, the levels of Aβ1-42, β-APP, BACE-1 and PS-1 in cultured neurons were detected, and the effects of inhibiting miR-142 on spatial learning and memory were also observed. Interestingly, we found that BAI3, an important regulator of excitatory synapses, was a potential target gene of miR-142-5p. Collectively, our findings suggest that miR-142 inhibition can alleviate the impairment of spatial learning and memory, reduce the level of apoptosis, and upregulate the expression of pCaMKII and BAI3 in the hippocampus of APP/PS1 transgenic mice; thus, appropriate interference of miR-142 may provide a potential therapeutic approach to rescue cognitive dysfunction in AD patients.
Collapse
Affiliation(s)
- Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xue-Yan Han
- Department of Neurology, Seventh People's Hospital of Jinan City, Jinan, China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Peng-Yin Nie
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yue-Dong Hu
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
37
|
Reynders M, Trauner D. Optical control of targeted protein degradation. Cell Chem Biol 2021; 28:969-986. [PMID: 34115971 DOI: 10.1016/j.chembiol.2021.05.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/12/2021] [Accepted: 05/14/2021] [Indexed: 12/13/2022]
Abstract
Molecular glues and proteolysis targeting chimeras (PROTACs) have emerged as small-molecule tools that selectively induce the degradation of a chosen protein and have shown therapeutic promise. Recently, several approaches employing light as an additional stimulus to control induced protein degradation have been reported. Here, we analyze the principles guiding the design of such systems, provide a survey of the literature published to date, and discuss opportunities for further development. Light-responsive degraders enable the precise temporal and spatial control of protein levels, making them useful research tools but also potential candidates for human precision medicine.
Collapse
Affiliation(s)
- Martin Reynders
- Department of Chemistry, New York University, New York, NY 10003, USA; Department of Chemistry, Ludwig Maximilians University of Munich, 81377 Munich, Germany
| | - Dirk Trauner
- Department of Chemistry, New York University, New York, NY 10003, USA; Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA; NYU Neuroscience Institute, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
38
|
Gindina S, Botsford B, Cowansage K, LeDoux J, Klann E, Hoeffer C, Ostroff L. Upregulation of eIF4E, but not other translation initiation factors, in dendritic spines during memory formation. J Comp Neurol 2021; 529:3112-3126. [PMID: 33864263 DOI: 10.1002/cne.25158] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/27/2021] [Accepted: 04/11/2021] [Indexed: 11/09/2022]
Abstract
Local translation can provide a rapid, spatially targeted supply of new proteins in distal dendrites to support synaptic changes that underlie learning. Learning and memory are especially sensitive to manipulations of translational control mechanisms, particularly those that target the initiation step, and translation initiation at synapses could be a means of maintaining synapse specificity during plasticity. Initiation predominantly occurs via recruitment of ribosomes to the 5' mRNA cap by complexes of eukaryotic initiation factors (eIFs), and the interaction between eIF4E and eIF4G1 is a particularly important target of translational control pathways. Pharmacological inhibition of eIF4E-eIF4G1 binding impairs formation of memory for aversive Pavlovian conditioning as well as the accompanying increase in polyribosomes in the heads of dendritic spines in the lateral amygdala (LA). This is consistent with a role for initiation at synapses in memory formation, but whether eIFs are even present near synapses is unknown. To determine whether dendritic spines contain eIFs and whether eIF distribution is affected by learning, we combined immunolabeling with serial section transmission electron microscopy (ssTEM) volume reconstructions of LA dendrites after Pavlovian conditioning. Labeling for eIF4E, eIF4G1, and eIF2α-another key target of regulation-occurred in roughly half of dendritic spines, but learning effects were only found for eIF4E, which was upregulated in the heads of dendritic spines. Our results support the possibility of regulated translation initiation as a means of synapse-specific protein targeting during learning and are consistent with the model of eIF4E availability as a central point of control.
Collapse
Affiliation(s)
- Sofya Gindina
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, USA
| | - Benjamin Botsford
- Center for Neural Science, New York University, New York, New York, USA
| | - Kiriana Cowansage
- Center for Neural Science, New York University, New York, New York, USA
| | - Joseph LeDoux
- Center for Neural Science, New York University, New York, New York, USA.,Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, New York, USA
| | - Charles Hoeffer
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado, USA
| | - Linnaea Ostroff
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
39
|
Goto-Silva L, Junqueira M. Single-cell proteomics: A treasure trove in neurobiology. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140658. [PMID: 33845200 DOI: 10.1016/j.bbapap.2021.140658] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022]
Abstract
Single-cell analysis came to change the way we look at cell populations. RNA sequencing of single cells allowed us to appreciate the diversity of cell types in the human brain in an unprecedented manner and its power to reveal cell-type specific changes in cell populations has just begun to be explored. In this context, looking at the proteome of single cells promises to bring functional information and contribute to completing the picture. The potential of single cell proteome, in developing a better understanding of the intricate connections between the very diverse cell populations in the brain, is huge. Whereas early approaches to address single-cell proteome have identified hundreds of proteins, today, techniques combining isobaric labelling and LC-MS can lead to the identification of thousands of proteins. In this review, we describe methods which have been used to identify and quantify proteins from single cells and propose that the application of isobaric labeling and label-free quantitative proteomics approach for single-cell analysis is ready to provide useful information for the neurobiology field.
Collapse
Affiliation(s)
- Livia Goto-Silva
- D'Or Institute for Research and Education (IDOR), 22281-100 Rio de Janeiro, Brazil
| | - Magno Junqueira
- Proteomics Unit, Department of Biochemistry, Chemistry Institute, Federal University of Rio de janeiro, 21941-909 Rio de Janeiro, Brazil.
| |
Collapse
|
40
|
Environmental Enrichment Enhances Ca v 2.1 Channel-Mediated Presynaptic Plasticity in Hypoxic-Ischemic Encephalopathy. Int J Mol Sci 2021; 22:ijms22073414. [PMID: 33810296 PMCID: PMC8037860 DOI: 10.3390/ijms22073414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Hypoxic–ischemic encephalopathy (HIE) is a devastating neonatal brain condition caused by lack of oxygen and limited blood flow. Environmental enrichment (EE) is a classic paradigm with a complex stimulation of physical, cognitive, and social components. EE can exert neuroplasticity and neuroprotective effects in immature brains. However, the exact mechanism of EE on the chronic condition of HIE remains unclear. HIE was induced by a permanent ligation of the right carotid artery, followed by an 8% O2 hypoxic condition for 1 h. At 6 weeks of age, HIE mice were randomly assigned to either standard cages or EE cages. In the behavioral assessments, EE mice showed significantly improved motor performances in rotarod tests, ladder walking tests, and hanging wire tests, compared with HIE control mice. EE mice also significantly enhanced cognitive performances in Y-maze tests. Particularly, EE mice showed a significant increase in Cav 2.1 (P/Q type) and presynaptic proteins by molecular assessments, and a significant increase of Cav 2.1 in histological assessments of the cerebral cortex and hippocampus. These results indicate that EE can upregulate the expression of the Cav 2.1 channel and presynaptic proteins related to the synaptic vesicle cycle and neurotransmitter release, which may be responsible for motor and cognitive improvements in HIE.
Collapse
|
41
|
Pascual M, López‐Hidalgo R, Montagud‐Romero S, Ureña‐Peralta JR, Rodríguez‐Arias M, Guerri C. Role of mTOR-regulated autophagy in spine pruning defects and memory impairments induced by binge-like ethanol treatment in adolescent mice. Brain Pathol 2021; 31:174-188. [PMID: 32876364 PMCID: PMC8018167 DOI: 10.1111/bpa.12896] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Adolescence is a brain maturation developmental period during which remodeling and changes in synaptic plasticity and neural connectivity take place in some brain regions. Different mechanism participates in adolescent brain maturation, including autophagy that plays a role in synaptic development and plasticity. Alcohol is a neurotoxic compound and its abuse in adolescence induces neuroinflammation, synaptic and myelin alterations, neural damage and behavioral impairments. Changes in synaptic plasticity and its regulation by mTOR have also been suggested to play a role in the behavioral dysfunction of binge ethanol drinking in adolescence. Therefore, by considering the critical role of mTOR in both autophagy and synaptic plasticity in the developing brain, the present study aims to evaluate whether binge ethanol treatment in adolescence would induce dysfunctions in synaptic plasticity and cognitive functions and if mTOR inhibition with rapamycin is capable of restoring both effects. Using C57BL/6 adolescent female and male mice (PND30) treated with ethanol (3 g/kg) on two consecutive days at 48-hour intervals over 2 weeks, we show that binge ethanol treatment alters the density and morphology of dendritic spines, effects that are associated with learning and memory impairments and changes in the levels of both transcription factor CREB phosphorylation and miRNAs. Rapamycin administration (3 mg/kg) prior to ethanol administration restores ethanol-induced changes in both plasticity and behavior dysfunctions in adolescent mice. These results support the critical role of mTOR/autophagy dysfunctions in the dendritic spines alterations and cognitive alterations induced by binge alcohol in adolescence.
Collapse
Affiliation(s)
- María Pascual
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
- Department of PhysiologySchool of Medicine and DentistryUniversity of ValenciaValenciaSpain
| | - Rosa López‐Hidalgo
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
| | | | - Juan R. Ureña‐Peralta
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
| | | | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of AlcoholPríncipe Felipe Research CenterValenciaSpain
| |
Collapse
|
42
|
Ullah R, Ikram M, Park TJ, Ahmad R, Saeed K, Alam SI, Rehman IU, Khan A, Khan I, Jo MG, Kim MO. Vanillic Acid, a Bioactive Phenolic Compound, Counteracts LPS-Induced Neurotoxicity by Regulating c-Jun N-Terminal Kinase in Mouse Brain. Int J Mol Sci 2020; 22:ijms22010361. [PMID: 33396372 PMCID: PMC7795830 DOI: 10.3390/ijms22010361] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE), a pattern recognition receptor signaling event, has been associated with several human illnesses, including neurodegenerative diseases, particularly in Alzheimer’s disease (AD). Vanillic acid (V.A), a flavoring agent, is a benzoic acid derivative having a broad range of biological activities, including antioxidant, anti-inflammatory, and neuroprotective effects. However, the underlying molecular mechanisms of V.A in exerting neuroprotection are not well investigated. The present study aims to explore the neuroprotective effects of V.A against lipopolysaccharides (LPS)-induced neuroinflammation, amyloidogenesis, synaptic/memory dysfunction, and neurodegeneration in mice brain. Behavioral tests and biochemical and immunofluorescence assays were applied. Our results indicated increased expression of RAGE and its downstream phospho-c-Jun n-terminal kinase (p-JNK) in the LPS-alone treated group, which was significantly reduced in the V.A + LPS co-treated group. We also found that systemic administration of LPS-injection induced glial cells (microglia and astrocytes) activation and significantly increased expression level of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-KB) and secretion of proinflammatory cytokines including tumor necrosis factor alpha (TNF-α), interleukin-1 β (IL1-β), and cyclooxygenase (COX-2). However, V.A + LPS co-treatment significantly inhibited the LPS-induced activation of glial cells and neuroinflammatory mediators. Moreover, we also noted that V.A treatment significantly attenuated LPS-induced increases in the expression of AD markers, such as β-site amyloid precursor protein (APP)–cleaving enzyme 1 (BACE1) and amyloid-β (Aβ). Furthermore, V.A treatment significantly reversed LPS-induced synaptic loss via enhancing the expression level of pre- and post-synaptic markers (PSD-95 and SYP), and improved memory performance in LPS-alone treated group. Taken together; we suggest that neuroprotective effects of V.A against LPS-induced neurotoxicity might be via inhibition of LPS/RAGE mediated JNK signaling pathway; and encourage future studies that V.A would be a potential neuroprotective and neurotherapeutic candidate in various neurological disorders.
Collapse
Affiliation(s)
- Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Muhammad Ikram
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow G12OZD, UK;
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Kamran Saeed
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Sayed Ibrar Alam
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Amjad Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Ibrahim Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Min Gi Jo
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
43
|
Moradi Majd R, Mayeli M, Rahmani F. Pathogenesis and promising therapeutics of Alzheimer disease through eIF2α pathway and correspondent kinases. Metab Brain Dis 2020; 35:1241-1250. [PMID: 32681467 DOI: 10.1007/s11011-020-00600-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/05/2020] [Indexed: 01/10/2023]
Abstract
Eukaryotic initiation factor 2 (eIF2α) pathway is overactivated in Alzheimer disease and is probably associated with synaptic and memory deficiencies. EIF2α protein is principally in charge of the regulation of protein synthesis in eukaryotic cells. Four kinases responsible for eIF2α phosphorylation at ser-51 are: General control non-derepressible-2 kinase (GCN2), double-stranded RNA-activated protein kinase (PKR), PKR-like endoplasmic reticulum kinase (PERK), and heme-regulated inhibitor kinase (HRI) are the four kinases. They lead to reduced levels of general translation and paradoxical increase of stress-responsive mRNAs expression including the B-secretase (BACE1) and the transcriptional modulator activating transcription factor 4 (ATF4), which in turn accelerates the beta-amyloidogenesis, tau phosphorylation, proapoptotic pathway induction and autophagy elements formation leading to the main pathological hallmarks of AD. Findings suggest that genetic or pharmacological inhibition of correspondent kinases can restore memory and prevent neurodegeneration. This implies that inhibition of eIF2α phosphorylation through respondent kinases is indeed a feasible prospect of clinical application. This review discusses recent therapeutic approaches targeting eIF2α pathway and provides an overview of the links between correspondent kinases overactivation with neurodegeneration in AD.
Collapse
Affiliation(s)
- Reza Moradi Majd
- Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Mahsa Mayeli
- Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Farzaneh Rahmani
- Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| |
Collapse
|
44
|
Frank MG. The Ontogenesis of Mammalian Sleep: Form and Function. CURRENT SLEEP MEDICINE REPORTS 2020; 6:267-279. [PMID: 33816063 PMCID: PMC8014960 DOI: 10.1007/s40675-020-00190-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW To present an up-to-date review and synthesis of findings about perinatal sleep development and function. I discuss landmark events in sleep ontogenesis, evidence that sleep promotes brain development and plasticity, and experimental considerations in this topic. RECENT FINDINGS Mammalian sleep undergoes dramatic changes in expression and regulation during perinatal development. This includes a progressive decrease in rapid-eye-movement (REM) sleep time, corresponding increases in nonREM sleep and wake time, and the appearance of mature sleep regulatory processes (homeostatic and circadian). These developmental events coincide with periods of rapid brain maturation and heightened synaptic plasticity. The latter involve an initial experience-independent phase, when circuit development is guided by spontaneous activity, and later occurring critical periods, when these circuits are shaped by experience. SUMMARY These ontogenetic changes suggest important interactions between sleep and brain development. More specifically, sleep may promote developmental programs of synaptogenesis and synaptic pruning and influence the opening and closing of critical periods of brain plasticity.
Collapse
Affiliation(s)
- Marcos G Frank
- Washington State University Spokane, Elson S. Floyd College of Medicine, Pharmaceutical and Biomedical Science Building 213, 412 E. Spokane Falls Blvd
| |
Collapse
|
45
|
Dopamine-Dependent QR2 Pathway Activation in CA1 Interneurons Enhances Novel Memory Formation. J Neurosci 2020; 40:8698-8714. [PMID: 33046554 DOI: 10.1523/jneurosci.1243-20.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 01/18/2023] Open
Abstract
The formation of memory for a novel experience is a critical cognitive capacity. The ability to form novel memories is sensitive to age-related pathologies and disease, to which prolonged metabolic stress is a major contributing factor. Presently, we describe a dopamine-dependent redox modulation pathway within the hippocampus of male mice that promotes memory consolidation. Namely, following novel information acquisition, quinone reductase 2 (QR2) is suppressed by miRNA-182 (miR-182) in the CA1 region of the hippocampus via dopamine D1 receptor (D1R) activation, a process largely facilitated by locus coeruleus activity. This pathway activation reduces ROS generated by QR2 enzymatic activity, a process that alters the intrinsic properties of CA1 interneurons 3 h following learning, in a form of oxidative eustress. Interestingly, novel experience decreases QR2 expression predominately in inhibitory interneurons. Additionally, we find that in aged animals this newly described QR2 pathway is chronically under activated, resulting in miR-182 underexpression and QR2 overexpression. This leads to accumulative oxidative stress, which can be seen in CA1 via increased levels of oxidized, inactivated potassium channel Kv2.1, which undergoes disulfide bridge oligomerization. This newly described interneuron-specific molecular pathway lies alongside the known mRNA translation-dependent processes necessary for long-term memory formation, entrained by dopamine in CA1. It is a process crucial for the distinguishing features of novel memory, and points to a promising new target for memory enhancement in aging and age-dependent diseases.SIGNIFICANCE STATEMENT One way in which evolution dictates which sensory information will stabilize as an internal representation, relies on information novelty. Dopamine is a central neuromodulator involved in this process in the mammalian hippocampus. Here, we describe for the first time a dopamine D1 receptor-dependent quinone reductase 2 pathway in interneurons. This is a targeted redox event necessary to delineate a novel experience to a robust long-term internal representation. Activation of this pathway alone can explain the effect novelty has on "flashbulb" memories, and it can become dysfunctional with age and diseases, such as Alzheimer's disease.
Collapse
|
46
|
Neuronal Localization of SENP Proteins with Super Resolution Microscopy. Brain Sci 2020; 10:brainsci10110778. [PMID: 33113832 PMCID: PMC7693135 DOI: 10.3390/brainsci10110778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 02/03/2023] Open
Abstract
SUMOylation of proteins plays a key role in modulating neuronal function. For this reason, the balance between protein SUMOylation and deSUMOylation requires fine regulation to guarantee the homeostasis of neural tissue. While extensive research has been carried out on the localization and function of small ubiquitin-related modifier (SUMO) variants in neurons, less attention has been paid to the SUMO-specific isopeptidases that constitute the human SUMO-specific isopeptidase (SENP)/Ubiquitin-Specific Protease (ULP) cysteine protease family (SENP1-3 and SENP5-7). Here, for the first time, we studied the localization of SENP1, SENP6, and SENP7 in cultured hippocampal primary neurons at a super resolution detail level, with structured illumination microscopy (SIM). We found that the deSUMOylases partially colocalize with pre- and post-synaptic markers such as synaptophysin and drebrin. Thus, further confirming the presence with synaptic markers of the negative regulators of the SUMOylation machinery.
Collapse
|
47
|
Khlebodarova TM, Kogai VV, Likhoshvai VA. On the dynamical aspects of local translation at the activated synapse. BMC Bioinformatics 2020; 21:258. [PMID: 32921299 PMCID: PMC7488754 DOI: 10.1186/s12859-020-03597-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 06/10/2020] [Indexed: 02/01/2023] Open
Abstract
Background The key role in the dynamic regulation of synaptic protein turnover belongs to the Fragile X Mental Retardation Protein, which regulates the efficiency of dendritic mRNA translation in response to stimulation of metabotropic glutamate receptors at excitatory synapses of the hippocampal pyramidal cells. Its activity is regulated via positive and negative regulatory loops that function in different time ranges, which is an absolute factor for the formation of chaotic regimes that lead to disrupted proteome stability. The indicated condition may cause a number of neuropsychiatric diseases, including autism and epilepsy. The present study is devoted to a theoretical analysis of the local translation system dynamic properties and identification of parameters affecting the chaotic potential of the system. Results A mathematical model that describes the maintenance of a specific pool of active receptors on the postsynaptic membrane via two mechanisms – de novo synthesis of receptor proteins and restoration of protein function during the recycling process – has been developed. Analysis of the model revealed that an increase in the values of the parameters describing the impact of protein recycling on the maintenance of a pool of active receptors in the membrane, duration of the signal transduction via the mammalian target of rapamycin pathway, influence of receptors on the translation activation, as well as reduction of the rate of synthesis and integration of de novo synthesized proteins into the postsynaptic membrane – contribute to the reduced complexity of the local translation system dynamic state. Formation of these patterns significantly depends on the complexity and non-linearity of the mechanisms of exposure of de novo synthesized receptors to the postsynaptic membrane, the correct evaluation of which is currently problematic. Conclusions The model predicts that an increase of “receptor recycling” and reduction of the rate of synthesis and integration of de novo synthesized proteins into the postsynaptic membrane contribute to the reduced complexity of the local translation system dynamic state. Herewith, stable stationary states occur much less frequently than cyclic states. It is possible that cyclical nature of functioning of the local translation system is its “normal” dynamic state.
Collapse
Affiliation(s)
- Tamara M Khlebodarova
- Department of Systems Biology, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia. .,Novosibirsk State University, Novosibirsk, 630090, Russia.
| | | | - Vitaly A Likhoshvai
- Department of Systems Biology, Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| |
Collapse
|
48
|
McCarberg B, Peppin J. Pain Pathways and Nervous System Plasticity: Learning and Memory in Pain. PAIN MEDICINE 2020; 20:2421-2437. [PMID: 30865778 DOI: 10.1093/pm/pnz017] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective This article reviews the structural and functional changes in pain chronification and explores the association between memory and the development of chronic pain. Methods PubMed was searched using the terms "chronic pain," "central sensitization," "learning," "memory," "long-term potentiation," "long-term depression," and "pain memory." Relevant findings were synthesized into a narrative of the processes affecting pain chronification. Results Pain pathways represent a complex sensory system with cognitive, emotional, and behavioral influences. Anatomically, the hippocampus, amygdala, and anterior cortex-central to the encoding and consolidation of memory-are also implicated in experiential aspects of pain. Common neurotransmitters and similar mechanisms of neural plasticity (eg, central sensitization, long-term potentiation) suggest a mechanistic overlap between chronic pain and memory. These anatomic and mechanistic correlates indicate that chronic pain and memory intimately interact on several levels. Longitudinal imaging studies suggest that spatiotemporal reorganization of brain activity accompanies the transition to chronic pain, during which the representation of pain gradually shifts from sensory to emotional and limbic structures. Conclusions The chronification of pain can be conceptualized as activity-induced plasticity of the limbic-cortical circuitry resulting in reorganization of the neocortex. The state of the limbic-cortical network determines whether nociceptive signals are transient or chronic by extinguishing pathways or amplifying signals that intensify the emotional component of nociceptive inputs. Thus, chronic pain can be seen as the persistence of the memory of pain and/or the inability to extinguish painful memories. Ideally, pharmacologic, physical, and/or psychological approaches should reverse the reorganization accompanying chronic pain.
Collapse
Affiliation(s)
- Bill McCarberg
- Chronic Pain Management Program, Kaiser Permanente, San Diego, California; †University of California, San Diego, California; ‡Neighborhood Health, San Diego, California; §College of Osteopathic Medicine, Marian University, Indianapolis, Indiana; ¶John F. Peppin, DO, LLC, Hamden, Connecticut
| | - John Peppin
- Chronic Pain Management Program, Kaiser Permanente, San Diego, California; †University of California, San Diego, California; ‡Neighborhood Health, San Diego, California; §College of Osteopathic Medicine, Marian University, Indianapolis, Indiana; ¶John F. Peppin, DO, LLC, Hamden, Connecticut
| |
Collapse
|
49
|
Hernández RB, Carrascal M, Abian J, Michalke B, Farina M, Gonzalez YR, Iyirhiaro GO, Moteshareie H, Burnside D, Golshani A, Suñol C. Manganese-induced neurotoxicity in cerebellar granule neurons due to perturbation of cell network pathways with potential implications for neurodegenerative disorders. Metallomics 2020; 12:1656-1678. [PMID: 33206086 DOI: 10.1039/d0mt00085j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Manganese (Mn) is essential for living organisms, playing an important role in nervous system function. Nevertheless, chronic and/or acute exposure to this metal, especially during early life stages, can lead to neurotoxicity and dementia by unclear mechanisms. Thus, based on previous works of our group with yeast and zebrafish, we hypothesized that the mechanisms mediating manganese-induced neurotoxicity can be associated with the alteration of protein metabolism. These mechanisms may also depend on the chemical speciation of manganese. Therefore, the current study aimed at investigating the mechanisms mediating the toxic effects of manganese in primary cultures of cerebellar granule neurons (CGNs). By exposing cultured CGNs to different chemical species of manganese ([[2-[(dithiocarboxy)amino]ethyl]carbamodithioato]](2-)-kS,kS']manganese, named maneb (MB), and [[1,2-ethanediylbis[carbamodithioato]](2-)]manganese mixture with [[1,2-ethanediylbis[carbamodithioato]](2-)]zinc, named mancozeb (MZ), and manganese chloride (MnCl2)), and using the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, we observed that both MB and MZ induced similar cytotoxicity (LC50∼ 7-9 μM), which was higher than that of MnCl2 (LC50∼ 27 μM). Subsequently, we applied systems biology approaches, including metallomics, proteomics, gene expression and bioinformatics, and revealed that independent of chemical speciation, for non-cytotoxic concentrations (0.3-3 μM), Mn-induced neurotoxicity in CGNs is associated with metal dyshomeostasis and impaired protein metabolism. In this way, we verified that MB induced more post-translational alterations than MnCl2, which can be a plausible explanation for cytotoxic differences between both chemical species. The metabolism of proteins is one of the most energy consuming cellular processes and its impairment appears to be a key event of some cellular stress processes reported separately in other studies such as cell cycle arrest, energy impairment, cell signaling, excitotoxicity, immune response, potential protein accumulation and apoptosis. Interestingly, we verified that Mn-induced neurotoxicity shares pathways associated with the development of Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, and Parkinson's disease. This has been observed in baker's yeast and zebrafish suggesting that the mode of action of Mn may be evolutionarily conserved.
Collapse
Affiliation(s)
- Raúl Bonne Hernández
- Laboratory of Bioinorganic and Environmental Toxicology - LABITA, Department of Exact and Earth Sciences, Federal University of São Paulo, Rua Prof. Artur Riedel, 275, CEP 09972-270, Diadema, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Akintunde JK, Akintola TE, Adenuga GO, Odugbemi ZA, Adetoye RO, Akintunde OG. Naringin attenuates Bisphenol-A mediated neurotoxicity in hypertensive rats by abrogation of cerebral nucleotide depletion, oxidative damage and neuroinflammation. Neurotoxicology 2020; 81:18-33. [PMID: 32810514 DOI: 10.1016/j.neuro.2020.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
We examined whether active fruit naringin can reduce the risk of BPA-mediated neurotoxicity in L-NAME induced hypertensive rats and whether the modulation could be linked to improvement of brain NO signaling. Male albino rats were randomly distributed into eight (n = 7) groups. Group I was control animals, Group II was orally-treated with L-NAME, Group III was orally treated with 100 mg/kg BPA, Group IV was orally-treated with L-NAME +100 mg/kg BPA. Group V was orally-administered with L-NAME +80 mg/kg NAR. Group VI was orally-administered with 100 mg/kg BPA +80 mg/kg NAR. Group VII was orally-administered with L-NAME+100 mg/kg BPA +80 mg/kg NAR. Lastly, group VIII was orally-treated with 80 mg/kg NAR. The treatment lasted for 14 days. Sub-acute exposure to L-NAME and BPA induced hypertension and mediated-neuroinflammation at CA-2 and CA-4 of hippocampus cells. It was evident by increase in PDE-51 and enzymes of ATP hydrolysis (ATPase, ADPase and AMPase) with corresponding upsurge in cholinergic (AChE and BuChE), dopaminergic (MAO-A) and adenosinergic (ADA) enzymes as well as movement disorder. The hypertensive-mediated neurotoxicity was related to alteration of NO signaling and higher release of pro-inflammatory cytokines (TNF-α and IL-1β), apoptotic proteins (P53 and caspace-9) and facilitated entry of T-lymphocytes (CD43+) into CNS through blood brain barrier potentiated by antigen presenting cells. Hence, these features of BPA-mediated neurotoxicity in L-NAME induced hypertensive rats were prohibited by co-administration of NAR through production of neuro-inflammatory mediators, stabilizing neurotransmitter enzymes, normalizing NO signaling and improving brain histology.
Collapse
Affiliation(s)
- J K Akintunde
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria.
| | - T E Akintola
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - G O Adenuga
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Z A Odugbemi
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria; Department of Physiology and Biochemistry, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| | - R O Adetoye
- Applied Biochemistry and Molecular Toxicology Research Group, Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - O G Akintunde
- Department of Physiology and Biochemistry, College of Veterinary Medicine, Federal University of Agriculture, Abeokuta, Nigeria
| |
Collapse
|