51
|
The Epigenetics of Aging in Invertebrates. Int J Mol Sci 2019; 20:ijms20184535. [PMID: 31540238 PMCID: PMC6769462 DOI: 10.3390/ijms20184535] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/03/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022] Open
Abstract
Aging is an unstoppable process coupled to the loss of physiological function and increased susceptibility to diseases. Epigenetic alteration is one of the hallmarks of aging, which involves changes in DNA methylation patterns, post-translational modification of histones, chromatin remodeling and non-coding RNA interference. Invertebrate model organisms, such as Drosophila melanogaster and Caenorhabditis elegans, have been used to investigate the biological mechanisms of aging because they show, evolutionarily, the conservation of many aspects of aging. In this review, we focus on recent advances in the epigenetic changes of aging with invertebrate models, providing insight into the relationship between epigenetic dynamics and aging.
Collapse
|
52
|
Wang G, Zhong J, Guttieres D, Man HY. Non-scaling regulation of AMPA receptors in homeostatic synaptic plasticity. Neuropharmacology 2019; 158:107700. [PMID: 31283924 DOI: 10.1016/j.neuropharm.2019.107700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/10/2019] [Accepted: 07/04/2019] [Indexed: 11/28/2022]
Abstract
Homeostatic synaptic plasticity (HSP) as an activity-dependent negative feedback regulation of synaptic strength plays important roles in the maintenance of neuronal and neural circuitry stability. A primary cellular substrate for HSP expression is alterations in synaptic accumulation of glutamatergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR). It is widely believed that during HSP, AMPAR accumulation changes with the same proportion at each synapse of a neuron, a process known as synaptic scaling. However, direct evidence on AMPAR synaptic scaling remains largely lacking. Here we report a direct examination of inactivity-induced homeostatic scaling of AMPAR at individual synapse by live-imaging. Surprisingly, instead of uniform up-scaling, a scattered pattern of changes in synaptic AMPAR was observed in cultured rat hippocampal neurons. While the majority of synapses showed up-regulation after activity inhibition, a reduction of AMPAR could be detected in certain synapses. More importantly, among the up-regulated synapses, a wide range of AMPAR changes was observed in synapses of the same neuron. We also found that synapses with higher levels of pre-existing AMPAR tend to be up-regulated by lesser extents, whereas the locations of synapses relative to the soma seem not affecting AMPAR scaling strengths. In addition, we observed strong competition between neighboring synapses during HSP. These results reveal that synaptic AMPAR may not be scaled during HSP, suggesting novel molecular mechanisms for information processing and storage at synapses.
Collapse
Affiliation(s)
- Guan Wang
- Department of Biology, Boston University, Boston, MA, USA; School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jia Zhong
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA; Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
53
|
MicroRNA-186-5p controls GluA2 surface expression and synaptic scaling in hippocampal neurons. Proc Natl Acad Sci U S A 2019; 116:5727-5736. [PMID: 30808806 DOI: 10.1073/pnas.1900338116] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Homeostatic synaptic scaling is a negative feedback response to fluctuations in synaptic strength induced by developmental or learning-related processes, which maintains neuronal activity stable. Although several components of the synaptic scaling apparatus have been characterized, the intrinsic regulatory mechanisms promoting scaling remain largely unknown. MicroRNAs may contribute to posttranscriptional control of mRNAs implicated in different stages of synaptic scaling, but their role in these mechanisms is still undervalued. Here, we report that chronic blockade of glutamate receptors of the AMPA and NMDA types in hippocampal neurons in culture induces changes in the neuronal mRNA and miRNA transcriptomes, leading to synaptic upscaling. Specifically, we show that synaptic activity blockade persistently down-regulates miR-186-5p. Moreover, we describe a conserved miR-186-5p-binding site within the 3'UTR of the mRNA encoding the AMPA receptor GluA2 subunit, and demonstrate that GluA2 is a direct target of miR-186-5p. Overexpression of miR-186 decreased GluA2 surface levels, increased synaptic expression of GluA2-lacking AMPA receptors, and blocked synaptic scaling, whereas inhibition of miR-186-5p increased GluA2 surface levels and the amplitude and frequency of AMPA receptor-mediated currents, and mimicked excitatory synaptic scaling induced by synaptic inactivity. Our findings elucidate an activity-dependent miRNA-mediated mechanism for regulation of AMPA receptor expression.
Collapse
|
54
|
Zampa F, Hartzell AL, Zolboot N, Lippi G. Non-coding RNAs: the gatekeepers of neural network activity. Curr Opin Neurobiol 2019; 57:54-61. [PMID: 30743177 DOI: 10.1016/j.conb.2019.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Non-coding RNAs have emerged as potent regulators of numerous cellular processes. In neurons and circuits, these molecules serve especially critical functions that ensure neural activity is maintained within appropriate physiological parameters. Their targets include synaptic proteins, ion channels, neurotransmitter receptors, and components of essential signaling cascades. Here, we discuss how several species of non-coding RNAs (ncRNAs) regulate intrinsic excitability and synaptic transmission, both during development and in mature circuits. Furthermore, we present the relationships between aberrant ncRNA expression and psychiatric disorders. The research presented here demonstrates how ncRNAs can be useful tools for elucidating fundamental neurobiology mechanisms and identifying the key molecular players.
Collapse
Affiliation(s)
- Federico Zampa
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Andrea L Hartzell
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Norjin Zolboot
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Giordano Lippi
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
55
|
Rocchi A, Moretti D, Lignani G, Colombo E, Scholz-Starke J, Baldelli P, Tkatch T, Benfenati F. Neurite-Enriched MicroRNA-218 Stimulates Translation of the GluA2 Subunit and Increases Excitatory Synaptic Strength. Mol Neurobiol 2019; 56:5701-5714. [DOI: 10.1007/s12035-019-1492-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022]
|
56
|
Kutsche LK, Gysi DM, Fallmann J, Lenk K, Petri R, Swiersy A, Klapper SD, Pircs K, Khattak S, Stadler PF, Jakobsson J, Nowick K, Busskamp V. Combined Experimental and System-Level Analyses Reveal the Complex Regulatory Network of miR-124 during Human Neurogenesis. Cell Syst 2018; 7:438-452.e8. [PMID: 30292704 PMCID: PMC6205824 DOI: 10.1016/j.cels.2018.08.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/12/2018] [Accepted: 08/23/2018] [Indexed: 02/07/2023]
Abstract
Non-coding RNAs regulate many biological processes including neurogenesis. The brain-enriched miR-124 has been assigned as a key player of neuronal differentiation via its complex but little understood regulation of thousands of annotated targets. To systematically chart its regulatory functions, we used CRISPR/Cas9 gene editing to disrupt all six miR-124 alleles in human induced pluripotent stem cells. Upon neuronal induction, miR-124-deleted cells underwent neurogenesis and became functional neurons, albeit with altered morphology and neurotransmitter specification. Using RNA-induced-silencing-complex precipitation, we identified 98 high-confidence miR-124 targets, of which some directly led to decreased viability. By performing advanced transcription-factor-network analysis, we identified indirect miR-124 effects on apoptosis, neuronal subtype differentiation, and the regulation of previously uncharacterized zinc finger transcription factors. Our data emphasize the need for combined experimental- and system-level analyses to comprehensively disentangle and reveal miRNA functions, including their involvement in the neurogenesis of diverse neuronal cell types found in the human brain. miR-124 is not essential for neurogenesis from human iPSCs miR-124 regulation mediates neuroprotection and refines neuronal cell fates miRNA knockout characterization by experimental and advanced computational analyses Identification of 98 targets including the neuronal feature repressor ZNF787
Collapse
Affiliation(s)
- Lisa K Kutsche
- Technische Universität Dresden, DFG Research Center for Regenerative Therapies, Dresden 01307, Germany
| | - Deisy M Gysi
- Department of Computer Science, Bioinformatics Group, Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig 04107, Germany; Faculty of Mathematics and Computer Science, Swarm Intelligence and Complex Systems Group, University of Leipzig, Leipzig 04109, Germany; Faculty for Biology, Chemistry and Pharmacy, Freie Universität Berlin, Institute for Biology, Berlin 14195, Germany
| | - Joerg Fallmann
- Department of Computer Science, Bioinformatics Group, Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig 04107, Germany
| | - Kerstin Lenk
- Technische Universität Dresden, DFG Research Center for Regenerative Therapies, Dresden 01307, Germany
| | - Rebecca Petri
- Department of Experimental Medical Science, Laboratory of Molecular Neurogenetics, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lunds Universitet, Lund 22184, Sweden
| | - Anka Swiersy
- Technische Universität Dresden, DFG Research Center for Regenerative Therapies, Dresden 01307, Germany
| | - Simon D Klapper
- Technische Universität Dresden, DFG Research Center for Regenerative Therapies, Dresden 01307, Germany
| | - Karolina Pircs
- Department of Experimental Medical Science, Laboratory of Molecular Neurogenetics, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lunds Universitet, Lund 22184, Sweden
| | - Shahryar Khattak
- Technische Universität Dresden, DFG Research Center for Regenerative Therapies, Dresden 01307, Germany
| | - Peter F Stadler
- Department of Computer Science, Bioinformatics Group, Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig 04107, Germany; Max Planck Institute for Mathematics in the Sciences, Leipzig 04103, Germany; Santa Fe Institute, 1399 Hyde Park Road, Santa Fe, NM 87501, USA
| | - Johan Jakobsson
- Department of Experimental Medical Science, Laboratory of Molecular Neurogenetics, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lunds Universitet, Lund 22184, Sweden
| | - Katja Nowick
- Faculty for Biology, Chemistry and Pharmacy, Freie Universität Berlin, Institute for Biology, Berlin 14195, Germany
| | - Volker Busskamp
- Technische Universität Dresden, DFG Research Center for Regenerative Therapies, Dresden 01307, Germany.
| |
Collapse
|
57
|
Walker SE, Spencer GE, Necakov A, Carlone RL. Identification and Characterization of microRNAs during Retinoic Acid-Induced Regeneration of a Molluscan Central Nervous System. Int J Mol Sci 2018; 19:E2741. [PMID: 30217012 PMCID: PMC6163488 DOI: 10.3390/ijms19092741] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/06/2018] [Accepted: 09/08/2018] [Indexed: 12/11/2022] Open
Abstract
Retinoic acid (RA) is the biologically active metabolite of vitamin A and has become a well-established factor that induces neurite outgrowth and regeneration in both vertebrates and invertebrates. However, the underlying regulatory mechanisms that may mediate RA-induced neurite sprouting remain unclear. In the past decade, microRNAs have emerged as important regulators of nervous system development and regeneration, and have been shown to contribute to processes such as neurite sprouting. However, few studies have demonstrated the role of miRNAs in RA-induced neurite sprouting. By miRNA sequencing analysis, we identify 482 miRNAs in the regenerating central nervous system (CNS) of the mollusc Lymnaeastagnalis, 219 of which represent potentially novel miRNAs. Of the remaining conserved miRNAs, 38 show a statistically significant up- or downregulation in regenerating CNS as a result of RA treatment. We further characterized the expression of one neuronally-enriched miRNA upregulated by RA, miR-124. We demonstrate, for the first time, that miR-124 is expressed within the cell bodies and neurites of regenerating motorneurons. Moreover, we identify miR-124 expression within the growth cones of cultured ciliary motorneurons (pedal A), whereas expression in the growth cones of another class of respiratory motorneurons (right parietal A) was absent in vitro. These findings support our hypothesis that miRNAs are important regulators of retinoic acid-induced neuronal outgrowth and regeneration in regeneration-competent species.
Collapse
Affiliation(s)
- Sarah E Walker
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| | - Aleksandar Necakov
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| | - Robert L Carlone
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
58
|
Lang WJ, Chen FY. The reciprocal link between EVI1 and miRNAs in human malignancies. Gene 2018; 672:56-63. [DOI: 10.1016/j.gene.2018.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 03/05/2018] [Accepted: 06/03/2018] [Indexed: 12/26/2022]
|
59
|
Corrà F, Agnoletto C, Minotti L, Baldassari F, Volinia S. The Network of Non-coding RNAs in Cancer Drug Resistance. Front Oncol 2018; 8:327. [PMID: 30211115 PMCID: PMC6123370 DOI: 10.3389/fonc.2018.00327] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Non-coding RNAs (ncRNAs) have been implicated in most cellular functions. The disruption of their function through somatic mutations, genomic imprinting, transcriptional and post-transcriptional regulation, plays an ever-increasing role in cancer development. ncRNAs, including notorious microRNAs, have been thus proposed to function as tumor suppressors or oncogenes, often in a context-dependent fashion. In parallel, ncRNAs with altered expression in cancer have been reported to exert a key role in determining drug sensitivity or restoring drug responsiveness in resistant cells. Acquisition of resistance to anti-cancer drugs is a major hindrance to effective chemotherapy and is one of the most important causes of relapse and mortality in cancer patients. For these reasons, non-coding RNAs have become recent focuses as prognostic agents and modifiers of chemo-sensitivity. This review starts with a brief outline of the role of most studied non-coding RNAs in cancer and then highlights the modulation of cancer drug resistance via known ncRNAs based mechanisms. We identified from literature 388 ncRNA-drugs interactions and analyzed them using an unsupervised approach. Essentially, we performed a network analysis of the non-coding RNAs with direct relations with cancer drugs. Within such a machine-learning framework we detected the most representative ncRNAs-drug associations and groups. We finally discussed the higher integration of the drug-ncRNA clusters with the goal of disentangling effectors from downstream effects and further clarify the involvement of ncRNAs in the cellular mechanisms underlying resistance to cancer treatments.
Collapse
Affiliation(s)
- Fabio Corrà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Chiara Agnoletto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Linda Minotti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Federica Baldassari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
60
|
Gomes C, Cunha C, Nascimento F, Ribeiro JA, Vaz AR, Brites D. Cortical Neurotoxic Astrocytes with Early ALS Pathology and miR-146a Deficit Replicate Gliosis Markers of Symptomatic SOD1G93A Mouse Model. Mol Neurobiol 2018; 56:2137-2158. [DOI: 10.1007/s12035-018-1220-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/29/2018] [Indexed: 12/13/2022]
|
61
|
Loera-Valencia R, Piras A, Ismail MAM, Manchanda S, Eyjolfsdottir H, Saido TC, Johansson J, Eriksdotter M, Winblad B, Nilsson P. Targeting Alzheimer's disease with gene and cell therapies. J Intern Med 2018; 284:2-36. [PMID: 29582495 DOI: 10.1111/joim.12759] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) causes dementia in both young and old people affecting more than 40 million people worldwide. The two neuropathological hallmarks of the disease, amyloid beta (Aβ) plaques and neurofibrillary tangles consisting of protein tau are considered the major contributors to the disease. However, a more complete picture reveals significant neurodegeneration and decreased cell survival, neuroinflammation, changes in protein and energy homeostasis and alterations in lipid and cholesterol metabolism. In addition, gene and cell therapies for severe neurodegenerative disorders have recently improved technically in terms of safety and efficiency and have translated to the clinic showing encouraging results. Here, we review broadly current data within the field for potential targets that could modify AD through gene and cell therapy strategies. We envision that not only Aβ will be targeted in a disease-modifying treatment strategy but rather that a combination of treatments, possibly at different intervention times may prove beneficial in curing this devastating disease. These include decreased tau pathology, neuronal growth factors to support neurons and modulation of neuroinflammation for an appropriate immune response. Furthermore, cell based therapies may represent potential strategies in the future.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - A Piras
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M A M Ismail
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Neuro, Diseases of the Nervous System Patient Flow, Karolinska University Hospital, Huddinge, Sweden
| | - S Manchanda
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - H Eyjolfsdottir
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - T C Saido
- RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - J Johansson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - P Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
62
|
Chowdhury D, Hell JW. Homeostatic synaptic scaling: molecular regulators of synaptic AMPA-type glutamate receptors. F1000Res 2018; 7:234. [PMID: 29560257 PMCID: PMC5832907 DOI: 10.12688/f1000research.13561.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/20/2018] [Indexed: 01/31/2023] Open
Abstract
The ability of neurons and circuits to maintain their excitability and activity levels within the appropriate dynamic range by homeostatic mechanisms is fundamental for brain function. Neuronal hyperactivity, for instance, could cause seizures. One such homeostatic process is synaptic scaling, also known as synaptic homeostasis. It involves a negative feedback process by which neurons adjust (scale) their postsynaptic strength over their whole synapse population to compensate for increased or decreased overall input thereby preventing neuronal hyper- or hypoactivity that could otherwise result in neuronal network dysfunction. While synaptic scaling is well-established and critical, our understanding of the underlying molecular mechanisms is still in its infancy. Homeostatic adaptation of synaptic strength is achieved through upregulation (upscaling) or downregulation (downscaling) of the functional availability of AMPA-type glutamate receptors (AMPARs) at postsynaptic sites. Understanding how synaptic AMPARs are modulated in response to alterations in overall neuronal activity is essential to gain valuable insights into how neuronal networks adapt to changes in their environment, as well as the genesis of an array of neurological disorders. Here we discuss the key molecular mechanisms that have been implicated in tuning the synaptic abundance of postsynaptic AMPARs in order to maintain synaptic homeostasis.
Collapse
Affiliation(s)
| | - Johannes W Hell
- Department of Pharmacology, University of California Davis, Davis, California, USA
| |
Collapse
|
63
|
Miao N, Jin J, Kim SN, Sun T. Hippocampal MicroRNAs Respond to Administration of Antidepressant Fluoxetine in Adult Mice. Int J Mol Sci 2018; 19:ijms19030671. [PMID: 29495532 PMCID: PMC5877532 DOI: 10.3390/ijms19030671] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/23/2018] [Accepted: 02/24/2018] [Indexed: 12/13/2022] Open
Abstract
Current antidepressant treatments to anxiety and depression remain inadequate, burdened by a significant percentage of misuse and drug side-effects, due to unclear mechanisms of actions of antidepressants. To better understand the regulatory roles of antidepressant fluoxetine-related drug reactions, we here investigate changes of expression levels of hippocampal microRNAs (miRNAs) after administration of fluoxetine in normal adult mice. We find that 64 miRNAs showed significant changes between fluoxetine treatment and control groups by analyzing 626 mouse miRNAs. Many miRNAs in response to fluoxetine are involved in neural-related signaling pathways by analyzing miRNA-target gene pairs using the Kyoto encyclopedia of genes and genomes (KEGG) and Gene Ontology (GO). Moreover, miRNAs with altered expression are mainly associated with the repression of the dopaminergic synapse signals, which may affect hippocampal function after fluoxetine treatment. Our results demonstrate that a number of miRNAs respond to antidepressants even in normal mice and may affect target gene expression, which supports the safety consideration of inappropriate treatment and off-label use of antidepressant drugs.
Collapse
Affiliation(s)
- Nan Miao
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen 361021, China.
| | - Junghee Jin
- Department of Cell and Developmental Biology, Cornell University Weill Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA.
| | - Seung-Nam Kim
- Department of Cell and Developmental Biology, Cornell University Weill Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA.
- College of Korean Medicine, Dongguk University, Ilsandonggu, Goyangsi 10326, Gyeonggido, Korea.
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen 361021, China.
- Department of Cell and Developmental Biology, Cornell University Weill Medical College, 1300 York Avenue, Box 60, New York, NY 10065, USA.
| |
Collapse
|
64
|
Gillet V, Hunting DJ, Takser L. Turing Revisited: Decoding the microRNA Messages in Brain Extracellular Vesicles for Early Detection of Neurodevelopmental Disorders. Curr Environ Health Rep 2018; 3:188-201. [PMID: 27301443 DOI: 10.1007/s40572-016-0093-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The prevention of neurodevelopmental disorders (NDD) of prenatal origin suffers from the lack of objective tools for early detection of susceptible individuals and the long time lag, usually in years, between the neurotoxic exposure and the diagnosis of mental dysfunction. Human data on the effects of alcohol, lead, and mercury and experimental data from animals on developmental neurotoxins and their long-term behavioral effects have achieved a critical mass, leading to the concept of the Developmental Origin of Health and Disease (DOHaD). However, there is currently no way to evaluate the degree of brain damage early after birth. We propose that extracellular vesicles (EVs) and particularly exosomes, released by brain cells into the fetal blood, may offer us a non-invasive means of assessing brain damage by neurotoxins. We are inspired by the strategy applied by Alan Turing (a cryptanalyst working for the British government), who created a first computer to decrypt German intelligence communications during World War II. Given the growing evidence that microRNAs (miRNAs), which are among the molecules carried by EVs, are involved in cell-cell communication, we propose that decrypting messages from EVs can allow us to detect damage thus offering an opportunity to cure, reverse, or prevent the development of NDD. This review summarizes recent findings on miRNAs associated with selected environmental toxicants known to be involved in the pathophysiology of NDD.
Collapse
Affiliation(s)
- Virginie Gillet
- Département Pédiatrie, Faculté de Médecine et Sciences de la Santé de l'Université de Sherbrooke, 3001, 12ème avenue Nord, Sherbrooke, Québec, Canada, J1H 5N4
| | - Darel John Hunting
- Département Radiobiologie, Faculté de Médecine et Sciences de la Santé de l'Université de Sherbrooke, 3001, 12ème avenue Nord, Sherbrooke, Québec, Canada, J1H 5N4
| | - Larissa Takser
- Département Pédiatrie, Faculté de Médecine et Sciences de la Santé de l'Université de Sherbrooke, 3001, 12ème avenue Nord, Sherbrooke, Québec, Canada, J1H 5N4.
| |
Collapse
|
65
|
Gilbert J, Man HY. Fundamental Elements in Autism: From Neurogenesis and Neurite Growth to Synaptic Plasticity. Front Cell Neurosci 2017; 11:359. [PMID: 29209173 PMCID: PMC5701944 DOI: 10.3389/fncel.2017.00359] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/31/2017] [Indexed: 01/12/2023] Open
Abstract
Autism spectrum disorder (ASD) is a set of neurodevelopmental disorders with a high prevalence and impact on society. ASDs are characterized by deficits in both social behavior and cognitive function. There is a strong genetic basis underlying ASDs that is highly heterogeneous; however, multiple studies have highlighted the involvement of key processes, including neurogenesis, neurite growth, synaptogenesis and synaptic plasticity in the pathophysiology of neurodevelopmental disorders. In this review article, we focus on the major genes and signaling pathways implicated in ASD and discuss the cellular, molecular and functional studies that have shed light on common dysregulated pathways using in vitro, in vivo and human evidence. HighlightsAutism spectrum disorder (ASD) has a prevalence of 1 in 68 children in the United States. ASDs are highly heterogeneous in their genetic basis. ASDs share common features at the cellular and molecular levels in the brain. Most ASD genes are implicated in neurogenesis, structural maturation, synaptogenesis and function.
Collapse
Affiliation(s)
- James Gilbert
- Department of Biology, Boston University, Boston, MA, United States
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, United States.,Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
66
|
Saraiva C, Esteves M, Bernardino L. MicroRNA: Basic concepts and implications for regeneration and repair of neurodegenerative diseases. Biochem Pharmacol 2017; 141:118-131. [DOI: 10.1016/j.bcp.2017.07.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/07/2017] [Indexed: 12/25/2022]
|
67
|
Zhang SF, Chen JC, Zhang J, Xu JG. miR-181a involves in the hippocampus-dependent memory formation via targeting PRKAA1. Sci Rep 2017; 7:8480. [PMID: 28814760 PMCID: PMC5559581 DOI: 10.1038/s41598-017-09095-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023] Open
Abstract
Post-transcriptional gene regulation by microRNAs (miRNAs) is involved in memory formation. However, the roles of individual miRNAs in these processes remain largely unknown. In this study, we want to clarify the role of miR-181a in hippocampus-dependent memory formation. A transient increase in miR-181a expression was observed after conditioned fear conditioning (CFC) and object location task (OLT) training. Selective overexpression or inhibition of miR-181a in the dorsal hippocampus (DH) via the injection of a miR-181a agomir or antagomir enhanced or impaired the CFC- and OLT-dependent memory formation, respectively. Using bioinformatics and luciferase assays, we identified PRKAA1 as a potential target gene of miR-181a. After CFC or OLT training, the expression and activity of PRKAA1 decreased as miR-181a expression increased and was effectively blocked by the miR-181a antagomir. Moreover, microinjection of the PRKAA1 agonist AICAR or inhibitor compound C in the DH reversed the roles of the miR-181a agomir or antagomir in CFC- and OLT-dependent memory formation. In conclusion, this work provides novel evidence describing the role and mechanism of miR-181a in hippocampus-dependent memory formation, which sheds light on the potential regulation of cognition and future treatments for cognitive disorders.
Collapse
Affiliation(s)
- Sun-Fu Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China.,Department of Neurosurgery, The First People's Hospital of Yibin, Yibin, Sichuan, P. R. China
| | - Jun-Chen Chen
- Department of Neurosurgery, Sichuan 81 Rehabilitation Center, Chengdu, Sichuan, P. R. China
| | - Jing Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Jian-Guo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China.
| |
Collapse
|
68
|
Rajman M, Metge F, Fiore R, Khudayberdiev S, Aksoy-Aksel A, Bicker S, Ruedell Reschke C, Raoof R, Brennan GP, Delanty N, Farrell MA, O'Brien DF, Bauer S, Norwood B, Veno MT, Krüger M, Braun T, Kjems J, Rosenow F, Henshall DC, Dieterich C, Schratt G. A microRNA-129-5p/Rbfox crosstalk coordinates homeostatic downscaling of excitatory synapses. EMBO J 2017; 36:1770-1787. [PMID: 28487411 DOI: 10.15252/embj.201695748] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 04/05/2017] [Accepted: 04/07/2017] [Indexed: 12/31/2022] Open
Abstract
Synaptic downscaling is a homeostatic mechanism that allows neurons to reduce firing rates during chronically elevated network activity. Although synaptic downscaling is important in neural circuit development and epilepsy, the underlying mechanisms are poorly described. We performed small RNA profiling in picrotoxin (PTX)-treated hippocampal neurons, a model of synaptic downscaling. Thereby, we identified eight microRNAs (miRNAs) that were increased in response to PTX, including miR-129-5p, whose inhibition blocked synaptic downscaling in vitro and reduced epileptic seizure severity in vivo Using transcriptome, proteome, and bioinformatic analysis, we identified the calcium pump Atp2b4 and doublecortin (Dcx) as miR-129-5p targets. Restoring Atp2b4 and Dcx expression was sufficient to prevent synaptic downscaling in PTX-treated neurons. Furthermore, we characterized a functional crosstalk between miR-129-5p and the RNA-binding protein (RBP) Rbfox1. In the absence of PTX, Rbfox1 promoted the expression of Atp2b4 and Dcx. Upon PTX treatment, Rbfox1 expression was downregulated by miR-129-5p, thereby allowing the repression of Atp2b4 and Dcx. We therefore identified a novel activity-dependent miRNA/RBP crosstalk during synaptic scaling, with potential implications for neural network homeostasis and epileptogenesis.
Collapse
Affiliation(s)
- Marek Rajman
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg, Germany
| | - Franziska Metge
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology, Department of Internal Medicine III, German Center for Cardiovascular Research (DZHK), University Hospital Heidelberg, Heidelberg, Germany
| | - Roberto Fiore
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg, Germany
| | - Sharof Khudayberdiev
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg, Germany
| | - Ayla Aksoy-Aksel
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg, Germany
| | - Silvia Bicker
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg, Germany
| | | | - Rana Raoof
- Physiology & Medical Physics Department, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Gary P Brennan
- Physiology & Medical Physics Department, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | | | - Sebastian Bauer
- Epilepsiezentrum Frankfurt Rhein-Main, Neurozentrum, Goethe-Universität Frankfurt, Frankfurt, Germany.,Epilepsiezentrum Hessen - Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Braxton Norwood
- Epilepsiezentrum Frankfurt Rhein-Main, Neurozentrum, Goethe-Universität Frankfurt, Frankfurt, Germany.,Epilepsiezentrum Hessen - Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Morten T Veno
- Department of Molecular Biology and Genetics and Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Marcus Krüger
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics and Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Felix Rosenow
- Epilepsiezentrum Frankfurt Rhein-Main, Neurozentrum, Goethe-Universität Frankfurt, Frankfurt, Germany.,Epilepsiezentrum Hessen - Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - David C Henshall
- Physiology & Medical Physics Department, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Klaus Tschira Institute for Integrative Computational Cardiology, Department of Internal Medicine III, German Center for Cardiovascular Research (DZHK), University Hospital Heidelberg, Heidelberg, Germany
| | - Gerhard Schratt
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
69
|
Tan MC, Widagdo J, Chau YQ, Zhu T, Wong JJL, Cheung A, Anggono V. The Activity-Induced Long Non-Coding RNA Meg3 Modulates AMPA Receptor Surface Expression in Primary Cortical Neurons. Front Cell Neurosci 2017; 11:124. [PMID: 28515681 PMCID: PMC5413565 DOI: 10.3389/fncel.2017.00124] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/12/2017] [Indexed: 11/13/2022] Open
Abstract
Transcription of new RNA is crucial for maintaining synaptic plasticity, learning and memory. Although the importance of synaptic plasticity-related messenger RNAs (mRNAs) is well established, the role of a large group of long non-coding RNAs (lncRNAs) in long-term potentiation (LTP) is not known. In this study, we demonstrated the expression of a lncRNA cluster, namely maternally expressed gene 3 (Meg3), retrotransposon-like gene 1-anti-sense (Rtl1-AS), Meg8 and Meg9, which is located in the maternally imprinted Dlk1-Dio3 region on mouse chromosome 12qF1, in primary cortical neurons following glycine stimulation in an N-Methyl-D-aspartate receptor (NMDAR)-dependent manner. Importantly, we also validated the expression of Meg3, Meg8 and Meg9 in the hippocampus of mice following cued fear conditioning in vivo. Interestingly, Meg3 is the only lncRNA that is expressed in the nucleus and cytoplasm. Further analysis revealed that Meg3 loss of function blocked the glycine-induced increase of the GluA1 subunit of AMPA receptors on the plasma membrane, a major hallmark of LTP. This aberrant trafficking of AMPA receptors correlated with the dysregulation of the phosphatidylinoside-3-kinase (PI3K)/AKT signaling pathway and the downregulation of the lipid phosphatase and tensin homolog (PTEN). These findings provide the first evidence for a functional role of the lncRNA Meg3 in the intricate regulation of the PTEN/PI3K/AKT signaling cascade during synaptic plasticity in neurons.
Collapse
Affiliation(s)
- Men C Tan
- Clem Jones Centre for Ageing Dementia Research, The University of QueenslandBrisbane, QLD, Australia.,Queensland Brain Institute, The University of QueenslandBrisbane, QLD, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, The University of QueenslandBrisbane, QLD, Australia.,Queensland Brain Institute, The University of QueenslandBrisbane, QLD, Australia
| | - Yu Q Chau
- Clem Jones Centre for Ageing Dementia Research, The University of QueenslandBrisbane, QLD, Australia.,Queensland Brain Institute, The University of QueenslandBrisbane, QLD, Australia
| | - Tianyi Zhu
- Clem Jones Centre for Ageing Dementia Research, The University of QueenslandBrisbane, QLD, Australia.,Queensland Brain Institute, The University of QueenslandBrisbane, QLD, Australia
| | - Justin J-L Wong
- Gene and Stem Cell Therapy Program, Centenary InstituteSydney, NSW, Australia.,Sydney Medical School, University of SydneySydney, NSW, Australia
| | - Allen Cheung
- Queensland Brain Institute, The University of QueenslandBrisbane, QLD, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, The University of QueenslandBrisbane, QLD, Australia.,Queensland Brain Institute, The University of QueenslandBrisbane, QLD, Australia
| |
Collapse
|
70
|
Hu Z, Li Z. miRNAs in synapse development and synaptic plasticity. Curr Opin Neurobiol 2017; 45:24-31. [PMID: 28334640 DOI: 10.1016/j.conb.2017.02.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/23/2017] [Accepted: 02/26/2017] [Indexed: 01/21/2023]
Abstract
Synapses are functional units of the nervous system, through which information is transferred between neurons. The development and activity-dependent modification of synapses require temporally and spatially controlled modulation of gene expression. microRNAs (miRNAs) have emerged as essential regulators of gene expression. They are small non-coding RNAs that regulate mRNA stability and translation by interacting with the 3' untranslated region (3' UTR) of mRNAs. miRNAs are located to neuronal processes to regulate protein synthesis locally and their expression is regulated by synaptic activity. This article reviews recent findings on the role of miRNAs in synapse development and synaptic plasticity.
Collapse
Affiliation(s)
- Zhonghua Hu
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, United States; Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, United States
| | - Zheng Li
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, United States.
| |
Collapse
|
71
|
Adaptor Complex 2 Controls Dendrite Morphology via mTOR-Dependent Expression of GluA2. Mol Neurobiol 2017; 55:1590-1606. [PMID: 28190237 PMCID: PMC5820378 DOI: 10.1007/s12035-017-0436-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 02/03/2017] [Indexed: 11/26/2022]
Abstract
The formation of dendritic arbors in neurons is a highly regulated process. Among the regulators of dendritogenesis are numerous membrane proteins that are eventually internalized via clathrin-mediated endocytosis. AP2 is an adaptor complex that is responsible for recruiting endocytic machinery to internalized cargo. Its direct involvement in dendritogenesis in mammalian neurons has not yet been tested. We found that the knockdown of AP2b1 (β2-adaptin), an AP2 subunit, reduced the number of dendrites in developing rat hippocampal neurons and decreased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA2 levels by inhibiting mechanistic/mammalian target of rapamycin (mTOR). The dendritic tree abruption that was caused by AP2b1 knockdown was rescued by the overexpression of GluA2 or restoration of the activity of the mTOR effector p70S6 kinase (S6K1). Altogether, this work provides evidence that the AP2 adaptor complex is needed for the dendritogenesis of mammalian neurons and reveals that mTOR-dependent GluA2 biosynthesis contributes to this process.
Collapse
|
72
|
Gilbert J, Shu S, Yang X, Lu Y, Zhu LQ, Man HY. β-Amyloid triggers aberrant over-scaling of homeostatic synaptic plasticity. Acta Neuropathol Commun 2016; 4:131. [PMID: 27955702 PMCID: PMC5154098 DOI: 10.1186/s40478-016-0398-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 11/29/2016] [Indexed: 11/21/2022] Open
Abstract
The over-production of β-amyloid (Aβ) has been strongly correlated to neuronal dysfunction and altered synaptic plasticity in Alzheimer’s disease (AD). Accordingly, it has been proposed that disrupted synaptic transmission and neuronal network instability underlie memory failure that is evident in the early phases of AD. Homeostatic synaptic plasticity (HSP) serves to restrain neuronal activity within a physiological range. Therefore a disruption of this mechanism may lead to destabilization in synaptic and neural circuit function. Here, we report that during HSP by neuronal activity deprivation, application of Aβ results in an aberrant over-response of the up-regulation of AMPA receptor (AMPAR)-mediated synaptic currents and cell-surface AMPAR expression. In the visual cortex, in vivo HSP induced by visual deprivation shows a similar over-response following an Aβ local injection. Aβ increases the expression of GluA2-lacking, calcium permeable AMPARs (CP-AMPARs), which are required for the initiation, but not maintenance of HSP. Both GluA2-lacking and GluA2-containing AMPARs contribute to the Aβ-mediated over-scaling of HSP. We also find that Aβ induces the dissociation of HDAC1 from the miR124 transcription factor EVI1, leading to an up-regulation of miR124 expression and increased amount of CP-AMPARs. Thus, via aberrant stimulation of miR124 expression and biogenesis of CP-AMPARs, Aβ is able to induce an over response in HSP. This Aβ-mediated dysregulation in homeostatic plasticity may play an important role in the pathogenesis of altered neural function and memory deficits in the early stages of AD.
Collapse
|
73
|
Fernandes D, Carvalho AL. Mechanisms of homeostatic plasticity in the excitatory synapse. J Neurochem 2016; 139:973-996. [PMID: 27241695 DOI: 10.1111/jnc.13687] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 11/30/2022]
Abstract
Brain development, sensory information processing, and learning and memory processes depend on Hebbian forms of synaptic plasticity, and on the remodeling and pruning of synaptic connections. Neurons in networks implicated in these processes carry out their functions while facing constant perturbation; homeostatic responses are therefore required to maintain neuronal activity within functional ranges for proper brain function. Here, we will review in vitro and in vivo studies demonstrating that several mechanisms underlie homeostatic plasticity of excitatory synapses, and identifying participant molecular players. Emerging evidence suggests a link between disrupted homeostatic synaptic plasticity and neuropsychiatric and neurologic disorders. Hebbian forms of synaptic plasticity, such as long-term potentiation (LTP), induce long-lasting changes in synaptic strength, which can be destabilizing and drive activity to saturation. Conversely, homeostatic plasticity operates to compensate for prolonged activity changes, stabilizing neuronal firing within a dynamic physiological range. We review mechanisms underlying homeostatic plasticity, and address how neurons integrate distinct forms of plasticity for proper brain function. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".
Collapse
Affiliation(s)
- Dominique Fernandes
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,PDBEB-Doctoral Program in Experimental Biology and Biomedicine, Interdisciplinary Research Institute (III-UC), University of Coimbra, Coimbra, Portugal
| | - Ana Luísa Carvalho
- CNC-Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|