51
|
Tabuchi M, Coates KE, Bautista OB, Zukowski LH. Light/Clock Influences Membrane Potential Dynamics to Regulate Sleep States. Front Neurol 2021; 12:625369. [PMID: 33854471 PMCID: PMC8039321 DOI: 10.3389/fneur.2021.625369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
The circadian rhythm is a fundamental process that regulates the sleep-wake cycle. This rhythm is regulated by core clock genes that oscillate to create a physiological rhythm of circadian neuronal activity. However, we do not know much about the mechanism by which circadian inputs influence neurons involved in sleep-wake architecture. One possible mechanism involves the photoreceptor cryptochrome (CRY). In Drosophila, CRY is receptive to blue light and resets the circadian rhythm. CRY also influences membrane potential dynamics that regulate neural activity of circadian clock neurons in Drosophila, including the temporal structure in sequences of spikes, by interacting with subunits of the voltage-dependent potassium channel. Moreover, several core clock molecules interact with voltage-dependent/independent channels, channel-binding protein, and subunits of the electrogenic ion pump. These components cooperatively regulate mechanisms that translate circadian photoreception and the timing of clock genes into changes in membrane excitability, such as neural firing activity and polarization sensitivity. In clock neurons expressing CRY, these mechanisms also influence synaptic plasticity. In this review, we propose that membrane potential dynamics created by circadian photoreception and core clock molecules are critical for generating the set point of synaptic plasticity that depend on neural coding. In this way, membrane potential dynamics drive formation of baseline sleep architecture, light-driven arousal, and memory processing. We also discuss the machinery that coordinates membrane excitability in circadian networks found in Drosophila, and we compare this machinery to that found in mammalian systems. Based on this body of work, we propose future studies that can better delineate how neural codes impact molecular/cellular signaling and contribute to sleep, memory processing, and neurological disorders.
Collapse
Affiliation(s)
- Masashi Tabuchi
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | | | | | | |
Collapse
|
52
|
Tiwari V, Mishra A, Singh S, Mishra SK, Sahu KK, Parul, Kulkarni MJ, Shukla R, Shukla S. Protriptyline improves spatial memory and reduces oxidative damage by regulating NFκB-BDNF/CREB signaling axis in streptozotocin-induced rat model of Alzheimer's disease. Brain Res 2021; 1754:147261. [PMID: 33422534 DOI: 10.1016/j.brainres.2020.147261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023]
Abstract
Antidepressants are well known to exert their role via upregulation of brain derived neurotrophic factor (BDNF). BDNF has been reported to exerts its neuroprotective effect in rodent and primate models as well as in patients of Alzheimer's disease (AD). The aim of our study was to evaluate the effect of protriptyline (PRT), a tricyclic antidepressant, in streptozotocin (STZ)- induced rat model of AD. Total 10 µl of STZ was injected into each ventricle (1 mg/kg). PRT (10 mg/kg, i.p.) treatment was started 3-day post STZ administration and continued till 21 days. We found that STZ treatment significantly increased pTau, Aβ42 and BACE-1 expression, oxidative stress and neurodegeneration in hippocampus and cortex of adult rats. STZ induced impairment in spatial learning and retention memory was associated with increased NFκB and reduced CREB and BDNF expression in cortex and hippocampus. Interestingly, PRT treatment significantly reduced pTau, Aβ42 and BACE-1 levels, neurodegeneration, oxidative stress and glial activation, contributing to the improved spatial learning and retention memory in STZ treated rats. Moreover, PRT treatment significantly improved p-ERK/ERK ratio and enhanced BDNF and CREB levels by reducing NFκB and GFAP expression in STZ treated rats. Our data suggest that impaired NFκB and CREB signaling potentially contribute in AD pathogenesis by elevating oxidative stress and neuroinflammation mediated neurodegeneration. Our study has established protriptyline as a multi target molecule in pre-clinical model of AD and further investigations on PRT like molecules could pave way for further development of effective new treatments in neurodegenerative disorders.
Collapse
Affiliation(s)
- Virendra Tiwari
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Akanksha Mishra
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 01595, USA
| | - Sonu Singh
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Department of Neuroscience, School of Medicine, University of Connecticut (Uconn) Health Center, 263 Farmington Avenue, L-4078, Farmington, CT 06030, USA
| | - Sandeep Kumar Mishra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Department of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U. P, India
| | - Kiran Kumari Sahu
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Parul
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Mahesh J Kulkarni
- Proteomics Facility, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune 411008, India
| | - Rakesh Shukla
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Department of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U. P, India
| | - Shubha Shukla
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
53
|
Rpd3/CoRest-mediated activity-dependent transcription regulates the flexibility in memory updating in Drosophila. Nat Commun 2021; 12:628. [PMID: 33504795 PMCID: PMC7840730 DOI: 10.1038/s41467-021-20898-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/23/2020] [Indexed: 01/30/2023] Open
Abstract
Consolidated memory can be preserved or updated depending on the environmental change. Although such conflicting regulation may happen during memory updating, the flexibility of memory updating may have already been determined in the initial memory consolidation process. Here, we explored the gating mechanism for activity-dependent transcription in memory consolidation, which is unexpectedly linked to the later memory updating in Drosophila. Through proteomic analysis, we discovered that the compositional change in the transcriptional repressor, which contains the histone deacetylase Rpd3 and CoRest, acts as the gating mechanism that opens and closes the time window for activity-dependent transcription. Opening the gate through the compositional change in Rpd3/CoRest is required for memory consolidation, but closing the gate through Rpd3/CoRest is significant to limit future memory updating. Our data indicate that the flexibility of memory updating is determined through the initial activity-dependent transcription, providing a mechanism involved in defining memory state.
Collapse
|
54
|
Eschment M, Franz HR, Güllü N, Hölscher LG, Huh KE, Widmann A. Insulin signaling represents a gating mechanism between different memory phases in Drosophila larvae. PLoS Genet 2020; 16:e1009064. [PMID: 33104728 PMCID: PMC7644093 DOI: 10.1371/journal.pgen.1009064] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 11/05/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
The ability to learn new skills and to store them as memory entities is one of the most impressive features of higher evolved organisms. However, not all memories are created equal; some are short-lived forms, and some are longer lasting. Formation of the latter is energetically costly and by the reason of restricted availability of food or fluctuations in energy expanses, efficient metabolic homeostasis modulating different needs like survival, growth, reproduction, or investment in longer lasting memories is crucial. Whilst equipped with cellular and molecular pre-requisites for formation of a protein synthesis dependent long-term memory (LTM), its existence in the larval stage of Drosophila remains elusive. Considering it from the viewpoint that larval brain structures are completely rebuilt during metamorphosis, and that this process depends completely on accumulated energy stores formed during the larval stage, investing in LTM represents an unnecessary expenditure. However, as an alternative, Drosophila larvae are equipped with the capacity to form a protein synthesis independent so-called larval anaesthesia resistant memory (lARM), which is consolidated in terms of being insensitive to cold-shock treatments. Motivated by the fact that LTM formation causes an increase in energy uptake in Drosophila adults, we tested the idea of whether an energy surplus can induce the formation of LTM in the larval stage. Suprisingly, increasing the metabolic state by feeding Drosophila larvae the disaccharide sucrose directly before aversive olfactory conditioning led to the formation of a protein synthesis dependent longer lasting memory. Moreover, formation of this memory component is accompanied by the suppression of lARM. We ascertained that insulin receptors (InRs) expressed in the mushroom body Kenyon cells suppresses the formation of lARM and induces the formation of a protein synthesis dependent longer lasting memory in Drosophila larvae. Given the numerical simplicity of the larval nervous system this work offers a unique prospect to study the impact of insulin signaling on the formation of protein synthesis dependent memories on a molecular level.
Collapse
Affiliation(s)
- Melanie Eschment
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Hanna R. Franz
- Department of Molecular Neurobiology of Behavior, University of Göttingen, Göttingen, Germany
| | - Nazlı Güllü
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Luis G. Hölscher
- Department of Molecular Neurobiology of Behavior, University of Göttingen, Göttingen, Germany
| | - Ko-Eun Huh
- Department of Molecular Neurobiology of Behavior, University of Göttingen, Göttingen, Germany
| | - Annekathrin Widmann
- Department of Biology, University of Konstanz, Konstanz, Germany
- Department of Molecular Neurobiology of Behavior, University of Göttingen, Göttingen, Germany
| |
Collapse
|
55
|
Lee WP, Chiang MH, Chang LY, Lee JY, Tsai YL, Chiu TH, Chiang HC, Fu TF, Wu T, Wu CL. Mushroom body subsets encode CREB2-dependent water-reward long-term memory in Drosophila. PLoS Genet 2020; 16:e1008963. [PMID: 32780743 PMCID: PMC7418956 DOI: 10.1371/journal.pgen.1008963] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/29/2020] [Indexed: 11/18/2022] Open
Abstract
Long-term memory (LTM) formation depends on the conversed cAMP response element-binding protein (CREB)-dependent gene transcription followed by de novo protein synthesis. Thirsty fruit flies can be trained to associate an odor with water reward to form water-reward LTM (wLTM), which can last for over 24 hours without a significant decline. The role of de novo protein synthesis and CREB-regulated gene expression changes in neural circuits that contribute to wLTM remains unclear. Here, we show that acute inhibition of protein synthesis in the mushroom body (MB) αβ or γ neurons during memory formation using a cold-sensitive ribosome-inactivating toxin disrupts wLTM. Furthermore, adult stage-specific expression of dCREB2b in αβ or γ neurons also disrupts wLTM. The MB αβ and γ neurons can be further classified into five different neuronal subsets including αβ core, αβ surface, αβ posterior, γ main, and γ dorsal. We observed that the neurotransmission from αβ surface and γ dorsal neuron subsets is required for wLTM retrieval, whereas the αβ core, αβ posterior, and γ main are dispensable. Adult stage-specific expression of dCREB2b in αβ surface and γ dorsal neurons inhibits wLTM formation. In vivo calcium imaging revealed that αβ surface and γ dorsal neurons form wLTM traces with different dynamic properties, and these memory traces are abolished by dCREB2b expression. Our results suggest that a small population of neurons within the MB circuits support long-term storage of water-reward memory in Drosophila.
Collapse
Affiliation(s)
- Wang-Pao Lee
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Meng-Hsuan Chiang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Li-Yun Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Jhen-Yi Lee
- School of Medicine, College of Medicine, Chang Gung University, Taiwan
| | - Ya-Lun Tsai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Tai-Hsiang Chiu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taiwan
| | | | - Tsai-Feng Fu
- Department of Applied Chemistry, National Chi Nan University, Taiwan
| | - Tony Wu
- Department of Neurology, Chang Gung Memorial Hospital, Taiwan
| | - Chia-Lin Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Taiwan
- Department of Biochemistry, College of Medicine, Chang Gung University, Taiwan
- * E-mail:
| |
Collapse
|
56
|
Salazar JL, Yang SA, Yamamoto S. Post-Developmental Roles of Notch Signaling in the Nervous System. Biomolecules 2020; 10:biom10070985. [PMID: 32630239 PMCID: PMC7408554 DOI: 10.3390/biom10070985] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Since its discovery in Drosophila, the Notch signaling pathway has been studied in numerous developmental contexts in diverse multicellular organisms. The role of Notch signaling in nervous system development has been extensively investigated by numerous scientists, partially because many of the core Notch signaling components were initially identified through their dramatic ‘neurogenic’ phenotype of developing fruit fly embryos. Components of the Notch signaling pathway continue to be expressed in mature neurons and glia cells, which is suggestive of a role in the post-developmental nervous system. The Notch pathway has been, so far, implicated in learning and memory, social behavior, addiction, and other complex behaviors using genetic model organisms including Drosophila and mice. Additionally, Notch signaling has been shown to play a modulatory role in several neurodegenerative disease model animals and in mediating neural toxicity of several environmental factors. In this paper, we summarize the knowledge pertaining to the post-developmental roles of Notch signaling in the nervous system with a focus on discoveries made using the fruit fly as a model system as well as relevant studies in C elegans, mouse, rat, and cellular models. Since components of this pathway have been implicated in the pathogenesis of numerous psychiatric and neurodegenerative disorders in human, understanding the role of Notch signaling in the mature brain using model organisms will likely provide novel insights into the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Jose L. Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
- Department of Neuroscience, BCM, Houston, TX 77030, USA
- Program in Developmental Biology, BCM, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-832-824-8119
| |
Collapse
|
57
|
Jacob PF, Waddell S. Spaced Training Forms Complementary Long-Term Memories of Opposite Valence in Drosophila. Neuron 2020; 106:977-991.e4. [PMID: 32289250 PMCID: PMC7302427 DOI: 10.1016/j.neuron.2020.03.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/25/2020] [Accepted: 03/16/2020] [Indexed: 12/30/2022]
Abstract
Forming long-term memory (LTM) often requires repetitive experience spread over time. Studies in Drosophila suggest aversive olfactory LTM is optimal after spaced training, multiple trials of differential odor conditioning with rest intervals. Memory after spaced training is frequently compared to that after the same number of trials without intervals. Here we show that, after spaced training, flies acquire additional information and form an aversive memory for the shock-paired odor and a slowly emerging and more persistent "safety-memory" for the explicitly unpaired odor. Safety-memory acquisition requires repetition, order, and spacing of the training trials and relies on triggering specific rewarding dopaminergic neurons. Co-existence of aversive and safety memories is evident as depression of odor-specific responses at different combinations of junctions in the mushroom body output network; combining two outputs appears to signal relative safety. Having complementary aversive and safety memories augments LTM performance after spaced training by making the odor preference more certain.
Collapse
Affiliation(s)
- Pedro F Jacob
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Scott Waddell
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3TA, UK.
| |
Collapse
|
58
|
Wang L, Nie Q, Gao M, Yang L, Xiang JW, Xiao Y, Liu FY, Gong XD, Fu JL, Wang Y, Nguyen QD, Liu Y, Liu M, Li DWC. The transcription factor CREB acts as an important regulator mediating oxidative stress-induced apoptosis by suppressing αB-crystallin expression. Aging (Albany NY) 2020; 12:13594-13617. [PMID: 32554860 PMCID: PMC7377838 DOI: 10.18632/aging.103474] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 05/03/2020] [Indexed: 12/17/2022]
Abstract
The general transcription factor, CREB has been shown to play an essential role in promoting cell proliferation, neuronal survival and synaptic plasticity in the nervous system. However, its function in stress response remains to be elusive. In the present study, we demonstrated that CREB plays a major role in mediating stress response. In both rat lens organ culture and mouse lens epithelial cells (MLECs), CREB promotes oxidative stress-induced apoptosis. To confirm that CREB is a major player mediating the above stress response, we established stable lines of MLECs stably expressing CREB and found that they are also very sensitive to oxidative stress-induced apoptosis. To define the underlying mechanism, RNAseq analysis was conducted. It was found that CREB significantly suppressed expression of the αB-crystallin gene to sensitize CREB-expressing cells undergoing oxidative stress-induced apoptosis. CREB knockdown via CRISPR/CAS9 technology led to upregulation of αB-crystallin and enhanced resistance against oxidative stress-induced apoptosis. Moreover, overexpression of exogenous human αB-crystallin can restore the resistance against oxidative stress-induced apoptosis. Finally, we provided first evidence that CREB directly regulates αB-crystallin gene. Together, our results demonstrate that CREB is an important transcription factor mediating stress response, and it promotes oxidative stress-induced apoptosis by suppressing αB-crystallin expression.
Collapse
Affiliation(s)
- Ling Wang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Qian Nie
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Meng Gao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
- Medical College, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Lan Yang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Jia-Wen Xiang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Yuan Xiao
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Fang-Yuan Liu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Xiao-Dong Gong
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Jia-Ling Fu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Yan Wang
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Quan Dong Nguyen
- Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94303, USA
| | - Yizhi Liu
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan 430074, Hubei, China
| | - David Wan-Cheng Li
- The State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510230, Guangdong, China
| |
Collapse
|
59
|
Kepler LD, McDiarmid TA, Rankin CH. Habituation in high-throughput genetic model organisms as a tool to investigate the mechanisms of neurodevelopmental disorders. Neurobiol Learn Mem 2020; 171:107208. [DOI: 10.1016/j.nlm.2020.107208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/14/2020] [Accepted: 03/02/2020] [Indexed: 10/24/2022]
|
60
|
PGC1α Controls Sucrose Taste Sensitization in Drosophila. Cell Rep 2020; 31:107480. [DOI: 10.1016/j.celrep.2020.03.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 02/12/2020] [Accepted: 03/13/2020] [Indexed: 11/19/2022] Open
|
61
|
Hagenston AM, Bading H, Bas-Orth C. Functional Consequences of Calcium-Dependent Synapse-to-Nucleus Communication: Focus on Transcription-Dependent Metabolic Plasticity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035287. [PMID: 31570333 DOI: 10.1101/cshperspect.a035287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the nervous system, calcium signals play a major role in the conversion of synaptic stimuli into transcriptional responses. Signal-regulated gene transcription is fundamental for a range of long-lasting adaptive brain functions that include learning and memory, structural plasticity of neurites and synapses, acquired neuroprotection, chronic pain, and addiction. In this review, we summarize the diverse mechanisms governing calcium-dependent transcriptional regulation associated with central nervous system plasticity. We focus on recent advances in the field of synapse-to-nucleus communication that include studies of the signal-regulated transcriptome in human neurons, identification of novel regulatory mechanisms such as activity-induced DNA double-strand breaks, and the identification of novel forms of activity- and transcription-dependent adaptations, in particular, metabolic plasticity. We summarize the reciprocal interactions between different kinds of neuroadaptations and highlight the emerging role of activity-regulated epigenetic modifiers in gating the inducibility of signal-regulated genes.
Collapse
Affiliation(s)
- Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, 69120 Heidelberg, Germany
| | - Carlos Bas-Orth
- Department of Medical Cell Biology, Institute for Anatomy and Cell Biology, Heidelberg University, 69120 Heidelberg, Germany
| |
Collapse
|
62
|
Li CH, Yang JL. Wolfberry extracts inhibit Aβ1-42 aggregation and rescue memory loss of AD drosophila. FOOD SCIENCE AND HUMAN WELLNESS 2020. [DOI: 10.1016/j.fshw.2019.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
63
|
Shang P, Zheng F, Han F, Song Y, Pan Z, Yu S, Zhuang X, Chen S. Lipin1 mediates cognitive impairment in fld mice via PKD-ERK pathway. Biochem Biophys Res Commun 2020; 525:286-291. [PMID: 32087966 DOI: 10.1016/j.bbrc.2020.02.070] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/10/2020] [Indexed: 01/20/2023]
Abstract
Lipin1 is important in lipid synthesis because of its phosphatidate phosphatase activity, and it also functions as transcriptional coactivators to regulate the expression of genes involved in lipid metabolism. We found that fld mice exhibit cognitive impairment, and it is related to the DAG-PKD-ERK pathway. We used fld mice to explore the relationship between lipin1 and cognitive function. Our results confirmed the presence of cognitive impairment in the hippocampus of lipin1-deficient mice. As shown in behavioral test, the spatial learning and memory ability of fld mice was much worse than that of wild-type mice. Electron microscopy results showed that the number of synapses in hippocampus of fld mice was significantly reduced. BDNF,SYP, PSD95 were significantly reduced. These results suggest that lipin1 impairs synaptic plasticity. Hence,a deficiency of lipin1 leads to decreased DAG levels and inhibits PKD activation, thereby affecting the phosphorylation of ERK and the CREB.
Collapse
Affiliation(s)
- Pan Shang
- School of Medicine,Shandong University, Jinan, Shandong, 250012, China; Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Fengjie Zheng
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Feng Han
- The People's Hospital of Zhangqiu Area, Jinan, Shandong, 250200, China
| | - Yuwen Song
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Zhe Pan
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Shuyan Yu
- Department of Physiology, Shandong University, School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Xianghua Zhuang
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China.
| | - Shihong Chen
- Department of Endocrinology and Metabolism, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China.
| |
Collapse
|
64
|
Pan Y, Monje M. Activity Shapes Neural Circuit Form and Function: A Historical Perspective. J Neurosci 2020; 40:944-954. [PMID: 31996470 PMCID: PMC6988998 DOI: 10.1523/jneurosci.0740-19.2019] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 12/30/2022] Open
Abstract
The brilliant and often prescient hypotheses of Ramon y Cajal have proven foundational for modern neuroscience, but his statement that "In adult centers the nerve paths are something fixed, ended, immutable … " is an exception that did not stand the test of empirical study. Mechanisms of cellular and circuit-level plasticity continue to shape and reshape many regions of the adult nervous system long after the neurodevelopmental period. Initially focused on neurons alone, the field has followed a meteoric trajectory in understanding of activity-regulated neurodevelopment and ongoing neuroplasticity with an arc toward appreciating neuron-glial interactions and the role that each neural cell type plays in shaping adaptable neural circuity. In this review, as part of a celebration of the 50th anniversary of Society for Neuroscience, we provide a historical perspective, following this arc of inquiry from neuronal to neuron-glial mechanisms by which activity and experience modulate circuit structure and function. The scope of this consideration is broad, and it will not be possible to cover the wealth of knowledge about all aspects of activity-dependent circuit development and plasticity in depth.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305
| |
Collapse
|
65
|
Serway CN, Dunkelberger BS, Del Padre D, Nolan NWC, Georges S, Freer S, Andres AJ, de Belle JS. Importin-α2 mediates brain development, learning and memory consolidation in Drosophila. J Neurogenet 2020; 34:69-82. [PMID: 31965871 DOI: 10.1080/01677063.2019.1709184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Neuronal development and memory consolidation are conserved processes that rely on nuclear-cytoplasmic transport of signaling molecules to regulate gene activity and initiate cascades of downstream cellular events. Surprisingly, few reports address and validate this widely accepted perspective. Here we show that Importin-α2 (Imp-α2), a soluble nuclear transporter that shuttles cargoes between the cytoplasm and nucleus, is vital for brain development, learning and persistent memory in Drosophila melanogaster. Mutations in importin-α2 (imp-α2, known as Pendulin or Pen and homologous with human KPNA2) are alleles of mushroom body miniature B (mbmB), a gene known to regulate aspects of brain development and influence adult behavior in flies. Mushroom bodies (MBs), paired associative centers in the brain, are smaller than normal due to defective proliferation of specific intrinsic Kenyon cell (KC) neurons in mbmB mutants. Extant KCs projecting to the MB β-lobe terminate abnormally on the contralateral side of the brain. mbmB adults have impaired olfactory learning but normal memory decay in most respects, except that protein synthesis-dependent long-term memory (LTM) is abolished. This observation supports an alternative mechanism of persistent memory in which mutually exclusive protein-synthesis-dependent and -independent forms rely on opposing cellular mechanisms or circuits. We propose a testable model of Imp-α2 and nuclear transport roles in brain development and conditioned behavior. Based on our molecular characterization, we suggest that mbmB is hereafter referred to as imp-α2mbmB.
Collapse
Affiliation(s)
- Christine N Serway
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA.,Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Brian S Dunkelberger
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA.,Las Vegas High School, Las Vegas, NV, USA
| | - Denise Del Padre
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA
| | - Nicole W C Nolan
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA.,Methodist Estabrook Cancer Center, Omaha, NE, USA
| | - Stephanie Georges
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA.,Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Stephanie Freer
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA.,Research Square Inc, Nashville, TN, USA
| | - Andrew J Andres
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA
| | - J Steven de Belle
- School of Life Sciences, University of Nevada, Las Vegas, NV, USA.,Department of Psychological Sciences, University of San Diego, San Diego, CA, USA
| |
Collapse
|
66
|
Environmental Light Is Required for Maintenance of Long-Term Memory in Drosophila. J Neurosci 2020; 40:1427-1439. [PMID: 31932417 DOI: 10.1523/jneurosci.1282-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/14/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
Long-term memory (LTM) is stored as functional modifications of relevant neural circuits in the brain. A large body of evidence indicates that the initial establishment of such modifications through the process known as memory consolidation requires learning-dependent transcriptional activation and de novo protein synthesis. However, it remains poorly understood how the consolidated memory is maintained for a long period in the brain, despite constant turnover of molecular substrates. Using the Drosophila courtship conditioning assay of adult males as a memory paradigm, here, we show that in Drosophila, environmental light plays a critical role in LTM maintenance. LTM is impaired when flies are kept in constant darkness (DD) during the memory maintenance phase. Because light activates the brain neurons expressing the neuropeptide pigment-dispersing factor (Pdf), we examined the possible involvement of Pdf neurons in LTM maintenance. Temporal activation of Pdf neurons compensated for the DD-dependent LTM impairment, whereas temporal knockdown of Pdf during the memory maintenance phase impaired LTM in light/dark cycles. Furthermore, we demonstrated that the transcription factor cAMP response element-binding protein (CREB) is required in the memory center, namely, the mushroom bodies (MBs), for LTM maintenance, and Pdf signaling regulates light-dependent transcription via CREB. Our results demonstrate for the first time that universally available environmental light plays a critical role in LTM maintenance by activating the evolutionarily conserved memory modulator CREB in MBs via the Pdf signaling pathway.SIGNIFICANCE STATEMENT Temporary memory can be consolidated into long-term memory (LTM) through de novo protein synthesis and functional modifications of neuronal circuits in the brain. Once established, LTM requires continual maintenance so that it is kept for an extended period against molecular turnover and cellular reorganization that may disrupt memory traces. How is LTM maintained mechanistically? Despite the critical importance of LTM maintenance, its molecular and cellular underpinnings remain elusive. This study using Drosophila is significant because it revealed for the first time in any organism that universally available environmental light plays an essential role in LTM maintenance. Interestingly, light does so by activating the evolutionarily conserved transcription factor cAMP response element-binding protein via peptidergic signaling.
Collapse
|
67
|
Rivi V, Benatti C, Colliva C, Radighieri G, Brunello N, Tascedda F, Blom JMC. Lymnaea stagnalis as model for translational neuroscience research: From pond to bench. Neurosci Biobehav Rev 2019; 108:602-616. [PMID: 31786320 DOI: 10.1016/j.neubiorev.2019.11.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/24/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022]
Abstract
The purpose of this review is to illustrate how a reductionistic, but sophisticated, approach based on the use of a simple model system such as the pond snail Lymnaea stagnalis (L. stagnalis), might be useful to address fundamental questions in learning and memory. L. stagnalis, as a model, provides an interesting platform to investigate the dialog between the synapse and the nucleus and vice versa during memory and learning. More importantly, the "molecular actors" of the memory dialogue are well-conserved both across phylogenetic groups and learning paradigms, involving single- or multi-trials, aversion or reward, operant or classical conditioning. At the same time, this model could help to study how, where and when the memory dialog is impaired in stressful conditions and during aging and neurodegeneration in humans and thus offers new insights and targets in order to develop innovative therapies and technology for the treatment of a range of neurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- V Rivi
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - C Benatti
- Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - C Colliva
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - G Radighieri
- Dept. of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - N Brunello
- Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - F Tascedda
- Dept. of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy
| | - J M C Blom
- Dept. of Education and Human Sciences, University of Modena and Reggio Emilia, Modena, Italy; Centre of Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
68
|
Zhao B, Sun J, Zhang X, Mo H, Niu Y, Li Q, Wang L, Zhong Y. Long-term memory is formed immediately without the need for protein synthesis-dependent consolidation in Drosophila. Nat Commun 2019; 10:4550. [PMID: 31591396 PMCID: PMC6779902 DOI: 10.1038/s41467-019-12436-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 09/04/2019] [Indexed: 12/21/2022] Open
Abstract
It is believed that long-term memory (LTM) cannot be formed immediately because it must go through a protein synthesis-dependent consolidation process. However, the current study uses Drosophila aversive olfactory conditioning to show that such processes are dispensable for context-dependent LTM (cLTM). Single-trial conditioning yields cLTM that is formed immediately in a protein-synthesis independent manner and is sustained over 14 days without decay. Unlike retrieval of traditional LTM, which requires only the conditioned odour and is mediated by mushroom-body neurons, cLTM recall requires both the conditioned odour and reinstatement of the training-environmental context. It is mediated through lateral-horn neurons that connect to multiple sensory brain regions. The cLTM cannot be retrieved if synaptic transmission from any one of these centres is blocked, with effects similar to those of altered encoding context during retrieval. The present study provides strong evidence that long-term memory can be formed easily without the need for consolidation. New protein synthesis is known to be indispensable for the consolidation of long-term memory. Here, the authors report that an olfactory memory can be successfully recalled after 14 days without protein synthesis when the training context is also provided in addition to the conditioned odor.
Collapse
Affiliation(s)
- Bohan Zhao
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Jiameng Sun
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xuchen Zhang
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Han Mo
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Yijun Niu
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Qian Li
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Lianzhang Wang
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Yi Zhong
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research and School of Life Sciences, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
69
|
Siegenthaler D, Escribano B, Bräuler V, Pielage J. Selective suppression and recall of long-term memories in Drosophila. PLoS Biol 2019; 17:e3000400. [PMID: 31454345 PMCID: PMC6711512 DOI: 10.1371/journal.pbio.3000400] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 07/22/2019] [Indexed: 11/18/2022] Open
Abstract
Adaptive decision-making depends on the formation of novel memories. In Drosophila, the mushroom body (MB) is the site of associative olfactory long-term memory (LTM) storage. However, due to the sparse and stochastic representation of olfactory information in Kenyon cells (KCs), genetic access to individual LTMs remains elusive. Here, we develop a cAMP response element (CRE)-activity–dependent memory engram label (CAMEL) tool that genetically tags KCs responding to the conditioned stimulus (CS). CAMEL activity depends on protein-synthesis–dependent aversive LTM conditioning and reflects the time course of CRE binding protein 2 (CREB2) activity during natural memory formation. We demonstrate that inhibition of LTM-induced CAMEL neurons reduces memory expression and that artificial optogenetic reactivation is sufficient to evoke aversive behavior phenocopying memory recall. Together, our data are consistent with CAMEL neurons marking a subset of engram KCs encoding individual memories. This study provides new insights into memory circuitry organization and an entry point towards cellular and molecular understanding of LTM storage. A novel genetic approach enables the visualization and manipulation of memory engram cells in Drosophila, providing a key methodological opportunity to characterize associative memory at the cellular and circuit level.
Collapse
Affiliation(s)
- Dominique Siegenthaler
- Division of Neurobiology and Zoology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Benjamin Escribano
- Division of Neurobiology and Zoology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Vanessa Bräuler
- Division of Neurobiology and Zoology, University of Kaiserslautern, Kaiserslautern, Germany
| | - Jan Pielage
- Division of Neurobiology and Zoology, University of Kaiserslautern, Kaiserslautern, Germany
- * E-mail:
| |
Collapse
|
70
|
Hegde AN, Smith SG. Recent developments in transcriptional and translational regulation underlying long-term synaptic plasticity and memory. ACTA ACUST UNITED AC 2019; 26:307-317. [PMID: 31416904 PMCID: PMC6699410 DOI: 10.1101/lm.048769.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022]
Abstract
Formation of long-term synaptic plasticity that underlies long-term memory requires new protein synthesis. Years of research has elucidated some of the transcriptional and translational mechanisms that contribute to the production of new proteins. Early research on transcription focused on the transcription factor cAMP-responsive element binding protein. Since then, other transcription factors, such as the Nuclear Receptor 4 family of proteins that play a role in memory formation and maintenance have been identified. In addition, several studies have revealed details of epigenetic mechanisms consisting of new types of chemical alterations of DNA such as hydroxymethylation, and various histone modifications in long-term synaptic plasticity and memory. Our understanding of translational control critical for memory formation began with the identification of molecules that impinge on the 5′ and 3′ untranslated regions of mRNAs and continued with the appreciation for local translation near synaptic sites. Lately, a role for noncoding RNAs such as microRNAs in regulating translation factors and other molecules critical for memory has been found. This review describes the past research in brief and mainly focuses on the recent work on molecular mechanisms of transcriptional and translational regulation that form the underpinnings of long-term synaptic plasticity and memory.
Collapse
Affiliation(s)
- Ashok N Hegde
- Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, Georgia 31061, USA
| | - Spencer G Smith
- Department of Biological and Environmental Sciences, Georgia College and State University, Milledgeville, Georgia 31061, USA
| |
Collapse
|
71
|
Awata H, Takakura M, Kimura Y, Iwata I, Masuda T, Hirano Y. The neural circuit linking mushroom body parallel circuits induces memory consolidation in Drosophila. Proc Natl Acad Sci U S A 2019; 116:16080-16085. [PMID: 31337675 PMCID: PMC6690006 DOI: 10.1073/pnas.1901292116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Memory consolidation is augmented by repeated learning following rest intervals, which is known as the spacing effect. Although the spacing effect has been associated with cumulative cellular responses in the neurons engaged in memory, here, we report the neural circuit-based mechanism for generating the spacing effect in the memory-related mushroom body (MB) parallel circuits in Drosophila To investigate the neurons activated during the training, we monitored expression of phosphorylation of mitogen-activated protein kinase (MAPK), ERK [phosphorylation of extracellular signal-related kinase (pERK)]. In an olfactory spaced training paradigm, pERK expression in one of the parallel circuits, consisting of γm neurons, was progressively inhibited via dopamine. This inhibition resulted in reduced pERK expression in a postsynaptic GABAergic neuron that, in turn, led to an increase in pERK expression in a dopaminergic neuron specifically in the later session during spaced training, suggesting that disinhibition of the dopaminergic neuron occurs during spaced training. The dopaminergic neuron was significant for gene expression in the different MB parallel circuits consisting of α/βs neurons for memory consolidation. Our results suggest that the spacing effect-generating neurons and the neurons engaged in memory reside in the distinct MB parallel circuits and that the spacing effect can be a consequence of evolved neural circuit architecture.
Collapse
Affiliation(s)
- Hiroko Awata
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, 606-8507 Kyoto, Japan
| | - Mai Takakura
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, 606-8507 Kyoto, Japan
| | - Yoko Kimura
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, 606-8507 Kyoto, Japan
| | - Ikuko Iwata
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, 606-8507 Kyoto, Japan
| | - Tomoko Masuda
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, 606-8507 Kyoto, Japan
| | - Yukinori Hirano
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, 606-8507 Kyoto, Japan
| |
Collapse
|
72
|
Zhu A, Lao C, Wang Z, Chen Y, Bai C. Characterization of Crocetin-Monoglucuronide as a Neuron-Protective Metabolite of Crocin-1. Mol Nutr Food Res 2019; 63:e1900024. [PMID: 31034723 DOI: 10.1002/mnfr.201900024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/19/2019] [Indexed: 01/24/2023]
Abstract
SCOPE Crocins are important apocarotenoids in the extract of saffron (Crocus sativus L.) and fruits of gardenia (Gardenia jasminoides E.). Crocins have shown versatile biological activities and thus they have been considered to be promising therapeutics for neurodegenerative and heart diseases. Metabolism studies report that crocetin-monoglucuronide (CM) is produced and detected in rat blood plasma after oral administration of crocins. However, due to the lack of standard compound of CM, its unambiguous identification, quantification, and bioactivity studies have been hindered. METHODS AND RESULTS CM is synthetized and its existence in blood plasma in Sprague-Dawley (SD) rats is quantified. The pharmacokinetic studies show that CM is produced in blood and brain after oral administration of crocin-1. It is then discovered that CM possesses neuroprotective activity against beta-amyloid (Aβ) toxicity in PC-12 cells and drosophila models. The enzymatic assay shows that CM can effectively inhibit AChE and docking studies indicate that CM may bind in the gorge channel of AChE. CONCLUSION The work discovered that CM is a neuron-protective metabolite of the orally administrated crocin-1. It is demonstrated that AChE can be one of the targets of CM in its neuron-protective activity.
Collapse
Affiliation(s)
- Anyun Zhu
- Department of Biopharmaceuticals and Materials Engineering, Zhuhai Key Laboratory of Fundamental and Applied Research in Traditional Chinese Medicine, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Chuyu Lao
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ze Wang
- Department of Biopharmaceuticals and Materials Engineering, Zhuhai Key Laboratory of Fundamental and Applied Research in Traditional Chinese Medicine, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Yang Chen
- Department of Biopharmaceuticals and Materials Engineering, Zhuhai Key Laboratory of Fundamental and Applied Research in Traditional Chinese Medicine, Zunyi Medical University, Zhuhai Campus, Zhuhai, 519041, China
| | - Chuan Bai
- Faculty of Forensic Medicine, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| |
Collapse
|
73
|
Castora FJ. Mitochondrial function and abnormalities implicated in the pathogenesis of ASD. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:83-108. [PMID: 30599156 DOI: 10.1016/j.pnpbp.2018.12.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 12/18/2022]
Abstract
Mitochondria are the powerhouse that generate over 90% of the ATP produced in cells. In addition to its role in energy production, the mitochondrion also plays a major role in carbohydrate, fatty acid, amino acid and nucleotide metabolism, programmed cell death (apoptosis), generation of and protection against reactive oxygen species (ROS), immune response, regulation of intracellular calcium ion levels and even maintenance of gut microbiota. With its essential role in bio-energetic as well as non-energetic biological processes, it is not surprising that proper cellular, tissue and organ function is dependent upon proper mitochondrial function. Accordingly, mitochondrial dysfunction has been shown to be directly linked to a variety of medical disorders, particularly neuromuscular disorders and increasing evidence has linked mitochondrial dysfunction to neurodegenerative and neurodevelopmental disorders such as Alzheimer's Disease (AD), Parkinson's Disease (PD), Rett Syndrome (RS) and Autism Spectrum Disorders (ASD). Over the last 40 years there has been a dramatic increase in the diagnosis of ASD and, more recently, an increasing body of evidence indicates that mitochondrial dysfunction plays an important role in ASD development. In this review, the latest evidence linking mitochondrial dysfunction and abnormalities in mitochondrial DNA (mtDNA) to the pathogenesis of autism will be presented. This review will also summarize the results of several recent `approaches used for improving mitochondrial function that may lead to new therapeutic approaches to managing and/or treating ASD.
Collapse
Affiliation(s)
- Frank J Castora
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA; Department of Neurology, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
74
|
Drosophila Acquires a Long-Lasting Body-Size Memory from Visual Feedback. Curr Biol 2019; 29:1833-1841.e3. [PMID: 31104933 DOI: 10.1016/j.cub.2019.04.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/11/2019] [Accepted: 04/12/2019] [Indexed: 11/23/2022]
Abstract
Grasping an object or crossing a trench requires the integration of information on the operating distance of our limbs with precise distance estimation. The reach of our hands and step size of our legs are learned by the visual feedback we get during our actions. This implicit knowledge of our peripersonal space is first acquired during infancy but will be continuously updated throughout our whole life [1]. In contrast, body size of holometabolous insects does not change after metamorphosis; nevertheless, they do have to learn their body reaches at least once. The body size of Drosophila imagines can vary by about 15% depending on environmental factors like food quality and temperature [2]. To investigate how flies acquire knowledge about and memorize their body size, we studied their decisions to either refrain from or initiate climbing over gaps exceeding their body size [3]. Naive (dark-reared) flies overestimate their size and have to learn it from the parallax motion of the retinal images of objects in their environment while walking. Naive flies can be trained in a striped arena and manipulated to underestimate their size, but once consolidated, this memory seems to last for a lifetime. Consolidation of this memory is stress sensitive only in the first 2 h after training but cannot be retrieved for the next 12 h. We have identified a set of intrinsic, lateral neurons of the protocerebral bridge of the central complex [4, 5] that depend on dCREB2 transcriptional activity for long-term memory consolidation and maintenance.
Collapse
|
75
|
Bartolotti N, Lazarov O. CREB signals as PBMC-based biomarkers of cognitive dysfunction: A novel perspective of the brain-immune axis. Brain Behav Immun 2019; 78:9-20. [PMID: 30641141 PMCID: PMC6488430 DOI: 10.1016/j.bbi.2019.01.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/26/2022] Open
Abstract
To date, there is no reliable biomarker for the assessment or determination of cognitive dysfunction in Alzheimer's disease and related dementia. Such a biomarker would not only aid in diagnostics, but could also serve as a measure of therapeutic efficacy. It is widely acknowledged that the hallmarks of Alzheimer's disease, namely, amyloid deposits and neurofibrillary tangles, as well as their precursors and metabolites, are poorly correlated with cognitive function and disease stage and thus have low diagnostic or prognostic value. A lack of biomarkers is one of the major roadblocks in diagnosing the disease and in assessing the efficacy of potential therapies. The phosphorylation of cAMP Response Element Binding protein (pCREB) plays a major role in memory acquisition and consolidation. In the brain, CREB activation by phosphorylation at Ser133 and the recruitment of transcription cofactors such as CREB binding protein (CBP) is a critical step for the formation of memory. This set of processes is a prerequisite for the transcription of genes thought to be important for synaptic plasticity, such as Egr-1. Interestingly, recent work suggests that the expression of pCREB in peripheral blood mononuclear cells (PBMC) positively correlates with pCREB expression in the postmortem brain of Alzheimer's patients, suggesting not only that pCREB expression in PBMC might serve as a biomarker of cognitive dysfunction, but also that the dysfunction of CREB signaling may not be limited to the brain in AD, and that a link may exist between the regulation of CREB in the blood and in the brain. In this review we consider the evidence suggesting a correlation between the level of CREB signals in the brain and blood, the current knowledge about CREB in PBMC and its association with CREB in the brain, and the implications and mechanisms for a neuro-immune cross talk that may underlie this communication. This Review will discuss the possibility that peripheral dysregulation of CREB is an early event in AD pathogenesis, perhaps as a facet of immune system dysfunction, and that this impairment in peripheral CREB signaling modifies CREB signaling in the brain, thus exacerbating cognitive decline in AD. A more thorough understanding of systemic dysregulation of CREB in AD will facilitate the search for a biomarker of cognitive function in AD, and also aid in the understanding of the mechanisms underlying cognitive decline in AD.
Collapse
Affiliation(s)
- Nancy Bartolotti
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
76
|
Ri H, Lee J, Sonn JY, Yoo E, Lim C, Choe J. Drosophila CrebB is a Substrate of the Nonsense-Mediated mRNA Decay Pathway that Sustains Circadian Behaviors. Mol Cells 2019; 42:301-312. [PMID: 31091556 PMCID: PMC6530642 DOI: 10.14348/molcells.2019.2451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/23/2022] Open
Abstract
Post-transcriptional regulation underlies the circadian control of gene expression and animal behaviors. However, the role of mRNA surveillance via the nonsense-mediated mRNA decay (NMD) pathway in circadian rhythms remains elusive. Here, we report that Drosophila NMD pathway acts in a subset of circadian pacemaker neurons to maintain robust 24 h rhythms of free-running locomotor activity. RNA interference-mediated depletion of key NMD factors in timeless-expressing clock cells decreased the amplitude of circadian locomotor behaviors. Transgenic manipulation of the NMD pathway in clock neurons expressing a neuropeptide PIGMENT-DISPERSING FACTOR (PDF) was sufficient to dampen or lengthen free-running locomotor rhythms. Confocal imaging of a transgenic NMD reporter revealed that arrhythmic Clock mutants exhibited stronger NMD activity in PDF-expressing neurons than wild-type. We further found that hypomorphic mutations in Suppressor with morphogenetic effect on genitalia 5 (Smg5 ) or Smg6 impaired circadian behaviors. These NMD mutants normally developed PDF-expressing clock neurons and displayed daily oscillations in the transcript levels of core clock genes. By contrast, the loss of Smg5 or Smg6 function affected the relative transcript levels of cAMP response element-binding protein B (CrebB ) in an isoform-specific manner. Moreover, the overexpression of a transcriptional repressor form of CrebB rescued free-running locomotor rhythms in Smg5-depleted flies. These data demonstrate that CrebB is a rate-limiting substrate of the genetic NMD pathway important for the behavioral output of circadian clocks in Drosophila.
Collapse
Affiliation(s)
- Hwajung Ri
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| | - Jongbin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| | - Jun Young Sonn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| | - Eunseok Yoo
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919,
Korea
| | - Chunghun Lim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919,
Korea
| | - Joonho Choe
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141,
Korea
| |
Collapse
|
77
|
Smolen P, Baxter DA, Byrne JH. How can memories last for days, years, or a lifetime? Proposed mechanisms for maintaining synaptic potentiation and memory. ACTA ACUST UNITED AC 2019; 26:133-150. [PMID: 30992383 PMCID: PMC6478248 DOI: 10.1101/lm.049395.119] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/12/2019] [Indexed: 01/24/2023]
Abstract
With memory encoding reliant on persistent changes in the properties of synapses, a key question is how can memories be maintained from days to months or a lifetime given molecular turnover? It is likely that positive feedback loops are necessary to persistently maintain the strength of synapses that participate in encoding. Such feedback may occur within signal-transduction cascades and/or the regulation of translation, and it may occur within specific subcellular compartments or within neuronal networks. Not surprisingly, numerous positive feedback loops have been proposed. Some posited loops operate at the level of biochemical signal-transduction cascades, such as persistent activation of Ca2+/calmodulin kinase II (CaMKII) or protein kinase Mζ. Another level consists of feedback loops involving transcriptional, epigenetic and translational pathways, and autocrine actions of growth factors such as BDNF. Finally, at the neuronal network level, recurrent reactivation of cell assemblies encoding memories is likely to be essential for late maintenance of memory. These levels are not isolated, but linked by shared components of feedback loops. Here, we review characteristics of some commonly discussed feedback loops proposed to underlie the maintenance of memory and long-term synaptic plasticity, assess evidence for and against their necessity, and suggest experiments that could further delineate the dynamics of these feedback loops. We also discuss crosstalk between proposed loops, and ways in which such interaction can facilitate the rapidity and robustness of memory formation and storage.
Collapse
Affiliation(s)
- Paul Smolen
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Douglas A Baxter
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John H Byrne
- Department of Neurobiology and Anatomy, W. M. Keck Center for the Neurobiology of Learning and Memory, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
78
|
Lyra E Silva NDM, Gonçalves RA, Boehnke SE, Forny-Germano L, Munoz DP, De Felice FG. Understanding the link between insulin resistance and Alzheimer's disease: Insights from animal models. Exp Neurol 2019; 316:1-11. [PMID: 30930096 DOI: 10.1016/j.expneurol.2019.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease affecting millions of people worldwide. AD is characterized by a profound impairment of higher cognitive functions and still lacks any effective disease-modifying treatment. Defective insulin signaling has been implicated in AD pathophysiology, but the mechanisms underlying this process are not fully understood. Here, we review the molecular mechanisms underlying defective brain insulin signaling in rodent models of AD, and in a non-human primate (NHP) model of the disease that recapitulates features observed in AD brains. We further highlight similarities between the NHP and human brains and discuss why NHP models of AD are important to understand disease mechanisms and to improve the translation of effective therapies to humans. We discuss how studies using different animal models have contributed to elucidate the link between insulin resistance and AD.
Collapse
Affiliation(s)
| | | | - Susan E Boehnke
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada; Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil.
| |
Collapse
|
79
|
Sun C, Zhu L, Ma R, Ren J, Wang J, Gao S, Yang D, Ning K, Ling B, Lu B, Chen X, Xu J. Astrocytic miR-324-5p is essential for synaptic formation by suppressing the secretion of CCL5 from astrocytes. Cell Death Dis 2019; 10:141. [PMID: 30760705 PMCID: PMC6374376 DOI: 10.1038/s41419-019-1329-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/01/2018] [Accepted: 01/04/2019] [Indexed: 12/18/2022]
Abstract
There is accumulating evidence that astrocytes play an important role in synaptic formation, plasticity, and pruning. Dicer and the fine-tuning of microRNA (miRNA) network are important for maintaining the normal functions of central nervous system and dysregulation of miRNAs is implicated in neurological disorders. However, little is known about the role of Dicer and miRNAs of astrocytes in the homeostasis of synapse as well as its plasticity. By selectively deleting Dicer in postnatal astrocytes, Dicer-deficient mice exhibited reactive astrogliosis and deficits in dendritic spine formation. Astrocyte-conditioned medium (ACM) collected from Dicer-null astrocytes caused synapse degeneration in cultured primary neurons. The expression of chemokine ligand 5 (CCL5) elevated in Dicer-deleted astrocytes which led to the significant augmentation of secreted CCL5 in ACM. In neurons treated with Dicer KO-ACM, CCL5 supplementation inhibited MAPK/CREB signaling pathway and exacerbated the synaptic formation deficiency, while CCL5 knockdown partially rescued the synapse degeneration. Moreover, we validated CCL5 as miR-324-5p targeted gene. ACM collected from miR-324-5p antagomir-transfected astrocytes mimicked the effect of CCL5 treatment on inhibiting synapse formation and MAPK/CREB signaling in Dicer KO-ACM-cocultured neurons. Furthermore, decreased miR-324-5p expression and elevated CCL5 expression were observed in the brain of aging mice. Our work reveals the non-cell-autonomous roles of astroglial miRNAs in regulation of astrocytic secretory milieu and neuronal synaptogenesis, implicating the loss or misregulation of astroglial miRNA network may contribute to neuroinflammation, neurodegeneration, and aging.
Collapse
Affiliation(s)
- Chenxi Sun
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Zhu
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rongjie Ma
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Ren
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Wang
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shane Gao
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Danjing Yang
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke Ning
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Bin Ling
- The Second People's Hospital of Yunnan Province, Kunming, China.
| | - Bing Lu
- East Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Xu Chen
- Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, China.
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
80
|
Shih MFM, Davis FP, Henry GL, Dubnau J. Nuclear Transcriptomes of the Seven Neuronal Cell Types That Constitute the Drosophila Mushroom Bodies. G3 (BETHESDA, MD.) 2019; 9:81-94. [PMID: 30397017 PMCID: PMC6325895 DOI: 10.1534/g3.118.200726] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/02/2018] [Indexed: 11/18/2022]
Abstract
The insect mushroom body (MB) is a conserved brain structure that plays key roles in a diverse array of behaviors. The Drosophila melanogaster MB is the primary invertebrate model of neural circuits related to memory formation and storage, and its development, morphology, wiring, and function has been extensively studied. MBs consist of intrinsic Kenyon Cells that are divided into three major neuron classes (γ, α'/β' and α/β) and 7 cell subtypes (γd, γm, α'/β'ap, α'/β'm, α/βp, α/βs and α/βc) based on their birth order, morphology, and connectivity. These subtypes play distinct roles in memory processing, however the underlying transcriptional differences are unknown. Here, we used RNA sequencing (RNA-seq) to profile the nuclear transcriptomes of each MB neuronal cell subtypes. We identified 350 MB class- or subtype-specific genes, including the widely used α/β class marker Fas2 and the α'/β' class marker trio Immunostaining corroborates the RNA-seq measurements at the protein level for several cases. Importantly, our data provide a full accounting of the neurotransmitter receptors, transporters, neurotransmitter biosynthetic enzymes, neuropeptides, and neuropeptide receptors expressed within each of these cell types. This high-quality, cell type-level transcriptome catalog for the Drosophila MB provides a valuable resource for the fly neuroscience community.
Collapse
Affiliation(s)
| | - Fred Pejman Davis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA; National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Gilbert Lee Henry
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Josh Dubnau
- Department of Anesthesiology, Stony Brook School of Medicine; Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY
| |
Collapse
|
81
|
The predictive value of serum p-CREB level on secondary cognitive impairment in patients with mild-to-moderate craniocerebral trauma. Neurosurg Rev 2019; 42:715-720. [DOI: 10.1007/s10143-018-01072-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022]
|
82
|
Miyashita T, Kikuchi E, Horiuchi J, Saitoe M. Long-Term Memory Engram Cells Are Established by c-Fos/CREB Transcriptional Cycling. Cell Rep 2018; 25:2716-2728.e3. [DOI: 10.1016/j.celrep.2018.11.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/13/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
|
83
|
Phan A, Thomas CI, Chakraborty M, Berry JA, Kamasawa N, Davis RL. Stromalin Constrains Memory Acquisition by Developmentally Limiting Synaptic Vesicle Pool Size. Neuron 2018; 101:103-118.e5. [PMID: 30503644 DOI: 10.1016/j.neuron.2018.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 08/24/2018] [Accepted: 11/01/2018] [Indexed: 12/14/2022]
Abstract
Stromalin, a cohesin complex protein, was recently identified as a novel memory suppressor gene, but its mechanism remained unknown. Here, we show that Stromalin functions as a negative regulator of synaptic vesicle (SV) pool size in Drosophila neurons. Stromalin knockdown in dopamine neurons during a critical developmental period enhances learning and increases SV pool size without altering the number of dopamine neurons, their axons, or synapses. The developmental effect of Stromalin knockdown persists into adulthood, leading to strengthened synaptic connections and enhanced olfactory memory acquisition in adult flies. Correcting the SV content in dopamine neuron axon terminals by impairing anterograde SV trafficking motor protein Unc104/KIF1A rescues the enhanced-learning phenotype in Stromalin knockdown flies. Our results identify a new mechanism for memory suppression and reveal that the size of the SV pool is controlled genetically and independent from other aspects of neuron structure and function through Stromalin.
Collapse
Affiliation(s)
- Anna Phan
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Connon I Thomas
- Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Molee Chakraborty
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Jacob A Berry
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Ronald L Davis
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
84
|
Abstract
The brain undergoes several changes at structural, molecular, and cellular levels leading to alteration in its functions and these processes are primarily maintained by proteostasis in cells. However, an imbalance in proteostasis due to the abnormal accumulation of protein aggregates induces endoplasmic reticulum (ER) stress. This event, in turn, activate the unfolded protein response; however, in most neurodegenerative conditions and brain injury, an uncontrolled unfolded protein response elicits memory dysfunction. Although the underlying signaling mechanism for impairment of memory function following induction of ER stress remains elusive, recent studies have highlighted that inactivation of a transcription factor, CREB, which is essential for synaptic function and memory formation, plays an essential role for ER stress-induced synaptic and memory dysfunction. In this review, current studies and most updated view on how ER stress affects memory function in both physiological and pathological conditions will be highlighted.
Collapse
Affiliation(s)
- Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
85
|
Widmer YF, Fritsch C, Jungo MM, Almeida S, Egger B, Sprecher SG. Multiple neurons encode CrebB dependent appetitive long-term memory in the mushroom body circuit. eLife 2018; 7:39196. [PMID: 30346271 PMCID: PMC6234028 DOI: 10.7554/elife.39196] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/19/2018] [Indexed: 11/28/2022] Open
Abstract
Lasting changes in gene expression are critical for the formation of long-term memories (LTMs), depending on the conserved CrebB transcriptional activator. While requirement of distinct neurons in defined circuits for different learning and memory phases have been studied in detail, only little is known regarding the gene regulatory changes that occur within these neurons. We here use the fruit fly as powerful model system to study the neural circuits of CrebB-dependent appetitive olfactory LTM. We edited the CrebB locus to create a GFP-tagged CrebB conditional knockout allele, allowing us to generate mutant, post-mitotic neurons with high spatial and temporal precision. Investigating CrebB-dependence within the mushroom body (MB) circuit we show that MB α/β and α’/β’ neurons as well as MBON α3, but not in dopaminergic neurons require CrebB for LTM. Thus, transcriptional memory traces occur in different neurons within the same neural circuit. Our brains can store different types of memories. You may have forgotten what you had for lunch yesterday, but still be able to remember a song from your childhood. Short-term memories and long-term memories form via different mechanisms. To establish long-term memories, the brain must produce new proteins, many of which are common to all members of the animal kingdom. By studying these proteins in organisms such as fruit flies, we can learn more about their role in our own memories. Widmer et al. used this approach to explore how a protein called CrebB helps fruit flies to remember for several days that a specific odor is associated with a sugary reward. These odor-reward memories form in a brain region called the mushroom body, which has three lobes. Input neurons supply information about the odor and the reward to the region, while output neurons pass on information to other parts of the fly brain. CrebB regulates the production of new proteins required to form these long-term odor-reward memories: but where exactly does CrebB act during this process? Using a gene editing technique called CRISPR, Widmer et al. generated mutant flies. In these insects CrebB could be easily deactivated ‘at will’ in either the entire brain, the whole mushroom body, each of the three lobes or in specific output neurons. The flies were then trained on the odor-reward task, and their memory tested 24 hours later. The results revealed that for the memories to form, CrebB is only required in two of the three lobes of the mushroom body, and in certain output neurons. Future studies can now focus on the cells shown to need CrebB to create long-term memories, and identify the other proteins involved in this process. In humans, defects in CrebB are associated with intellectual disability, addiction and depression. The mutant fly created by Widmer et al. could be a useful model in which to investigate how the protein may play a role in these conditions.
Collapse
Affiliation(s)
- Yves F Widmer
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Cornelia Fritsch
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Magali M Jungo
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Silvia Almeida
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Simon G Sprecher
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
86
|
Design, Synthesis of N-phenethyl Cinnamide Derivatives and Their Biological Activities for the Treatment of Alzheimer's Disease: Antioxidant, Beta-amyloid Disaggregating and Rescue Effects on Memory Loss. Molecules 2018; 23:molecules23102663. [PMID: 30332835 PMCID: PMC6222358 DOI: 10.3390/molecules23102663] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/03/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Gx-50 is a bioactive compound for the treatment of Alzheimer’s disease (AD) found in Sichuan pepper (Zanthoxylum bungeanum). In order to find a stronger anti-AD lead compound, 20 gx-50 (1–20) analogs have been designed and synthesized, and their molecular structures were determined based on nuclear magnetic resonance (NMR) and mass spectrometry (MS) analysis, as well as comparison with literature data. Compounds 1–20 were evaluated for their anti-AD potential by using DPPH radical scavenging assay for considering their anti-oxidant activity, thioflavin T (ThT) fluorescence assay for considering the inhibitory or disaggregate potency of Aβ, and transgenic Drosophila model assay for evaluating their rescue effect on memory loss. Finally, compound 13 was determined as a promising anti-AD candidate.
Collapse
|
87
|
Liu L, Qian X, Chao M, Zhao Y, Huang J, Wang T, Sun F, Ling E, Song H. Aluminum toxicity related to SOD and expression of presenilin and CREB in Bombyx mori. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2018; 99:e21480. [PMID: 29978503 DOI: 10.1002/arch.21480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Aluminum (Al) is an important environmental metal factor that can be potentially associated with pathological changes leading to neurotoxicity. The silkworm, Bombyx mori, is an important economic insect and has also been used as a model organism in various research areas. However, the toxicity of Al on silkworm physiology has not been reported. Here, we comprehensively investigate the toxic effects of Al on the silkworm, focusing on its effects on viability and development, superoxide dismutase (SOD) activity, and the expression of presenilin and cAMP response element-binding protein (CREB) in BmE cells and silkworm larvae. BmE cell viability decreased after treatment with aluminum chloride (AlCl3 ) in both dose- and time-dependent manners. When AlCl3 solution was injected into newly hatched fifth instar larvae, both larval weight gain and survival rate were significantly decreased in a manner correlating with AlCl3 dose and developmental stage. Furthermore, when BmE cells and silkworm larvae were exposed to AlCl3 , SOD activity decreased significantly relative to the control group, whereas presenilin expression increased more than twofold. Additionally, CREB and phosphorylated CREB (p-CREB) expression in the heads of fifth instar larvae decreased by 28.0% and 50.0%, respectively. These results indicate that Al inhibits the growth and development of silkworms in vitro and in vivo, altering SOD activity and the expressions of presenilin, CREB, and p-CREB. Our data suggest that B. mori can serve as a model animal for studying Al-induced neurotoxicity or neurodegeneration.
Collapse
Affiliation(s)
- Longhai Liu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiaoran Qian
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Mengling Chao
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yijiao Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Junyi Huang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Taichu Wang
- Sericultural Research Institute, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Fan Sun
- Sericultural Research Institute, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Erjun Ling
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Hongsheng Song
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
88
|
Malvaez M, Greenfield VY, Matheos DP, Angelillis NA, Murphy MD, Kennedy PJ, Wood MA, Wassum KM. Habits Are Negatively Regulated by Histone Deacetylase 3 in the Dorsal Striatum. Biol Psychiatry 2018; 84:383-392. [PMID: 29571524 PMCID: PMC6082729 DOI: 10.1016/j.biopsych.2018.01.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Optimal behavior and decision making result from a balance of control between two strategies, one cognitive/goal-directed and one habitual. These systems are known to rely on the anatomically distinct dorsomedial and dorsolateral striatum, respectively. However, the transcriptional regulatory mechanisms required to learn and transition between these strategies are unknown. Here we examined the role of one chromatin-based transcriptional regulator, histone modification via histone deacetylases (HDACs), in this process. METHODS We combined procedures that diagnose behavioral strategy in rats with pharmacological and viral-mediated HDAC manipulations, chromatin immunoprecipitation, and messenger RNA quantification. RESULTS The results indicate that dorsal striatal HDAC3 activity constrains habit formation. Systemic HDAC inhibition following instrumental (lever press → reward) conditioning increased histone acetylation throughout the dorsal striatum and accelerated habitual control of behavior. HDAC3 was removed from the promoters of key learning-related genes in the dorsal striatum as habits formed with overtraining and with posttraining HDAC inhibition. Decreasing HDAC3 function, either by selective pharmacological inhibition or by expression of dominant-negative mutated HDAC3, in either the dorsolateral striatum or the dorsomedial striatum accelerated habit formation, while HDAC3 overexpression in either region prevented habit. CONCLUSIONS These results challenge the strict dissociation between dorsomedial striatum and dorsolateral striatum function in goal-directed versus habitual behavioral control and identify dorsostriatal HDAC3 as a critical molecular directive of the transition to habit. Because this transition is disrupted in many neurodegenerative and psychiatric diseases, these data suggest a potential molecular mechanism for the negative behavioral symptoms of these conditions and a target for therapeutic intervention.
Collapse
Affiliation(s)
- Melissa Malvaez
- Department of Psychology, University of California, Los Angeles, California
| | - Venuz Y Greenfield
- Department of Psychology, University of California, Los Angeles, California
| | - Dina P Matheos
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California
| | | | - Michael D Murphy
- Department of Psychology, University of California, Los Angeles, California
| | - Pamela J Kennedy
- Department of Psychology, University of California, Los Angeles, California; Brain Research Institute, University of California, Los Angeles, Los Angeles, California
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, California
| | - Kate M Wassum
- Department of Psychology, University of California, Los Angeles, California; Brain Research Institute, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
89
|
Troup M, Yap MH, Rohrscheib C, Grabowska MJ, Ertekin D, Randeniya R, Kottler B, Larkin A, Munro K, Shaw PJ, van Swinderen B. Acute control of the sleep switch in Drosophila reveals a role for gap junctions in regulating behavioral responsiveness. eLife 2018; 7:37105. [PMID: 30109983 PMCID: PMC6117154 DOI: 10.7554/elife.37105] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 08/14/2018] [Indexed: 11/13/2022] Open
Abstract
Sleep is a dynamic process in most animals, involving distinct stages that probably perform multiple functions for the brain. Before sleep functions can be initiated, it is likely that behavioral responsiveness to the outside world needs to be reduced, even while the animal is still awake. Recent work in Drosophila has uncovered a sleep switch in the dorsal fan-shaped body (dFB) of the fly’s central brain, but it is not known whether these sleep-promoting neurons also govern the acute need to ignore salient stimuli in the environment during sleep transitions. We found that optogenetic activation of the sleep switch suppressed behavioral responsiveness to mechanical stimuli, even in awake flies, indicating a broader role for these neurons in regulating arousal. The dFB-mediated suppression mechanism and its associated neural correlates requires innexin6 expression, suggesting that the acute need to reduce sensory perception when flies fall asleep is mediated in part by electrical synapses.
Collapse
Affiliation(s)
- Michael Troup
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Melvyn Hw Yap
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Chelsie Rohrscheib
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Martyna J Grabowska
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Deniz Ertekin
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Roshini Randeniya
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Benjamin Kottler
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia.,King's College London, London, United Kingdom
| | - Aoife Larkin
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia.,University of Cambridge, Cambridge, United Kingdom
| | - Kelly Munro
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Paul J Shaw
- Washington University School of Medicine, St Louis, United States
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
90
|
Swanson A, Wolf T, Sitzmann A, Willette AA. Neuroinflammation in Alzheimer's disease: Pleiotropic roles for cytokines and neuronal pentraxins. Behav Brain Res 2018; 347:49-56. [PMID: 29462653 PMCID: PMC5988985 DOI: 10.1016/j.bbr.2018.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is a potential factor speculated to underlie Alzheimer's disease (AD) etiopathogenesis and progression. The overwhelming focus in this area of research to date has been on the chronic upregulation of pro-inflammatory cytokines to understand how neuroinflammatory mechanisms contribute to neurodegeneration. Yet, it is important to understand the pleiotropic roles of these cytokines in modulating neuroinflammation in which they cannot be labeled as a strictly "good" or "bad" biomarker phenotype. As such, biomarkers with more precise functions are needed to better understand how neuroinflammation impacts the brain in AD. Neuronal pentraxins are a concentration- dependent group of pro- or anti- inflammatory cytokines. There is contradictory evidence of these pentraxins as being both neuroprotective and potentially detrimental in AD. Potential neuroprotective examples include their ability to predict AD-related outcomes such as cognition, memory function and synaptic refinement. This review will briefly outline the basis of AD and subsequently summarize findings for neuropathological mechanisms of neuroinflammation, roles for traditional pro-and anti-inflammatory cytokines, and data found thus far on the neuronal pentraxins.
Collapse
Affiliation(s)
- Ashley Swanson
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Tovah Wolf
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Alli Sitzmann
- Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States.
| | - Auriel A Willette
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States; Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States; Department of Biomedical Sciences, Iowa State University, 2008 Veterinary Medicine, Ames, IA 50011, United States; Department of Neurology, University of Iowa, 2007 Roy Carver Pavilion, 200 Hawkins Drive, Iowa City, IA 52242, United States.
| |
Collapse
|
91
|
Alberini CM, Cruz E, Descalzi G, Bessières B, Gao V. Astrocyte glycogen and lactate: New insights into learning and memory mechanisms. Glia 2018; 66:1244-1262. [PMID: 29076603 PMCID: PMC5903986 DOI: 10.1002/glia.23250] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/05/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022]
Abstract
Memory, the ability to retain learned information, is necessary for survival. Thus far, molecular and cellular investigations of memory formation and storage have mainly focused on neuronal mechanisms. In addition to neurons, however, the brain comprises other types of cells and systems, including glia and vasculature. Accordingly, recent experimental work has begun to ask questions about the roles of non-neuronal cells in memory formation. These studies provide evidence that all types of glial cells (astrocytes, oligodendrocytes, and microglia) make important contributions to the processing of encoded information and storing memories. In this review, we summarize and discuss recent findings on the critical role of astrocytes as providers of energy for the long-lasting neuronal changes that are necessary for long-term memory formation. We focus on three main findings: first, the role of glucose metabolism and the learning- and activity-dependent metabolic coupling between astrocytes and neurons in the service of long-term memory formation; second, the role of astrocytic glucose metabolism in arousal, a state that contributes to the formation of very long-lasting and detailed memories; and finally, in light of the high energy demands of the brain during early development, we will discuss the possible role of astrocytic and neuronal glucose metabolisms in the formation of early-life memories. We conclude by proposing future directions and discussing the implications of these findings for brain health and disease. Astrocyte glycogenolysis and lactate play a critical role in memory formation. Emotionally salient experiences form strong memories by recruiting astrocytic β2 adrenergic receptors and astrocyte-generated lactate. Glycogenolysis and astrocyte-neuron metabolic coupling may also play critical roles in memory formation during development, when the energy requirements of brain metabolism are at their peak.
Collapse
Affiliation(s)
- Cristina M Alberini
- Center for Neural Science, New York University, New York, New York, 10003
- Associate Investigator, Neuroscience Institute, NYU Langone Medical Center, New York, New York, 10016
| | - Emmanuel Cruz
- Center for Neural Science, New York University, New York, New York, 10003
| | - Giannina Descalzi
- Center for Neural Science, New York University, New York, New York, 10003
| | - Benjamin Bessières
- Center for Neural Science, New York University, New York, New York, 10003
| | - Virginia Gao
- Center for Neural Science, New York University, New York, New York, 10003
| |
Collapse
|
92
|
Wang X, Li M, Zhu H, Yu Y, Xu Y, Zhang W, Bian C. Transcriptional Regulation Involved in Fear Memory Reconsolidation. J Mol Neurosci 2018; 65:127-140. [PMID: 29796837 DOI: 10.1007/s12031-018-1084-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/09/2018] [Indexed: 11/26/2022]
Abstract
Memory reconsolidation has been demonstrated to offer a potential target period during which the fear memories underlying fear disorders can be disrupted. Reconsolidation is a labile stage that consolidated memories re-enter after memories are reactivated. Reactivated memories, induced by cues related to traumatic events, are susceptible to strengthening and weakening. Gene transcription regulation and protein synthesis have been suggested to be required for fear memory reconsolidation. Investigating the transcriptional regulation mechanisms underlying reconsolidation may provide a therapeutic method for the treatment of fear disorders such as post-traumatic stress disorder (PTSD). However, the therapeutic effect of treating a fear disorder through interfering with reconsolidation is still contradictory. In this review, we summarize several transcription factors that have been linked to fear memory reconsolidation and propose that transcription factors, as well as related signaling pathways can serve as targets for fear memory interventions. Then, we discuss the application of pharmacological and behavioral interventions during reconsolidation that may or not efficiently treat fear disorders.
Collapse
Affiliation(s)
- Xu Wang
- Department of Military Psychology, College of Psychology, Army Medical University, Chongqing, 400038, China
- Forth Battalion of Cadet Brigade, Army Medical University, Chongqing, 400038, China
| | - Min Li
- Department of Military Psychology, College of Psychology, Army Medical University, Chongqing, 400038, China
| | - Haitao Zhu
- Medical Company, Troops 95848 of People's Liberation Army, Xiaogan, 432100, China
| | - Yongju Yu
- Department of Military Psychology, College of Psychology, Army Medical University, Chongqing, 400038, China
| | - Yuanyuan Xu
- Department of Military Psychology, College of Psychology, Army Medical University, Chongqing, 400038, China
| | - Wenmo Zhang
- Department of Fundamental, Army Logistical University of PLA, Chongqing, 401331, China
| | - Chen Bian
- Department of Military Psychology, College of Psychology, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
93
|
Zhang X, Li Q, Wang L, Liu ZJ, Zhong Y. Active Protection: Learning-Activated Raf/MAPK Activity Protects Labile Memory from Rac1-Independent Forgetting. Neuron 2018; 98:142-155.e4. [DOI: 10.1016/j.neuron.2018.02.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/20/2017] [Accepted: 02/23/2018] [Indexed: 12/20/2022]
|
94
|
Widmann A, Eichler K, Selcho M, Thum AS, Pauls D. Odor-taste learning in Drosophila larvae. JOURNAL OF INSECT PHYSIOLOGY 2018; 106:47-54. [PMID: 28823531 DOI: 10.1016/j.jinsphys.2017.08.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/07/2017] [Accepted: 08/16/2017] [Indexed: 06/07/2023]
Abstract
The Drosophila larva is an attractive model system to study fundamental questions in the field of neuroscience. Like the adult fly, the larva offers a seemingly unlimited genetic toolbox, which allows one to visualize, silence or activate neurons down to the single cell level. This, combined with its simplicity in terms of cell numbers, offers a useful system to study the neuronal correlates of complex processes including associative odor-taste learning and memory formation. Here, we summarize the current knowledge about odor-taste learning and memory at the behavioral level and integrate the recent progress on the larval connectome to shed light on the sub-circuits that allow Drosophila larvae to integrate present sensory input in the context of past experience and to elicit an appropriate behavioral response.
Collapse
Affiliation(s)
| | - Katharina Eichler
- Department of Biology, University of Konstanz, D-78464 Konstanz, Germany; HHMI Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Mareike Selcho
- Department of Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, D-97074 Würzburg, Germany
| | - Andreas S Thum
- Department of Biology, University of Konstanz, D-78464 Konstanz, Germany; Department of Genetics, University of Leipzig, D-04103 Leipzig, Germany.
| | - Dennis Pauls
- Department of Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, D-97074 Würzburg, Germany.
| |
Collapse
|
95
|
Vagnoni A, Bullock SL. A cAMP/PKA/Kinesin-1 Axis Promotes the Axonal Transport of Mitochondria in Aging Drosophila Neurons. Curr Biol 2018; 28:1265-1272.e4. [PMID: 29606421 PMCID: PMC5912900 DOI: 10.1016/j.cub.2018.02.048] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 11/15/2022]
Abstract
Mitochondria play fundamental roles within cells, including energy provision, calcium homeostasis, and the regulation of apoptosis. The transport of mitochondria by microtubule-based motors is critical for neuronal structure and function. This process allows local requirements for mitochondrial functions to be met and also facilitates recycling of these organelles [1, 2]. An age-related reduction in mitochondrial transport has been observed in neurons of mammalian and non-mammalian organisms [3, 4, 5, 6], and has been proposed to contribute to the broader decline in neuronal function that occurs during aging [3, 5, 6, 7]. However, the factors that influence mitochondrial transport in aging neurons are poorly understood. Here we provide evidence using the tractable Drosophila wing nerve system that the cyclic AMP/protein kinase A (cAMP/PKA) pathway promotes the axonal transport of mitochondria in adult neurons. The level of the catalytic subunit of PKA decreases during aging, and acute activation of the cAMP/PKA pathway in aged flies strongly stimulates mitochondrial motility. Thus, the age-related impairment of transport is reversible. The expression of many genes is increased by PKA activation in aged flies. However, our results indicate that elevated mitochondrial transport is due in part to upregulation of the heavy chain of the kinesin-1 motor, the level of which declines during aging. Our study identifies evolutionarily conserved factors that can strongly influence mitochondrial motility in aging neurons. cAMP/PKA pathway promotes mitochondrial transport in adult Drosophila wing neurons Pathway activation in aged flies suppresses age-related reduction in transport Levels of PKAc and kinesin-1 motor decline during aging Kinesin-1 upregulation is an important output of PKA activation in aged flies
Collapse
Affiliation(s)
- Alessio Vagnoni
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK; Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London SE5 9RX, UK.
| | - Simon L Bullock
- Division of Cell Biology, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
96
|
Korneev SA, Vavoulis DV, Naskar S, Dyakonova VE, Kemenes I, Kemenes G. A CREB2-targeting microRNA is required for long-term memory after single-trial learning. Sci Rep 2018; 8:3950. [PMID: 29500383 PMCID: PMC5834643 DOI: 10.1038/s41598-018-22278-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 02/12/2018] [Indexed: 02/01/2023] Open
Abstract
Although single-trial induced long-term memories (LTM) have been of major interest in neuroscience, how LTM can form after a single episode of learning remains largely unknown. We hypothesized that the removal of molecular inhibitory constraints by microRNAs (miRNAs) plays an important role in this process. To test this hypothesis, first we constructed small non-coding RNA (sncRNA) cDNA libraries from the CNS of Lymnaea stagnalis subjected to a single conditioning trial. Then, by next generation sequencing of these libraries, we identified a specific pool of miRNAs regulated by training. Of these miRNAs, we focussed on Lym-miR-137 whose seed region shows perfect complementarity to a target sequence in the 3' UTR of the mRNA for CREB2, a well-known memory repressor. We found that Lym-miR-137 was transiently up-regulated 1 h after single-trial conditioning, preceding a down-regulation of Lym-CREB2 mRNA. Furthermore, we discovered that Lym-miR-137 is co-expressed with Lym-CREB2 mRNA in an identified neuron with an established role in LTM. Finally, using an in vivo loss-of-function approach we demonstrated that Lym-miR-137 is required for single-trial induced LTM.
Collapse
Affiliation(s)
- Sergei A Korneev
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK.
| | - Dimitris V Vavoulis
- RDM Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Clifton, BS8 1UB, UK
| | - Souvik Naskar
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Varvara E Dyakonova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Ildikó Kemenes
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - György Kemenes
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| |
Collapse
|
97
|
Abstract
A recent study has found that pathogen exposure early in the life of the nematode Caenorhabditis elegans leads to a long-lasting aversion that requires distinct sets of neurons for the formation and retrieval of the imprinted memory.
Collapse
Affiliation(s)
- Diego Rayes
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, UNS-CONICET, B-8000FWB, Bahía Blanca, Argentina.
| | - Mark J Alkema
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
98
|
Hansen RT, Zhang HT. The Past, Present, and Future of Phosphodiesterase-4 Modulation for Age-Induced Memory Loss. ADVANCES IN NEUROBIOLOGY 2018; 17:169-199. [PMID: 28956333 DOI: 10.1007/978-3-319-58811-7_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The purpose of this chapter is to highlight the state of progress for phosphodiesterase-4 (PDE4) modulation as a potential therapeutic for psychiatric illness, and to draw attention to particular hurdles and obstacles that must be overcome in future studies to develop PDE4-mediated therapeutics. Pathological and non-pathological related memory loss will be the focus of the chapter; however, we will at times also touch upon other psychiatric illnesses like anxiety and depression. First, we will provide a brief background of PDE4, and the rationale for its extensive study in cognition. Second, we will explore fundamental differences in individual PDE4 subtypes, and then begin to address differences between pathological and non-pathological aging. Alterations of cAMP/PDE4 signaling that occur within normal vs. pathological aging, and the potential for PDE4 modulation to combat these alterations within each context will be described. Finally, we will finish the chapter with obstacles that have hindered the field, and future studies and alternative viewpoints that need to be addressed. Overall, we hope this chapter will demonstrate the incredible complexity of PDE4 signaling in the brain, and will be useful in forming a strategy to develop future PDE4-mediated therapeutics for psychiatric illnesses.
Collapse
Affiliation(s)
- Rolf T Hansen
- Departments of Behavioral Medicine & Psychiatry and Physiology & Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506-9137, USA
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Institute of Pharmacology, Taishan Medical University, Taian, 271016, China.
| |
Collapse
|
99
|
Briskin-Luchinsky V, Levy R, Halfon M, Susswein AJ. Molecular correlates of separate components of training that contribute to long-term memory formation after learning that food is inedible in Aplysia. ACTA ACUST UNITED AC 2018; 25:90-99. [PMID: 29339560 PMCID: PMC5772390 DOI: 10.1101/lm.046326.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Abstract
Training Aplysia with inedible food for a period that is too brief to produce long-term memory becomes effective in producing memory when training is paired with a nitric oxide (NO) donor. Lip stimulation for the same period of time paired with an NO donor is ineffective. Using qPCR, we examined molecular correlates of brief training versus lip stimulation, of treatment with an NO donor versus saline, and of the combined stimuli producing long-term memory. Changes were examined in mRNA expression of Aplysia homologs of C/EBP, CREB1, CREB1α, CREB1β, and CREB2, in both the buccal and cerebral ganglia controlling feeding. Both the brief training and the NO donor increased expression of C/EBP, CREB1, CREB1α, and CREB1β, but not CREB2 in the buccal ganglia. For CREB1α, there was a significant interaction between the effects of the brief training and of the NO donor. In addition, the NO donor, but not brief training, increased expression of all of the genes in the cerebral ganglion. These findings show that the components of learning that alone do not produce memory produce molecular changes in different ganglia. Thus, long-term memory is likely to arise by both additive and interactive increases in gene expression.
Collapse
Affiliation(s)
- Valeria Briskin-Luchinsky
- The Mina and Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Roi Levy
- The Mina and Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Maayan Halfon
- The Mina and Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Abraham J Susswein
- The Mina and Everard Goodman Faculty of Life Sciences and The Leslie and Susan Gonda (Goldschmied) Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan, 52900, Israel
| |
Collapse
|
100
|
Eagle AL, Gajewski PA, Robison AJ. Role of hippocampal activity-induced transcription in memory consolidation. Rev Neurosci 2018; 27:559-73. [PMID: 27180338 DOI: 10.1515/revneuro-2016-0010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 03/26/2016] [Indexed: 01/15/2023]
Abstract
Experience-dependent changes in the strength of connections between neurons in the hippocampus (HPC) are critical for normal learning and memory consolidation, and disruption of this process drives a variety of neurological and psychiatric diseases. Proper HPC function relies upon discrete changes in gene expression driven by transcription factors (TFs) induced by neuronal activity. Here, we describe the induction and function of many of the most well-studied HPC TFs, including cyclic-AMP response element binding protein, serum-response factor, AP-1, and others, and describe their role in the learning process. We also discuss the known target genes of many of these TFs and the purported mechanisms by which they regulate long-term changes in HPC synaptic strength. Moreover, we propose that future research in this field will depend upon unbiased identification of additional gene targets for these activity-dependent TFs and subsequent meta-analyses that identify common genes or pathways regulated by multiple TFs in the HPC during learning or disease.
Collapse
|