1
|
Liu S, Zhang X, Gong X, Yu J, Lin T, Xiang Q, Zeng X, Liu J. Molecular and pharmacological characterization of the dopamine receptors in the oriental fruit fly, Bactrocera dorsalis. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 180:104312. [PMID: 40245998 DOI: 10.1016/j.ibmb.2025.104312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/21/2025] [Accepted: 04/12/2025] [Indexed: 04/19/2025]
Abstract
Dopamine (DA) is a critical molecule within the insect nervous system, known to regulate a myriad of physiological functions and instigate behavioral shifts in insects. It exerts its effects by interacting with specific dopamine receptors (DARs). In this study, three DARs cDNAs from Bactrocera dorsalis (Hendel) (Diptera: Tephritidae) (BdDOP1, BdDOP2 and BdDOP3) were cloned using molecular biology techniques. These receptors exhibited high sequence identity with their orthologous DARs, and phylogenetic analyses also clustered these receptors within their respective receptor subtype. Additionally, the high expression levels of these DARs in the head suggest their prominent role in the central nervous system of B. dorsalis. To investigate the pharmacological properties of these receptors, expression vectors for BdDOP1, BdDOP2 and BdDOP3 were constructed and expressed in HEK-293T cells. Our results demonstrated that DA and synthetic agonists activated these receptors in a dose-dependent manner, and DA activation can be competitively inhibited by various antagonists, exhibiting distinct potencies for each dopamine receptor type. Among the tested antagonists, SCH-23390, methiothepin, and metoclopramide were identified as the most potent inhibitors of BdDOP1, BdDOP2 and BdDOP3, respectively. This study provides valuable insights into the molecular and pharmacological characteristics of DARs in B. dorsalis, offering a theoretical foundation for the development of novel behavioral modulators targeting these receptors. The findings also serve as a reference for the functional analyses of DARs in other insect species.
Collapse
Affiliation(s)
- Shiyan Liu
- State Key Laboratory of Green Pesticide/Guangdong Engineering Research Center for Insect Behavior Regulation/College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China
| | - Xuefeng Zhang
- State Key Laboratory of Green Pesticide/Guangdong Engineering Research Center for Insect Behavior Regulation/College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China
| | - Xin Gong
- State Key Laboratory of Green Pesticide/Guangdong Engineering Research Center for Insect Behavior Regulation/College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China
| | - Jinxin Yu
- State Key Laboratory of Green Pesticide/Guangdong Engineering Research Center for Insect Behavior Regulation/College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China
| | - Tao Lin
- College of Life Science, Shangrao Normal University, Shangrao, 334001, China
| | - Qian Xiang
- State Key Laboratory of Green Pesticide/Guangdong Engineering Research Center for Insect Behavior Regulation/College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China
| | - Xinnian Zeng
- State Key Laboratory of Green Pesticide/Guangdong Engineering Research Center for Insect Behavior Regulation/College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China.
| | - Jiali Liu
- State Key Laboratory of Green Pesticide/Guangdong Engineering Research Center for Insect Behavior Regulation/College of Plant Protection, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
2
|
Jones JD, Holder BL, Montgomery AC, McAdams CV, He E, Burns AE, Eiken KR, Vogt A, Velarde AI, Elder AJ, McEllin JA, Dissel S. The dorsal fan-shaped body is a neurochemically heterogeneous sleep-regulating center in Drosophila. PLoS Biol 2025; 23:e3003014. [PMID: 40138668 DOI: 10.1371/journal.pbio.3003014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/03/2025] [Accepted: 01/13/2025] [Indexed: 03/29/2025] Open
Abstract
Sleep is a behavior that is conserved throughout the animal kingdom. Yet, despite extensive studies in humans and animal models, the exact function or functions of sleep remain(s) unknown. A complicating factor in trying to elucidate the function of sleep is the complexity and multiplicity of neuronal circuits that are involved in sleep regulation. It is conceivable that distinct sleep-regulating circuits are only involved in specific aspects of sleep and may underlie different sleep functions. Thus, it would be beneficial to assess the contribution of individual circuits in sleep's putative functions. The intricacy of the mammalian brain makes this task extremely difficult. However, the fruit fly Drosophila melanogaster, with its simpler brain organization, available connectomics, and unparalleled genetics, offers the opportunity to interrogate individual sleep-regulating centers. In Drosophila, neurons projecting to the dorsal fan-shaped body (dFB) have been proposed to be key regulators of sleep, particularly sleep homeostasis. We recently demonstrated that the most widely used genetic tool to manipulate dFB neurons, the 23E10-GAL4 driver, expresses in 2 sleep-regulating neurons (VNC-SP neurons) located in the ventral nerve cord (VNC), the fly analog of the vertebrate spinal cord. Since most data supporting a role for the dFB in sleep regulation have been obtained using 23E10-GAL4, it is unclear whether the sleep phenotypes reported in these studies are caused by dFB neurons or VNC-SP cells. A recent publication replicated our finding that 23E10-GAL4 contains sleep-promoting neurons in the VNC. However, it also proposed that the dFB is not involved in sleep regulation at all, but this suggestion was made using genetic tools that are not dFB-specific and a very mild sleep deprivation protocol. In this study, using a newly created dFB-specific genetic driver line, we demonstrate that optogenetic activation of the majority of 23E10-GAL4 dFB neurons promotes sleep and that these neurons are involved in sleep homeostasis. We also show that dFB neurons require stronger stimulation than VNC-SP cells to promote sleep. In addition, we demonstrate that dFB-induced sleep can consolidate short-term memory (STM) into long-term memory (LTM), suggesting that the benefit of sleep on memory is not circuit-specific. Finally, we show that dFB neurons are neurochemically heterogeneous and can be divided in 3 populations. Most dFB neurons express both glutamate and acetylcholine, while a minority of cells expresses only one of these 2 neurotransmitters. Importantly, dFB neurons do not express GABA, as previously suggested. Using neurotransmitter-specific dFB tools, our data also points at cholinergic dFB neurons as particularly potent at regulating sleep and sleep homeostasis.
Collapse
Affiliation(s)
- Joseph D Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Brandon L Holder
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Andrew C Montgomery
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Chloe V McAdams
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Emily He
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Anna E Burns
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Kiran R Eiken
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Alex Vogt
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Adriana I Velarde
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Alexandra J Elder
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Jennifer A McEllin
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Stephane Dissel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| |
Collapse
|
3
|
Sabandal PR, Kim YC, Sabandal JM, Han KA. Social context and dopamine signaling converge in the mushroom body to drive impulsivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639508. [PMID: 40027633 PMCID: PMC11870619 DOI: 10.1101/2025.02.21.639508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Organisms adapt their behaviors flexibly in response to various internal and environmental factors. However, how and where these factors converge in the brain to alter behavior is not well understood. In this study, we examine how social context interacts with dopamine activity to influence inhibitory control in Drosophila . We found that, regardless of social context-whether isolated or in groups-wild-type flies consistently showed strong movement suppression in a go/no-go task that measures action restraint. In contrast, flies with enhanced dopamine activity suppressed their movements when tested alone or with potential mates but exhibited impulsive behaviors when exposed to same-sex peers. This social-context-dependent impulsivity was shown to rely on dopamine-D1 receptor-cAMP signaling in mushroom body (MB) neurons. Remarkably, activating the MB was sufficient to induce impulsivity, even without dopamine input or a social context. Our findings highlight MB as a critical hub where social context and dopamine signaling converge to regulate impulsive behavior in Drosophila . Signficance statement This study demonstrates that impulsivity results from the interplay between elevated dopamine levels and social context, rather than dopamine alone, with the mushroom body (MB) serving as a key neural hub for integrating these signals in Drosophila . Social stimuli, such as the presence of same-sex peers, disrupt inhibitory control in a context-dependent manner, highlighting the importance of multimodal sensory inputs and MB activity. These findings challenge the isolation-focused approach in traditional impulsivity research and underscore the need to account for social influences when investigating cognitive processes and disorders like ADHD, autism, and substance use, where social settings often amplify symptoms. Classification Genetics / Neuroscience.
Collapse
|
4
|
Dai X, Le JQ, Ma D, Rosbash M. Four SpsP neurons are an integrating sleep regulation hub in Drosophila. SCIENCE ADVANCES 2024; 10:eads0652. [PMID: 39576867 PMCID: PMC11584021 DOI: 10.1126/sciadv.ads0652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Sleep is essential and highly conserved, yet its regulatory mechanisms remain largely unknown. To identify sleep drive neurons, we imaged Drosophila brains with calcium-modulated photoactivatable ratiometric integrator (CaMPARI). The results indicate that the activity of the protocerebral bridge (PB) correlates with sleep drive. We further identified a key three-layer PB circuit, EPG-SpsP-PEcG, in which the four SpsP neurons in the PB respond to ellipsoid body (EB) signals from EPG neurons and send signals back to the EB through PEcG neurons. This circuit is strengthened by sleep deprivation, indicating a plasticity response to sleep drive. SpsP neurons also receive inputs from the sensorimotor brain region, suggesting that they may encode sleep drive by integrating sensorimotor and navigation cues. Together, our experiments show that the four SpsP neurons and their sleep regulatory circuit play an important and dynamic role in sleep regulation.
Collapse
Affiliation(s)
- Xihuimin Dai
- Howard Hughes Medical Institute, Brandeis University, Waltham MA 02454, USA
| | - Jasmine Quynh Le
- Howard Hughes Medical Institute, Brandeis University, Waltham MA 02454, USA
| | - Dingbang Ma
- Howard Hughes Medical Institute, Brandeis University, Waltham MA 02454, USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Michael Rosbash
- Howard Hughes Medical Institute, Brandeis University, Waltham MA 02454, USA
| |
Collapse
|
5
|
Wani AR, Chowdhury B, Luong J, Chaya GM, Patel K, Isaacman-Beck J, Kayser MS, Syed MH. Stem cell-specific ecdysone signaling regulates the development of dorsal fan-shaped body neurons and sleep homeostasis. Curr Biol 2024; 34:4951-4967.e5. [PMID: 39383867 PMCID: PMC11537841 DOI: 10.1016/j.cub.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 08/09/2024] [Accepted: 09/11/2024] [Indexed: 10/11/2024]
Abstract
Complex behaviors arise from neural circuits that assemble from diverse cell types. Sleep is a conserved behavior essential for survival, yet little is known about how the nervous system generates neuron types of a sleep-wake circuit. Here, we focus on the specification of Drosophila 23E10-labeled dorsal fan-shaped body (dFB) long-field tangential input neurons that project to the dorsal layers of the fan-shaped body neuropil in the central complex. We use lineage analysis and genetic birth dating to identify two bilateral type II neural stem cells (NSCs) that generate 23E10 dFB neurons. We show that adult 23E10 dFB neurons express ecdysone-induced protein 93 (E93) and that loss of ecdysone signaling or E93 in type II NSCs results in their misspecification. Finally, we show that E93 knockdown in type II NSCs impairs adult sleep behavior. Our results provide insight into how extrinsic hormonal signaling acts on NSCs to generate the neuronal diversity required for adult sleep behavior. These findings suggest that some adult sleep disorders might derive from defects in stem cell-specific temporal neurodevelopmental programs.
Collapse
Affiliation(s)
- Adil R Wani
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, Albuquerque, NM 87131, USA
| | - Budhaditya Chowdhury
- The Advanced Science Research Center, City University of New York, New York, NY 10031, USA
| | - Jenny Luong
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gonzalo Morales Chaya
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, Albuquerque, NM 87131, USA
| | - Krishna Patel
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, Albuquerque, NM 87131, USA
| | | | - Matthew S Kayser
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Mubarak Hussain Syed
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, Albuquerque, NM 87131, USA.
| |
Collapse
|
6
|
Sitaraman D, Vecsey CG, Koochagian C. Activity Monitoring for Analysis of Sleep in Drosophila melanogaster. Cold Spring Harb Protoc 2024; 2024:pdb.top108095. [PMID: 38336390 PMCID: PMC11827337 DOI: 10.1101/pdb.top108095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Sleep is important for survival, and the need for sleep is conserved across species. In the past two decades, the fruit fly Drosophila melanogaster has emerged as a promising system in which to study the genetic, neural, and physiological bases of sleep. Through significant advances in our understanding of the regulation of sleep in flies, the field is poised to address several open questions about sleep, such as how the need for sleep is encoded, how molecular regulators of sleep are situated within brain networks, and what the functions of sleep are. Here, we describe key findings, open questions, and commonly used methods that have been used to inform existing theories and develop new ways of thinking about the function, regulation, and adaptability of sleep behavior.
Collapse
Affiliation(s)
- Divya Sitaraman
- Department of Psychology, College of Science, California State University, Hayward, California 94542, USA
| | | | - Casey Koochagian
- Neuroscience Program, Skidmore College, Saratoga Springs, New York 12866, USA
| |
Collapse
|
7
|
Cazalé-Debat L, Scheunemann L, Day M, Fernandez-D V Alquicira T, Dimtsi A, Zhang Y, Blackburn LA, Ballardini C, Greenin-Whitehead K, Reynolds E, Lin AC, Owald D, Rezaval C. Mating proximity blinds threat perception. Nature 2024; 634:635-643. [PMID: 39198656 PMCID: PMC11485238 DOI: 10.1038/s41586-024-07890-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 07/31/2024] [Indexed: 09/01/2024]
Abstract
Romantic engagement can bias sensory perception. This 'love blindness' reflects a common behavioural principle across organisms: favouring pursuit of a coveted reward over potential risks1. In the case of animal courtship, such sensory biases may support reproductive success but can also expose individuals to danger, such as predation2,3. However, how neural networks balance the trade-off between risk and reward is unknown. Here we discover a dopamine-governed filter mechanism in male Drosophila that reduces threat perception as courtship progresses. We show that during early courtship stages, threat-activated visual neurons inhibit central courtship nodes via specific serotonergic neurons. This serotonergic inhibition prompts flies to abort courtship when they see imminent danger. However, as flies advance in the courtship process, the dopaminergic filter system reduces visual threat responses, shifting the balance from survival to mating. By recording neural activity from males as they approach mating, we demonstrate that progress in courtship is registered as dopaminergic activity levels ramping up. This dopamine signalling inhibits the visual threat detection pathway via Dop2R receptors, allowing male flies to focus on courtship when they are close to copulation. Thus, dopamine signalling biases sensory perception based on perceived goal proximity, to prioritize between competing behaviours.
Collapse
Affiliation(s)
- Laurie Cazalé-Debat
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
| | - Lisa Scheunemann
- Freie Universität Berlin, Institute of Biology, Berlin, Germany
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Megan Day
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
| | - Tania Fernandez-D V Alquicira
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Dimtsi
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Youchong Zhang
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford, UK
| | - Lauren A Blackburn
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
- School of Science and the Environment, University of Worcester, Worcester, UK
| | - Charles Ballardini
- School of Biosciences, University of Birmingham, Birmingham, UK
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK
| | - Katie Greenin-Whitehead
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Eric Reynolds
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andrew C Lin
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - David Owald
- Institut für Neurophysiologie and NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carolina Rezaval
- School of Biosciences, University of Birmingham, Birmingham, UK.
- Birmingham Centre for Neurogenetics, University of Birmingham, Birmingham, UK.
| |
Collapse
|
8
|
Le JQ, Ma D, Dai X, Rosbash M. Light and dopamine impact two circadian neurons to promote morning wakefulness. Curr Biol 2024; 34:3941-3954.e4. [PMID: 39142287 PMCID: PMC11404089 DOI: 10.1016/j.cub.2024.07.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/13/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024]
Abstract
In both mammals and flies, circadian brain neurons orchestrate physiological oscillations and behaviors like wake and sleep-these neurons can be subdivided by morphology and by gene expression patterns. Recent single-cell sequencing studies identified 17 Drosophila circadian neuron groups. One of these includes only two lateral neurons (LNs), which are marked by the expression of the neuropeptide ion transport peptide (ITP). Although these two ITP+ LNs have long been grouped with five other circadian evening activity cells, inhibiting the two neurons alone strongly reduces morning activity, indicating that they also have a prominent morning function. As dopamine signaling promotes activity in Drosophila, like in mammals, we considered that dopamine might influence this morning activity function. Moreover, the ITP+ LNs express higher mRNA levels than other LNs of the type 1-like dopamine receptor Dop1R1. Consistent with the importance of Dop1R1, cell-specific CRISPR-Cas9 mutagenesis of this receptor in the two ITP+ LNs renders flies significantly less active in the morning, and ex vivo live imaging shows Dop1R1-dependent cyclic AMP (cAMP) responses to dopamine in these two neurons. Notably, the response is more robust in the morning, reflecting higher morning Dop1R1 mRNA levels in the two neurons. As mRNA levels are not elevated in constant darkness, this suggests light-dependent upregulation of morning Dop1R1 transcript levels. Taken together with the enhanced morning cAMP response to dopamine, the data indicate how light and dopamine promote morning wakefulness in flies, mimicking the important effect of light on morning wakefulness in humans.
Collapse
Affiliation(s)
- Jasmine Quynh Le
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Dingbang Ma
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA; Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xihuimin Dai
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA.
| |
Collapse
|
9
|
O’Hara MK, Saul C, Handa A, Cho B, Zheng X, Sehgal A, Williams JA. The NFκB Dif is required for behavioral and molecular correlates of sleep homeostasis in Drosophila. Sleep 2024; 47:zsae096. [PMID: 38629438 PMCID: PMC11321855 DOI: 10.1093/sleep/zsae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/18/2024] [Indexed: 05/07/2024] Open
Abstract
The nuclear factor binding the κ light chain in B-cells (NFκB) is involved in a wide range of cellular processes including development, growth, innate immunity, and sleep. However, genetic studies of the role of specific NFκB transcription factors in sleep have been limited. Drosophila fruit flies carry three genes encoding NFκB transcription factors, Dorsal, Dorsal Immunity Factor (Dif), and Relish. We previously found that loss of the Relish gene from fat body suppressed daily nighttime sleep, and abolished infection-induced sleep. Here we show that Dif regulates daily sleep and recovery sleep following prolonged wakefulness. Mutants of Dif showed reduced daily sleep and suppressed recovery in response to sleep deprivation. Pan-neuronal knockdown of Dif strongly suppressed daily sleep, indicating that in contrast to Relish, Dif functions from the central nervous system to regulate sleep. Based on the unique expression pattern of a Dif- GAL4 driver, we hypothesized that its effects on sleep were mediated by the pars intercerebralis (PI). While RNAi knock-down of Dif in the PI reduced daily sleep, it had no effect on the recovery response to sleep deprivation. However, recovery sleep was suppressed when RNAi knock-down of Dif was distributed across a wider range of neurons. Induction of the nemuri (nur) antimicrobial peptide by sleep deprivation was reduced in Dif mutants and pan-neuronal overexpression of nur also suppressed the Dif mutant phenotype by significantly increasing sleep and reducing nighttime arousability. Together, these findings indicate that Dif functions from brain to target nemuri and to promote deep sleep.
Collapse
Affiliation(s)
- Michael K O’Hara
- Department of Neuroscience, Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | - Bumsik Cho
- Department of Neuroscience, Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Amita Sehgal
- Department of Neuroscience, Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Howard Hughes Medical Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Julie A Williams
- Department of Neuroscience, Chronobiology and Sleep Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
10
|
Dan C, Hulse BK, Kappagantula R, Jayaraman V, Hermundstad AM. A neural circuit architecture for rapid learning in goal-directed navigation. Neuron 2024; 112:2581-2599.e23. [PMID: 38795708 DOI: 10.1016/j.neuron.2024.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/16/2024] [Accepted: 04/30/2024] [Indexed: 05/28/2024]
Abstract
Anchoring goals to spatial representations enables flexible navigation but is challenging in novel environments when both representations must be acquired simultaneously. We propose a framework for how Drosophila uses internal representations of head direction (HD) to build goal representations upon selective thermal reinforcement. We show that flies use stochastically generated fixations and directed saccades to express heading preferences in an operant visual learning paradigm and that HD neurons are required to modify these preferences based on reinforcement. We used a symmetric visual setting to expose how flies' HD and goal representations co-evolve and how the reliability of these interacting representations impacts behavior. Finally, we describe how rapid learning of new goal headings may rest on a behavioral policy whose parameters are flexible but whose form is genetically encoded in circuit architecture. Such evolutionarily structured architectures, which enable rapidly adaptive behavior driven by internal representations, may be relevant across species.
Collapse
Affiliation(s)
- Chuntao Dan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Brad K Hulse
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Ramya Kappagantula
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Vivek Jayaraman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Ann M Hermundstad
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| |
Collapse
|
11
|
Zhang Y, Karadas M, Liu J, Gu X, Vöröslakos M, Li Y, Tsien RW, Buzsáki G. Interaction of acetylcholine and oxytocin neuromodulation in the hippocampus. Neuron 2024; 112:1862-1875.e5. [PMID: 38537642 PMCID: PMC11156550 DOI: 10.1016/j.neuron.2024.02.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/17/2024] [Accepted: 02/29/2024] [Indexed: 06/09/2024]
Abstract
A postulated role of subcortical neuromodulators is to control brain states. Mechanisms by which different neuromodulators compete or cooperate at various temporal scales remain an open question. We investigated the interaction of acetylcholine (ACh) and oxytocin (OXT) at slow and fast timescales during various brain states. Although these neuromodulators fluctuated in parallel during NREM packets, transitions from NREM to REM were characterized by a surge of ACh but a continued decrease of OXT. OXT signaling lagged behind ACh. High ACh was correlated with population synchrony and gamma oscillations during active waking, whereas minimum ACh predicts sharp-wave ripples (SPW-Rs). Optogenetic control of ACh and OXT neurons confirmed the active role of these neuromodulators in the observed correlations. Synchronous hippocampal activity consistently reduced OXT activity, whereas inactivation of the lateral septum-hypothalamus path attenuated this effect. Our findings demonstrate how cooperative actions of these neuromodulators allow target circuits to perform specific functions.
Collapse
Affiliation(s)
| | | | | | - Xinyi Gu
- Neuroscience Institute, New York, NY, USA
| | | | - Yulong Li
- School of Life Science, Peking University, Beijing, China
| | - Richard W Tsien
- Neuroscience Institute, New York, NY, USA; Department of Neurology, Langone Medical Center, New York University, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA
| | - György Buzsáki
- Neuroscience Institute, New York, NY, USA; Department of Neurology, Langone Medical Center, New York University, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
12
|
Le JQ, Ma D, Dai X, Rosbash M. Light and dopamine impact two circadian neurons to promote morning wakefulness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583333. [PMID: 38496661 PMCID: PMC10942368 DOI: 10.1101/2024.03.04.583333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
In both mammals and flies, circadian brain neurons orchestrate physiological oscillations and behaviors like wake and sleep; these neurons can be subdivided by morphology and by gene expression patterns. Recent single-cell sequencing studies identified 17 Drosophila circadian neuron groups. One of these include only two lateral neurons (LNs), which are marked by the expression of the neuropeptide ion transport peptide (ITP). Although these two ITP+ LNs have long been grouped with five other circadian evening activity cells, inhibiting the two neurons alone strongly reduces morning activity; this indicates that they are prominent morning neurons. As dopamine signaling promotes activity in Drosophila like in mammals, we considered that dopamine might influence this morning activity function. Moreover, the ITP+ LNs express higher mRNA levels than other LNs of the type 1-like dopamine receptor Dop1R1. Consistent with the importance of Dop1R1, CRISPR/Cas9 mutagenesis of this receptor only in the two ITP+ LNs renders flies significantly less active in the morning, and ex vivo live imaging shows that dopamine increases cAMP levels in these two neurons; cell-specific mutagenesis of Dop1R1 eliminates this cAMP response to dopamine. Notably, the response is more robust in the morning, reflecting higher morning Dop1R1 mRNA levels in the two neurons. As morning levels are not elevated in constant darkness, this suggests light-dependent upregulation of morning Dop1R1 transcript levels. Taken together with enhanced morning cAMP response to dopamine, the data indicate how light stimulates morning wakefulness in flies, which mimics the important effect of light on morning wakefulness in humans.
Collapse
Affiliation(s)
- Jasmine Quynh Le
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| | - Dingbang Ma
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Xihuimin Dai
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| |
Collapse
|
13
|
Abhilash L, Shafer OT. A two-process model of Drosophila sleep reveals an inter-dependence between circadian clock speed and the rate of sleep pressure decay. Sleep 2024; 47:zsad277. [PMID: 37930351 PMCID: PMC11275470 DOI: 10.1093/sleep/zsad277] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 10/18/2023] [Indexed: 11/07/2023] Open
Abstract
Sleep is controlled by two processes-a circadian clock that regulates its timing and a homeostat that regulates the drive to sleep. Drosophila has been an insightful model for understanding both processes. For four decades, Borbély and Daan's two-process model has provided a powerful framework for understanding sleep regulation. However, the field of fly sleep has not employed such a model as a framework for the investigation of sleep. To this end, we have adapted the two-process model to the fly and established its utility by showing that it can provide empirically testable predictions regarding the circadian and homeostatic control of fly sleep. We show that the ultradian rhythms previously reported for loss-of-function clock mutants in the fly are robustly detectable and a predictable consequence of a functional sleep homeostat in the absence of a functioning circadian system. We find that a model in which the circadian clock speed and homeostatic rates act without influencing each other provides imprecise predictions regarding how clock speed influences the strength of sleep rhythms and the amount of daily sleep. We also find that quantitatively good fits between empirical values and model predictions were achieved only when clock speeds were positively correlated with rates of decay of sleep pressure. Our results indicate that longer sleep bouts better reflect the homeostatic process than the current definition of sleep as any inactivity lasting 5 minutes or more. This two-process model represents a powerful framework for work on the molecular and physiological regulation of fly sleep.
Collapse
Affiliation(s)
- Lakshman Abhilash
- The Advanced Science Research Center, The City University of New York, New York, NY, USA
| | - Orie Thomas Shafer
- The Advanced Science Research Center, The City University of New York, New York, NY, USA
| |
Collapse
|
14
|
Dopp J, Ortega A, Davie K, Poovathingal S, Baz ES, Liu S. Single-cell transcriptomics reveals that glial cells integrate homeostatic and circadian processes to drive sleep-wake cycles. Nat Neurosci 2024; 27:359-372. [PMID: 38263460 PMCID: PMC10849968 DOI: 10.1038/s41593-023-01549-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024]
Abstract
The sleep-wake cycle is determined by circadian and sleep homeostatic processes. However, the molecular impact of these processes and their interaction in different brain cell populations are unknown. To fill this gap, we profiled the single-cell transcriptome of adult Drosophila brains across the sleep-wake cycle and four circadian times. We show cell type-specific transcriptomic changes, with glia displaying the largest variation. Glia are also among the few cell types whose gene expression correlates with both sleep homeostat and circadian clock. The sleep-wake cycle and sleep drive level affect the expression of clock gene regulators in glia, and disrupting clock genes specifically in glia impairs homeostatic sleep rebound after sleep deprivation. These findings provide a comprehensive view of the effects of sleep homeostatic and circadian processes on distinct cell types in an entire animal brain and reveal glia as an interaction site of these two processes to determine sleep-wake dynamics.
Collapse
Affiliation(s)
- Joana Dopp
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Antonio Ortega
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Kristofer Davie
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Suresh Poovathingal
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - El-Sayed Baz
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Zoology Department, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Sha Liu
- Center for Brain & Disease Research, VIB-KU Leuven, Leuven, Belgium.
- Department of Neurosciences, KU Leuven, Leuven, Belgium.
- Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
15
|
Tabuchi M. Dynamic neuronal instability generates synaptic plasticity and behavior: Insights from Drosophila sleep. Neurosci Res 2024; 198:1-7. [PMID: 37385545 PMCID: PMC11033711 DOI: 10.1016/j.neures.2023.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/05/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
How do neurons encode the information that underlies cognition, internal states, and behavior? This review focuses on the neural circuit mechanisms underlying sleep in Drosophila and, to illustrate the power of addressing neural coding in this system, highlights a specific circuit mediating the circadian regulation of sleep quality. This circuit exhibits circadian cycling of sleep quality, which depends solely on the pattern (not the rate) of spiking. During the night, the stability of spike waveforms enhances the reliability of spike timing in these neurons to promote sleep quality. During the day, instability of the spike waveforms leads to uncertainty of spike timing, which remarkably produces synaptic plasticity to induce arousal. Investigation of the molecular and biophysical basis of these changes was greatly facilitated by its study in Drosophila, revealing direct connections between genes, molecules, spike biophysical properties, neural codes, synaptic plasticity, and behavior. Furthermore, because these patterns of neural activity change with aging, this model system holds promise for understanding the interplay between the circadian clock, aging, and sleep quality. It is proposed here that neurophysiological investigations of the Drosophila brain present an exceptional opportunity to tackle some of the most challenging questions related to neural coding.
Collapse
Affiliation(s)
- Masashi Tabuchi
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States.
| |
Collapse
|
16
|
Adams GJ, O'Brien PA. The unified theory of sleep: Eukaryotes endosymbiotic relationship with mitochondria and REM the push-back response for awakening. Neurobiol Sleep Circadian Rhythms 2023; 15:100100. [PMID: 37484687 PMCID: PMC10362302 DOI: 10.1016/j.nbscr.2023.100100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
The Unified Theory suggests that sleep is a process that developed in eukaryotic animals from a relationship with an endosymbiotic bacterium. Over evolutionary time the bacterium evolved into the modern mitochondrion that continues to exert an effect on sleep patterns, e.g. the bacterium Wolbachia establishes an endosymbiotic relationship with Drosophila and many other species of insects and is able to change the host's behaviour by making it sleep. The hypothesis is supported by other host-parasite relationships, e.g., Trypanosoma brucei which causes day-time sleepiness and night-time insomnia in humans and cattle. For eukaryotes such as Monocercomonoids that don't contain mitochondria we find no evidence of them sleeping. Mitochondria produce the neurotransmitter gamma aminobutyric acid (GABA), and ornithine a precursor of the neurotransmitter GABA, together with substances such as 3,4dihydroxy phenylalanine (DOPA) a precursor for the neurotransmitter dopamine: These substances have been shown to affect the sleep/wake cycles in animals such as Drosophilia and Hydra. Eukaryote animals have traded the very positive side of having mitochondria providing aerobic respiration for them with the negative side of having to sleep. NREM (Quiet sleep) is the process endosymbionts have imposed upon their host eukaryotes and REM (Active sleep) is the push-back adaptation of eukaryotes with brains, returning to wakefulness.
Collapse
Affiliation(s)
| | - Philip A. O'Brien
- College of Science, Health, Engineering and Education, Murdoch University, WA, Australia
| |
Collapse
|
17
|
Axelrod S, Li X, Sun Y, Lincoln S, Terceros A, O’Neil J, Wang Z, Nguyen A, Vora A, Spicer C, Shapiro B, Young MW. The Drosophila blood-brain barrier regulates sleep via Moody G protein-coupled receptor signaling. Proc Natl Acad Sci U S A 2023; 120:e2309331120. [PMID: 37831742 PMCID: PMC10589661 DOI: 10.1073/pnas.2309331120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/28/2023] [Indexed: 10/15/2023] Open
Abstract
Sleep is vital for most animals, yet its mechanism and function remain unclear. We found that permeability of the BBB (blood-brain barrier)-the organ required for the maintenance of homeostatic levels of nutrients, ions, and other molecules in the brain-is modulated by sleep deprivation (SD) and can cell-autonomously effect sleep changes. We observed increased BBB permeability in known sleep mutants as well as in acutely sleep-deprived animals. In addition to molecular tracers, SD-induced BBB changes also increased the penetration of drugs used in the treatment of brain pathologies. After chronic/genetic or acute SD, rebound sleep or administration of the sleeping aid gaboxadol normalized BBB permeability, showing that SD effects on the BBB are reversible. Along with BBB permeability, RNA levels of the BBB master regulator moody are modulated by sleep. Conversely, altering BBB permeability alone through glia-specific modulation of moody, gαo, loco, lachesin, or neuroglian-each a well-studied regulator of BBB function-was sufficient to induce robust sleep phenotypes. These studies demonstrate a tight link between BBB permeability and sleep and indicate a unique role for the BBB in the regulation of sleep.
Collapse
Affiliation(s)
- Sofia Axelrod
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Xiaoling Li
- International Personalized Cancer Center, Tianjin Cancer Hospital Airport Hospital, Tianjin300308, China
| | - Yingwo Sun
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Samantha Lincoln
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrea Terceros
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Jenna O’Neil
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Andrew Nguyen
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Aabha Vora
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Carmen Spicer
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Benjamin Shapiro
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| | - Michael W. Young
- Laboratory of Genetics, The Rockefeller University, New York, NY10065
| |
Collapse
|
18
|
O’Hara MK, Saul C, Handa A, Sehgal A, Williams JA. The NFκB Dif is required for behavioral and molecular correlates of sleep homeostasis in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.12.562029. [PMID: 37905096 PMCID: PMC10614778 DOI: 10.1101/2023.10.12.562029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The nuclear factor binding the κ light chain in B-cells (NFκB) is involved in a wide range of cellular processes including development, growth, innate immunity, and sleep. However, efforts have been limited toward understanding how specific NFκB transcription factors function in sleep. Drosophila fruit flies carry three genes encoding NFκB transcription factors, Dorsal, Dorsal Immunity Factor (Dif), and Relish. We previously found that loss of the Relish gene from fat body suppressed daily nighttime sleep, and abolished infection-induced sleep. Here we show that Dif regulates daily sleep and recovery sleep following prolonged wakefulness. Mutants of Dif showed reduced daily sleep and suppressed recovery in response to sleep deprivation. Pan-neuronal knockdown of Dif strongly suppressed daily sleep, indicating that in contrast to Relish, Dif functions from the central nervous system to regulate sleep. Based on the distribution of a Dif-associated GAL4 driver, we hypothesized that its effects on sleep were mediated by the pars intercerebralis (PI). While RNAi knock-down of Dif in the PI reduced daily sleep, it had no effect on the recovery response to sleep deprivation. However, recovery sleep was suppressed when RNAi knock-down of Dif was distributed across a wider range of neurons. Induction of the nemuri (nur) antimicrobial peptide by sleep deprivation was suppressed in Dif mutants and pan-neuronal over-expression of nur also suppressed the Dif mutant phenotype. Together, these findings indicate that Dif functions from brain to target nemuri and to promote sleep.
Collapse
Affiliation(s)
| | | | | | - Amita Sehgal
- Chronobiology and Sleep Institute, Department of Neuroscience
- Howard Hughes Medical Institute, University of Pennsylvania Perelman School of Medicine Philadelphia, PA 19104
| | | |
Collapse
|
19
|
Koutsoumparis A, Busack I, Chen CK, Hayashi Y, Braeckman BP, Meierhofer D, Bringmann H. Reverse genetic screening during L1 arrest reveals a role of the diacylglycerol kinase 1 gene dgk-1 and sphingolipid metabolism genes in sleep regulation. Genetics 2023; 225:iyad124. [PMID: 37682641 DOI: 10.1093/genetics/iyad124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 07/01/2023] [Indexed: 09/10/2023] Open
Abstract
Sleep is a fundamental state of behavioral quiescence and physiological restoration. Sleep is controlled by environmental conditions, indicating a complex regulation of sleep by multiple processes. Our knowledge of the genes and mechanisms that control sleep during various conditions is, however, still incomplete. In Caenorhabditis elegans, sleep is increased when development is arrested upon starvation. Here, we performed a reverse genetic sleep screen in arrested L1 larvae for genes that are associated with metabolism. We found over 100 genes that are associated with a reduced sleep phenotype. Enrichment analysis revealed sphingolipid metabolism as a key pathway that controls sleep. A strong sleep loss was caused by the loss of function of the diacylglycerol kinase 1 gene, dgk-1, a negative regulator of synaptic transmission. Rescue experiments indicated that dgk-1 is required for sleep in cholinergic and tyraminergic neurons. The Ring Interneuron S (RIS) neuron is crucial for sleep in C. elegans and activates to induce sleep. RIS activation transients were abolished in dgk-1 mutant animals. Calcium transients were partially rescued by a reduction-of-function mutation of unc-13, suggesting that dgk-1 might be required for RIS activation by limiting synaptic vesicle release. dgk-1 mutant animals had impaired L1 arrest survival and dampened expression of the protective heat shock factor gene hsp-12.6. These data suggest that dgk-1 impairment causes broad physiological deficits. Microcalorimetry and metabolomic analyses of larvae with impaired RIS showed that RIS is broadly required for energy conservation and metabolic control, including for the presence of sphingolipids. Our data support the notion that metabolism broadly influences sleep and that sleep is associated with profound metabolic changes. We thus provide novel insights into the interplay of lipids and sleep and provide a rich resource of mutants and metabolic pathways for future sleep studies.
Collapse
Affiliation(s)
- Anastasios Koutsoumparis
- Chair of Cellular Circuits and Systems, Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Am Tatzberg 47/49, Dresden, Saxony 01307, Germany
| | - Inka Busack
- Chair of Cellular Circuits and Systems, Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Am Tatzberg 47/49, Dresden, Saxony 01307, Germany
| | - Chung-Kuan Chen
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Yu Hayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Bart P Braeckman
- Laboratory of Aging Physiology and Molecular Evolution, Department of Biology, Ghent University, 9000 Ghent, Belgium
| | - David Meierhofer
- Mass Spectrometry Facility, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Henrik Bringmann
- Chair of Cellular Circuits and Systems, Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Am Tatzberg 47/49, Dresden, Saxony 01307, Germany
| |
Collapse
|
20
|
Wani AR, Chowdhury B, Luong J, Chaya GM, Patel K, Isaacman-Beck J, Shafer O, Kayser MS, Syed MH. Stem cell-specific ecdysone signaling regulates the development and function of a Drosophila sleep homeostat. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560022. [PMID: 37873323 PMCID: PMC10592846 DOI: 10.1101/2023.09.29.560022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Complex behaviors arise from neural circuits that are assembled from diverse cell types. Sleep is a conserved and essential behavior, yet little is known regarding how the nervous system generates neuron types of the sleep-wake circuit. Here, we focus on the specification of Drosophila sleep-promoting neurons-long-field tangential input neurons that project to the dorsal layers of the fan-shaped body neuropil in the central complex (CX). We use lineage analysis and genetic birth dating to identify two bilateral Type II neural stem cells that generate these dorsal fan-shaped body (dFB) neurons. We show that adult dFB neurons express Ecdysone-induced protein E93, and loss of Ecdysone signaling or E93 in Type II NSCs results in the misspecification of the adult dFB neurons. Finally, we show that E93 knockdown in Type II NSCs affects adult sleep behavior. Our results provide insight into how extrinsic hormonal signaling acts on NSCs to generate neuronal diversity required for adult sleep behavior. These findings suggest that some adult sleep disorders might derive from defects in stem cell-specific temporal neurodevelopmental programs.
Collapse
Affiliation(s)
- Adil R Wani
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, 87131 Albuquerque, NM, USA
| | - Budhaditya Chowdhury
- The Advanced Science Research Center, City University of New York, New York, NY 10031, USA
| | - Jenny Luong
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gonzalo Morales Chaya
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, 87131 Albuquerque, NM, USA
| | - Krishna Patel
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, 87131 Albuquerque, NM, USA
| | | | - Orie Shafer
- The Advanced Science Research Center, City University of New York, New York, NY 10031, USA
| | - Matthew S. Kayser
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Chronobiology Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mubarak Hussain Syed
- Neural Diversity Lab, Department of Biology, University of New Mexico, 219 Yale Blvd Ne, 87131 Albuquerque, NM, USA
| |
Collapse
|
21
|
Cuddapah VA, Hsu CT, Li Y, Shah HM, Saul C, Killiany S, Shon J, Yue Z, Gionet G, Putt ME, Sehgal A. Sleepiness, not total sleep amount, increases seizure risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560325. [PMID: 37873373 PMCID: PMC10592838 DOI: 10.1101/2023.09.30.560325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Sleep loss has been associated with increased seizure risk since antiquity. Despite this observation standing the test of time, how poor sleep drives susceptibility to seizures remains unclear. To identify underlying mechanisms, we restricted sleep in Drosophila epilepsy models and developed a method to identify spontaneous seizures using quantitative video tracking. Here we find that sleep loss exacerbates seizures but only when flies experience increased sleep need, or sleepiness , and not necessarily with reduced sleep quantity. This is supported by the paradoxical finding that acute activation of sleep-promoting circuits worsens seizures, because it increases sleep need without changing sleep amount. Sleep-promoting circuits become hyperactive after sleep loss and are associated with increased whole-brain activity. During sleep restriction, optogenetic inhibition of sleep-promoting circuits to reduce sleepiness protects against seizures. Downregulation of the 5HT1A serotonin receptor in sleep-promoting cells mediates the effect of sleep need on seizures, and we identify an FDA-approved 5HT1A agonist to mitigate seizures. Our findings demonstrate that while homeostatic sleep is needed to recoup lost sleep, it comes at the cost of increasing seizure susceptibility. We provide an unexpected perspective on interactions between sleep and seizures, and surprisingly implicate sleep- promoting circuits as a therapeutic target for seizure control.
Collapse
|
22
|
De J, Wu M, Lambatan V, Hua Y, Joiner WJ. Re-examining the role of the dorsal fan-shaped body in promoting sleep in Drosophila. Curr Biol 2023; 33:3660-3668.e4. [PMID: 37552985 PMCID: PMC10573663 DOI: 10.1016/j.cub.2023.07.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/10/2023] [Accepted: 07/20/2023] [Indexed: 08/10/2023]
Abstract
The needs fulfilled by sleep are unknown, though the effects of insufficient sleep are manifold. To better understand how the need to sleep is sensed and discharged, much effort has gone into identifying the neural circuits involved in regulating arousal, especially those that promote sleep. In prevailing models, the dorsal fan-shaped body (dFB) plays a central role in this process in the fly brain. In the present study we manipulated various properties of the dFB including its electrical activity, synaptic output, and endogenous gene expression. In each of these experimental contexts we were unable to identify any effect on sleep that could be unambiguously mapped to the dFB. Furthermore, we found evidence that sleep phenotypes previously attributed to the dFB were caused by genetic manipulations that inadvertently targeted the ventral nerve cord. We also examined expression of two genes whose purported effects have been attributed to functions within a specific subpopulation of dFB neurons. In both cases we found little to no expression in the expected cells. Collectively, our results cast doubt on the prevailing hypothesis that the dFB plays a central role in promoting sleep.
Collapse
Affiliation(s)
- Joydeep De
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Meilin Wu
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vanessa Lambatan
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yue Hua
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - William J Joiner
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA; Center for Circadian Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
23
|
Kniazkina M, Dyachuk V. Does EGFR Signaling Mediate Orexin System Activity in Sleep Initiation? Int J Mol Sci 2023; 24:ijms24119505. [PMID: 37298454 DOI: 10.3390/ijms24119505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Sleep-wake cycle disorders are an important symptom of many neurological diseases, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Circadian rhythms and sleep-wake cycles play a key role in maintaining the health of organisms. To date, these processes are still poorly understood and, therefore, need more detailed elucidation. The sleep process has been extensively studied in vertebrates, such as mammals and, to a lesser extent, in invertebrates. A complex, multi-step interaction of homeostatic processes and neurotransmitters provides the sleep-wake cycle. Many other regulatory molecules are also involved in the cycle regulation, but their functions remain largely unclear. One of these signaling systems is epidermal growth factor receptor (EGFR), which regulates the activity of neurons in the modulation of the sleep-wake cycle in vertebrates. We have evaluated the possible role of the EGFR signaling pathway in the molecular regulation of sleep. Understanding the molecular mechanisms that underlie sleep-wake regulation will provide critical insight into the fundamental regulatory functions of the brain. New findings of sleep-regulatory pathways may provide new drug targets and approaches for the treatment of sleep-related diseases.
Collapse
Affiliation(s)
- Marina Kniazkina
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Vyacheslav Dyachuk
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| |
Collapse
|
24
|
Jones JD, Holder BL, Eiken KR, Vogt A, Velarde AI, Elder AJ, McEllin JA, Dissel S. Regulation of sleep by cholinergic neurons located outside the central brain in Drosophila. PLoS Biol 2023; 21:e3002012. [PMID: 36862736 PMCID: PMC10013921 DOI: 10.1371/journal.pbio.3002012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 03/14/2023] [Accepted: 01/25/2023] [Indexed: 03/03/2023] Open
Abstract
Sleep is a complex and plastic behavior regulated by multiple brain regions and influenced by numerous internal and external stimuli. Thus, to fully uncover the function(s) of sleep, cellular resolution of sleep-regulating neurons needs to be achieved. Doing so will help to unequivocally assign a role or function to a given neuron or group of neurons in sleep behavior. In the Drosophila brain, neurons projecting to the dorsal fan-shaped body (dFB) have emerged as a key sleep-regulating area. To dissect the contribution of individual dFB neurons to sleep, we undertook an intersectional Split-GAL4 genetic screen focusing on cells contained within the 23E10-GAL4 driver, the most widely used tool to manipulate dFB neurons. In this study, we demonstrate that 23E10-GAL4 expresses in neurons outside the dFB and in the fly equivalent of the spinal cord, the ventral nerve cord (VNC). Furthermore, we show that 2 VNC cholinergic neurons strongly contribute to the sleep-promoting capacity of the 23E10-GAL4 driver under baseline conditions. However, in contrast to other 23E10-GAL4 neurons, silencing these VNC cells does not block sleep homeostasis. Thus, our data demonstrate that the 23E10-GAL4 driver contains at least 2 different types of sleep-regulating neurons controlling distinct aspects of sleep behavior.
Collapse
Affiliation(s)
- Joseph D. Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Brandon L. Holder
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Kiran R. Eiken
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Alex Vogt
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Adriana I. Velarde
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Alexandra J. Elder
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Jennifer A. McEllin
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Stephane Dissel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| |
Collapse
|
25
|
Ma D, Herndon N, Le JQ, Abruzzi KC, Zinn K, Rosbash M. Neural connectivity molecules best identify the heterogeneous clock and dopaminergic cell types in the Drosophila adult brain. SCIENCE ADVANCES 2023; 9:eade8500. [PMID: 36812309 PMCID: PMC9946362 DOI: 10.1126/sciadv.ade8500] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/26/2023] [Indexed: 05/25/2023]
Abstract
Our recent single-cell sequencing of most adult Drosophila circadian neurons indicated notable and unexpected heterogeneity. To address whether other populations are similar, we sequenced a large subset of adult brain dopaminergic neurons. Their gene expression heterogeneity is similar to that of clock neurons, i.e., both populations have two to three cells per neuron group. There was also unexpected cell-specific expression of neuron communication molecule messenger RNAs: G protein-coupled receptor or cell surface molecule (CSM) transcripts alone can define adult brain dopaminergic and circadian neuron cell type. Moreover, the adult expression of the CSM DIP-beta in a small group of clock neurons is important for sleep. We suggest that the common features of circadian and dopaminergic neurons are general, essential for neuronal identity and connectivity of the adult brain, and that these features underlie the complex behavioral repertoire of Drosophila.
Collapse
Affiliation(s)
- Dingbang Ma
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Nicholas Herndon
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Jasmine Quynh Le
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Katharine C. Abruzzi
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| |
Collapse
|
26
|
Neuropeptide diuretic hormone 31 mediates memory and sleep via distinct neural pathways in Drosophila. Neurosci Res 2023:S0168-0102(23)00037-8. [PMID: 36780946 DOI: 10.1016/j.neures.2023.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/13/2023]
Abstract
Memory formation and sleep regulation are critical for brain functions in animals from invertebrates to humans. Neuropeptides play a pivotal role in regulating physiological behaviors, including memory formation and sleep. However, the detailed mechanisms by which neuropeptides regulate these physiological behaviors remains unclear. Herein, we report that neuropeptide diuretic hormone 31 (DH31) positively regulates memory formation and sleep in Drosophila melanogaster. The expression of DH31 in the dorsal and ventral fan-shaped body (dFB and vFB) neurons of the central complex and ventral lateral clock neurons (LNvs) in the brain was responsive to sleep regulation. In addition, the expression of membrane-tethered DH31 in dFB neurons rescued sleep defects in Dh31 mutants, suggesting that DH31 secreted from dFB, vFB, and LNvs acts on the DH31 receptor in the dFB to regulate sleep partly in an autoregulatory feedback loop. Moreover, the expression of DH31 in octopaminergic neurons, but not in the dFB neurons, is involved in forming intermediate-term memory. Our results suggest that DH31 regulates memory formation and sleep through distinct neural pathways.
Collapse
|
27
|
Zhang T, Wang X, Li X, Li YN, Li Y, Wu S, Xu L, Zhou R, Yang J, Li G, Liu X, Zheng X, Zhang Z, Zhang H. MoLrp1-mediated signaling induces nuclear accumulation of MoMsn2 to facilitate fatty acid oxidation for infectious growth of the rice blast fungus. PLANT COMMUNICATIONS 2023:100561. [PMID: 36774535 PMCID: PMC10363509 DOI: 10.1016/j.xplc.2023.100561] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 01/05/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Fatty acid β-oxidation is critical for fatty acid degradation and cellular development. In the rice blast fungus Magnaporthe oryzae, fatty acid β-oxidation is reported to be important mainly for turgor generation in the appressorium. However, the role of fatty acid β-oxidation during invasive hyphal growth is rarely documented. We demonstrated that blocking peroxisomal fatty acid β-oxidation impaired lipid droplet (LD) degradation and infectious growth of M. oryzae. We found that the key regulator of pathogenesis, MoMsn2, which we identified previously, is involved in fatty acid β-oxidation by targeting MoDCI1 (encoding dienoyl-coenzyme A [CoA] isomerase), which is also important for LD degradation and infectious growth. Cytological observations revealed that MoMsn2 accumulated from the cytosol to the nucleus during early infection or upon treatment with oleate. We determined that the low-density lipoprotein receptor-related protein MoLrp1, which is also involved in fatty acid β-oxidation and infectious growth, plays a critical role in the accumulation of MoMsn2 from the cytosol to the nucleus by activating the cyclic AMP signaling pathway. Our results provide new insights into the importance of fatty acid oxidation during invasive hyphal growth, which is modulated by MoMsn2 and its related signaling pathways in M. oryzae.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xingyu Wang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xue Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Ya-Nan Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Yuhe Li
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Shuang Wu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Lele Xu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Ruiwen Zhou
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Jing Yang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Guotian Li
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, the Provincial Key Laboratory of Plant Pathology of Hubei Province, College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, China
| | - Xinyu Liu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Xiaobo Zheng
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Zhengguang Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China
| | - Haifeng Zhang
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, and Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, Nanjing, China; The Key Laboratory of Plant Immunity, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
28
|
Yan W, Lin H, Yu J, Wiggin TD, Wu L, Meng Z, Liu C, Griffith LC. Subtype-Specific Roles of Ellipsoid Body Ring Neurons in Sleep Regulation in Drosophila. J Neurosci 2023; 43:764-786. [PMID: 36535771 PMCID: PMC9899086 DOI: 10.1523/jneurosci.1350-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/22/2022] [Accepted: 10/26/2022] [Indexed: 12/23/2022] Open
Abstract
The ellipsoid body (EB) is a major structure of the central complex of the Drosophila melanogaster brain. Twenty-two subtypes of EB ring neurons have been identified based on anatomic and morphologic characteristics by light-level microscopy and EM connectomics. A few studies have associated ring neurons with the regulation of sleep homeostasis and structure. However, cell type-specific and population interactions in the regulation of sleep remain unclear. Using an unbiased thermogenetic screen of EB drivers using female flies, we found the following: (1) multiple ring neurons are involved in the modulation of amount of sleep and structure in a synergistic manner; (2) analysis of data for ΔP(doze)/ΔP(wake) using a mixed Gaussian model detected 5 clusters of GAL4 drivers which had similar effects on sleep pressure and/or depth: lines driving arousal contained R4m neurons, whereas lines that increased sleep pressure had R3m cells; (3) a GLM analysis correlating ring cell subtype and activity-dependent changes in sleep parameters across all lines identified several cell types significantly associated with specific sleep effects: R3p was daytime sleep-promoting, and R4m was nighttime wake-promoting; and (4) R3d cells present in 5HT7-GAL4 and in GAL4 lines, which exclusively affect sleep structure, were found to contribute to fragmentation of sleep during both day and night. Thus, multiple subtypes of ring neurons distinctively control sleep amount and/or structure. The unique highly interconnected structure of the EB suggests a local-network model worth future investigation; understanding EB subtype interactions may provide insight how sleep circuits in general are structured.SIGNIFICANCE STATEMENT How multiple brain regions, with many cell types, can coherently regulate sleep remains unclear, but identification of cell type-specific roles can generate opportunities for understanding the principles of integration and cooperation. The ellipsoid body (EB) of the fly brain exhibits a high level of connectivity and functional heterogeneity yet is able to tune multiple behaviors in real-time, including sleep. Leveraging the powerful genetic tools available in Drosophila and recent progress in the characterization of the morphology and connectivity of EB ring neurons, we identify several EB subtypes specifically associated with distinct aspects of sleep. Our findings will aid in revealing the rules of coding and integration in the brain.
Collapse
Affiliation(s)
- Wei Yan
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518000, China
| | - Hai Lin
- Central Research Institute, United Imaging Healthcare, Shanghai, 200032, China
| | - Junwei Yu
- Department of Biology, National Center for Behavioral Genomics and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02453
| | - Timothy D Wiggin
- Department of Biology, National Center for Behavioral Genomics and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02453
| | - Litao Wu
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518000, China
| | - Zhiqiang Meng
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518000, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen, 518000, China
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen, 518000, China
| | - Chang Liu
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518000, China
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen, 518000, China
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen, 518000, China
| | - Leslie C Griffith
- Department of Biology, National Center for Behavioral Genomics and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02453
| |
Collapse
|
29
|
Liang X, Holy TE, Taghert PH. Polyphasic circadian neural circuits drive differential activities in multiple downstream rhythmic centers. Curr Biol 2023; 33:351-363.e3. [PMID: 36610393 PMCID: PMC9877191 DOI: 10.1016/j.cub.2022.12.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 01/08/2023]
Abstract
Circadian clocks align various behaviors such as locomotor activity, sleep/wake, feeding, and mating to times of day that are most adaptive. How rhythmic information in pacemaker circuits is translated to neuronal outputs is not well understood. Here, we used brain-wide, 24-h in vivo calcium imaging in the Drosophila brain and searched for circadian rhythmic activity among identified clusters of dopaminergic (DA) and peptidergic neurosecretory (NS) neurons. Such rhythms were widespread and imposed by the PERIOD-dependent clock activity within the ∼150-cell circadian pacemaker network. The rhythms displayed either a morning (M), evening (E), or mid-day (MD) phase. Different subgroups of circadian pacemakers imposed neural activity rhythms onto different downstream non-clock neurons. Outputs from the canonical M and E pacemakers converged to regulate DA-PPM3 and DA-PAL neurons. E pacemakers regulate the evening-active DA-PPL1 neurons. In addition to these canonical M and E oscillators, we present evidence for a third dedicated phase occurring at mid-day: the l-LNv pacemakers present the MD activity peak, and they regulate the MD-active DA-PPM1/2 neurons and three distinct NS cell types. Thus, the Drosophila circadian pacemaker network is a polyphasic rhythm generator. It presents dedicated M, E, and MD phases that are functionally transduced as neuronal outputs to organize diverse daily activity patterns in downstream circuits.
Collapse
Affiliation(s)
- Xitong Liang
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Timothy E Holy
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Paul H Taghert
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
30
|
Marquand K, Roselli C, Cervantes-Sandoval I, Boto T. Sleep benefits different stages of memory in Drosophila. Front Physiol 2023; 14:1087025. [PMID: 36744027 PMCID: PMC9892949 DOI: 10.3389/fphys.2023.1087025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Understanding the physiological mechanisms that modulate memory acquisition and consolidation remains among the most ambitious questions in neuroscience. Massive efforts have been dedicated to deciphering how experience affects behavior, and how different physiological and sensory phenomena modulate memory. Our ability to encode, consolidate and retrieve memories depends on internal drives, and sleep stands out among the physiological processes that affect memory: one of the most relatable benefits of sleep is the aiding of memory that occurs in order to both prepare the brain to learn new information, and after a learning task, to consolidate those new memories. Drosophila lends itself to the study of the interactions between memory and sleep. The fruit fly provides incomparable genetic resources, a mapped connectome, and an existing framework of knowledge on the molecular, cellular, and circuit mechanisms of memory and sleep, making the fruit fly a remarkable model to decipher the sophisticated regulation of learning and memory by the quantity and quality of sleep. Research in Drosophila has stablished not only that sleep facilitates learning in wild-type and memory-impaired animals, but that sleep deprivation interferes with the acquisition of new memories. In addition, it is well-accepted that sleep is paramount in memory consolidation processes. Finally, studies in Drosophila have shown that that learning itself can promote sleep drive. Nevertheless, the molecular and network mechanisms underlying this intertwined relationship are still evasive. Recent remarkable work has shed light on the neural substrates that mediate sleep-dependent memory consolidation. In a similar way, the mechanistic insights of the neural switch control between sleep-dependent and sleep-independent consolidation strategies were recently described. This review will discuss the regulation of memory by sleep in Drosophila, focusing on the most recent advances in the field and pointing out questions awaiting to be investigated.
Collapse
Affiliation(s)
- Katie Marquand
- Department of Physiology, School of Medicine, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Camilla Roselli
- Trinity College Institute of Neuroscience, School of Genetics and Microbiology, Smurfit Institute of Genetics and School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | - Isaac Cervantes-Sandoval
- Department of Biology, Georgetown University, Washington, DC, United States
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
| | - Tamara Boto
- Department of Physiology, School of Medicine, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Damulewicz M, Tyszka A, Pyza E. Light exposure during development affects physiology of adults in Drosophila melanogaster. Front Physiol 2022; 13:1008154. [PMID: 36505068 PMCID: PMC9732085 DOI: 10.3389/fphys.2022.1008154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Light is one of most important factors synchronizing organisms to day/night cycles in the environment. In Drosophila it is received through compound eyes, Hofbauer-Buchner eyelet, ocelli, using phospholipase C-dependent phototransduction and by deep brain photoreceptors, like Cryptochrome. Even a single light pulse during early life induces larval-time memory, which synchronizes the circadian clock and maintains daily rhythms in adult flies. In this study we investigated several processes in adult flies after maintaining their embryos, larvae and pupae in constant darkness (DD) until eclosion. We found that the lack of external light during development affects sleep time, by reduction of night sleep, and in effect shift to the daytime. However, disruption of internal CRY- dependent photoreception annuls this effect. We also observed changes in the expression of genes encoding neurotransmitters and their receptors between flies kept in different light regime. In addition, the lack of light during development results in decreasing size of mushroom bodies, involved in sleep regulation. Taking together, our results show that presence of light during early life plays a key role in brain development and affects adult behavior.
Collapse
|
32
|
Ryu TH, Subramanian M, Yeom E, Yu K. The prominin-like Gene Expressed in a Subset of Dopaminergic Neurons Regulates Locomotion in Drosophila. Mol Cells 2022; 45:640-648. [PMID: 35993164 PMCID: PMC9448647 DOI: 10.14348/molcells.2022.0006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/15/2022] [Accepted: 05/02/2022] [Indexed: 11/27/2022] Open
Abstract
CD133, also known as prominin-1, was first identified as a biomarker of mammalian cancer and neural stem cells. Previous studies have shown that the prominin-like (promL) gene, an orthologue of mammalian CD133 in Drosophila, plays a role in glucose and lipid metabolism, body growth, and longevity. Because locomotion is required for food sourcing and ultimately the regulation of metabolism, we examined the function of promL in Drosophila locomotion. Both promL mutants and pan-neuronal promL inhibition flies displayed reduced spontaneous locomotor activity. As dopamine is known to modulate locomotion, we also examined the effects of promL inhibition on the dopamine concentration and mRNA expression levels of tyrosine hydroxylase (TH) and DOPA decarboxylase (Ddc), the enzymes responsible for dopamine biosynthesis, in the heads of flies. Compared with those in control flies, the levels of dopamine and the mRNAs encoding TH and Ddc were lower in promL mutant and pan-neuronal promL inhibition flies. In addition, an immunostaining analysis revealed that, compared with control flies, promL mutant and pan-neuronal promL inhibition flies had lower levels of the TH protein in protocerebral anterior medial (PAM) neurons, a subset of dopaminergic neurons. Inhibition of promL in these PAM neurons reduced the locomotor activity of the flies. Overall, these findings indicate that promL expressed in PAM dopaminergic neurons regulates locomotion by controlling dopamine synthesis in Drosophila.
Collapse
Affiliation(s)
- Tae Hoon Ryu
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Manivannan Subramanian
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Eunbyul Yeom
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Kweon Yu
- Metabolism and Neurophysiology Research Group, Disease Target Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
33
|
Dissel S, Klose MK, van Swinderen B, Cao L, Ford M, Periandri EM, Jones JD, Li Z, Shaw PJ. Sleep-promoting neurons remodel their response properties to calibrate sleep drive with environmental demands. PLoS Biol 2022; 20:e3001797. [PMID: 36173939 PMCID: PMC9521806 DOI: 10.1371/journal.pbio.3001797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 08/16/2022] [Indexed: 01/29/2023] Open
Abstract
Falling asleep at the wrong time can place an individual at risk of immediate physical harm. However, not sleeping degrades cognition and adaptive behavior. To understand how animals match sleep need with environmental demands, we used live-brain imaging to examine the physiological response properties of the dorsal fan-shaped body (dFB) following interventions that modify sleep (sleep deprivation, starvation, time-restricted feeding, memory consolidation) in Drosophila. We report that dFB neurons change their physiological response-properties to dopamine (DA) and allatostatin-A (AstA) in response to different types of waking. That is, dFB neurons are not simply passive components of a hard-wired circuit. Rather, the dFB neurons intrinsically regulate their response to the activity from upstream circuits. Finally, we show that the dFB appears to contain a memory trace of prior exposure to metabolic challenges induced by starvation or time-restricted feeding. Together, these data highlight that the sleep homeostat is plastic and suggests an underlying mechanism.
Collapse
Affiliation(s)
- Stephane Dissel
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
- * E-mail: (SD); (PJS)
| | - Markus K. Klose
- University of Pittsburgh School of Medicine, Department of Pharmacology & Chemical Biology, Pittsburgh, Pennsylvania, United States of America
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, St Lucia, Australia
| | - Lijuan Cao
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Melanie Ford
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Erica M. Periandri
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Joseph D. Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Zhaoyi Li
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul J. Shaw
- Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail: (SD); (PJS)
| |
Collapse
|
34
|
Gong NN, Luong HNB, Dang AH, Mainwaring B, Shields E, Schmeckpeper K, Bonasio R, Kayser MS. Intrinsic maturation of sleep output neurons regulates sleep ontogeny in Drosophila. Curr Biol 2022; 32:4025-4039.e3. [PMID: 35985328 PMCID: PMC9529826 DOI: 10.1016/j.cub.2022.07.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 06/06/2022] [Accepted: 07/21/2022] [Indexed: 11/19/2022]
Abstract
The maturation of sleep behavior across a lifespan (sleep ontogeny) is an evolutionarily conserved phenomenon. Mammalian studies have shown that in addition to increased sleep duration, early life sleep exhibits stark differences compared with mature sleep with regard to sleep states. How the intrinsic maturation of sleep output circuits contributes to sleep ontogeny is poorly understood. The fruit fly Drosophila melanogaster exhibits multifaceted changes to sleep from juvenile to mature adulthood. Here, we use a non-invasive probabilistic approach to investigate the changes in sleep architecture in juvenile and mature flies. Increased sleep in juvenile flies is driven primarily by a decreased probability of transitioning to wake and characterized by more time in deeper sleep states. Functional manipulations of sleep-promoting neurons in the dorsal fan-shaped body (dFB) suggest that these neurons differentially regulate sleep in juvenile and mature flies. Transcriptomic analysis of dFB neurons at different ages and a subsequent RNAi screen implicate the genes involved in dFB sleep circuit maturation. These results reveal that the dynamic transcriptional states of sleep output neurons contribute to the changes in sleep across the lifespan.
Collapse
Affiliation(s)
- Naihua N Gong
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hang Ngoc Bao Luong
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - An H Dang
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin Mainwaring
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Shields
- Department of Urology and Institute of Neuropathology, Medical Center-University of Freiburg, 79106 Freiburg, Germany; Epigenetics Institute and Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karl Schmeckpeper
- Department of Computer Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roberto Bonasio
- Epigenetics Institute and Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew S Kayser
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Active forgetting requires Sickie function in a dedicated dopamine circuit in Drosophila. Proc Natl Acad Sci U S A 2022; 119:e2204229119. [PMID: 36095217 PMCID: PMC9499536 DOI: 10.1073/pnas.2204229119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Forgetting is an essential component of the brain's memory management system, providing a balance to memory formation processes by removing unused or unwanted memories, or by suppressing their expression. However, the molecular, cellular, and circuit mechanisms underlying forgetting are poorly understood. Here we show that the memory suppressor gene, sickie, functions in a single dopamine neuron (DAn) by supporting the process of active forgetting in Drosophila. RNAi knockdown (KD) of sickie impairs forgetting by reducing the Ca2+ influx and DA release from the DAn that promotes forgetting. Coimmunoprecipitation/mass spectrometry analyses identified cytoskeletal and presynaptic active zone (AZ) proteins as candidates that physically interact with Sickie, and a focused RNAi screen of the candidates showed that Bruchpilot (Brp)-a presynaptic AZ protein that regulates calcium channel clustering and neurotransmitter release-impairs active forgetting like sickie KD. In addition, overexpression of brp rescued the impaired forgetting of sickie KD, providing evidence that they function in the same process. Moreover, we show that sickie KD in the DAn reduces the abundance and size of AZ markers but increases their number, suggesting that Sickie controls DAn activity for forgetting by modulating the presynaptic AZ structure. Our results identify a molecular and circuit mechanism for normal levels of active forgetting and reveal a surprising role of Sickie in maintaining presynaptic AZ structure for neurotransmitter release.
Collapse
|
36
|
Kaźmierczak M, Nicola SM. The Arousal-motor Hypothesis of Dopamine Function: Evidence that Dopamine Facilitates Reward Seeking in Part by Maintaining Arousal. Neuroscience 2022; 499:64-103. [PMID: 35853563 PMCID: PMC9479757 DOI: 10.1016/j.neuroscience.2022.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/28/2022] [Accepted: 07/12/2022] [Indexed: 10/17/2022]
Abstract
Dopamine facilitates approach to reward via its actions on dopamine receptors in the nucleus accumbens. For example, blocking either D1 or D2 dopamine receptors in the accumbens reduces the proportion of reward-predictive cues to which rats respond with cued approach. Recent evidence indicates that accumbens dopamine also promotes wakefulness and arousal, but the relationship between dopamine's roles in arousal and reward seeking remains unexplored. Here, we show that the ability of systemic or intra-accumbens injections of the D1 antagonist SCH23390 to reduce cued approach to reward depends on the animal's state of arousal. Handling the animal, a manipulation known to increase arousal, was sufficient to reverse the behavioral effects of the antagonist. In addition, SCH23390 reduced spontaneous locomotion and increased time spent in sleep postures, both consistent with reduced arousal, but also increased time spent immobile in postures inconsistent with sleep. In contrast, the ability of the D2 antagonist haloperidol to reduce cued approach was not reversible by handling. Haloperidol reduced spontaneous locomotion but did not increase sleep postures, instead increasing immobility in non-sleep postures. We place these results in the context of the extensive literature on dopamine's contributions to behavior, and propose the arousal-motor hypothesis. This novel synthesis, which proposes that two main functions of dopamine are to promote arousal and facilitate motor behavior, accounts both for our findings and many previous behavioral observations that have led to disparate and conflicting conclusions.
Collapse
Affiliation(s)
- Marcin Kaźmierczak
- Departments of Neuroscience and Psychiatry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Forchheimer 111, Bronx, NY 10461, USA
| | - Saleem M Nicola
- Departments of Neuroscience and Psychiatry, Albert Einstein College of Medicine, 1300 Morris Park Ave, Forchheimer 111, Bronx, NY 10461, USA.
| |
Collapse
|
37
|
Ze LJ, Wang P, Peng YC, Jin L, Li GQ. Silencing tyrosine hydroxylase or dopa decarboxylase gene disrupts cuticle tanning during larva-pupa-adult transformation in Henosepilachna vigintioctopunctata. PEST MANAGEMENT SCIENCE 2022; 78:3880-3893. [PMID: 35470957 DOI: 10.1002/ps.6948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/30/2022] [Accepted: 04/26/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The 28-spotted potato ladybird, Henosepilachna vigintioctopunctata, is a notorious defoliator of many solanaceous and cucurbitaceous plants. Tyrosine hydroxylase (TH) and dopa decarboxylase (DDC) are responsible for cuticle tanning pathway in insects. RESULTS We identified HvTH and HvDDC in H. vigintioctopunctata, and found that high levels of them were accumulated just before or right after molting. Injection of dsHvTH or feeding 3-iodo-tyrosine (3-IT) at the third instar larval stage repressed tanning of the larval cuticle, reduced larval feeding, inhibited larval growth, and consequently caused 100% of larval mortality. Knockdown of HvDDC at the third instar larval stage hardly affected the coloration of larval head, and partially inhibited pigmentation of larval bodies and around 80% of the HvDDC RNAi larvae developed into albino pupae and adults. Moreover, depletion of HvTH or HvDDC at the fourth instar larval stage resulted in albino pupae and adults. The HvTH or HvDDC hypomorph adults fully or partially failed to remove the larval/pupal exuviae, possessed pale and abnormal wings, and poorly tanned heads and bodies, and eventually, struggled for several days without feeding on leaves before death. CONCLUSION These results show that TH and DDC play key roles in larval and adult cuticle tanning and development in H. vigintioctopunctata. Also, these findings suggest that dopa- and dopamine-originated pigments are essential for larval and adult feeding behavior and the molting process during emergence. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Long-Ji Ze
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Pei Wang
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Ying-Chuan Peng
- Institute of Entomology, Jiangxi Agricultural University, Nanchang, China
| | - Lin Jin
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Guo-Qing Li
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
38
|
Satterfield LK, De J, Wu M, Qiu T, Joiner WJ. Inputs to the Sleep Homeostat Originate Outside the Brain. J Neurosci 2022; 42:5695-5704. [PMID: 35680412 PMCID: PMC9302467 DOI: 10.1523/jneurosci.2113-21.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 01/22/2023] Open
Abstract
The need to sleep is sensed and discharged in a poorly understood process that is homeostatically controlled over time. In flies, different contributions to this process have been attributed to peripheral ppk and central brain neurons, with the former serving as hypothetical inputs to the sleep homeostat and the latter reportedly serving as the homeostat itself. Here we re-evaluate these distinctions in light of new findings using female flies. First, activating neurons targeted by published ppk and brain drivers elicits similar phenotypes, namely, sleep deprivation followed by rebound sleep. Second, inhibiting activity or synaptic output with one type of driver suppresses sleep homeostasis induced using the other type of driver. Third, drivers previously used to implicate central neurons in sleep homeostasis unexpectedly also label ppk neurons. Fourth, activating only this subset of colabeled neurons is sufficient to elicit sleep homeostasis. Thus, many published contributions of central neurons to sleep homeostasis can be explained by previously unrecognized expression of brain drivers in peripheral ppk neurons, most likely those in the legs, which promote walking. Last, we show that activation of certain non-ppk neurons can also induce sleep homeostasis. Notably, axons of these as well as ppk neurons terminate in the same ventral brain region, suggesting that a previously undefined neural circuit element of a sleep homeostat may lie nearby.SIGNIFICANCE STATEMENT The biological needs that sleep fulfills are unknown, but they are reflected by the ability of an animal to compensate for prior sleep loss in a process called sleep homeostasis. Researchers have searched for the neural circuitry that comprises the sleep homeostat so that the information it conveys can shed light on the nature of sleep need. Here we demonstrate that neurons originating outside of the brain are responsible for phenotypes previously attributed to the proposed central brain sleep homeostat in flies. Our results support a revised neural circuit model for sensing and discharging sleep need in which peripheral inputs connect to a sleep homeostat through previously unrecognized neural circuit elements in the ventral brain.
Collapse
Affiliation(s)
- Lawrence K Satterfield
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California 92093
| | - Joydeep De
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - Meilin Wu
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - Tianhao Qiu
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
| | - William J Joiner
- Department of Pharmacology, University of California, San Diego, La Jolla, California 92093
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California 92093
- Center for Circadian Biology, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
39
|
Liu Z, Jiang L, Li C, Li C, Yang J, Yu J, Mao R, Rao Y. LKB1 Is Physiologically Required for Sleep from Drosophila melanogaster to the Mus musculus. Genetics 2022; 221:6586797. [PMID: 35579349 DOI: 10.1093/genetics/iyac082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/10/2022] [Indexed: 11/14/2022] Open
Abstract
Liver Kinase B1 (LKB1) is known as a master kinase for 14 kinases related to the adenosine monophosphate (AMP)-activated protein kinase (AMPK). Two of them salt inducible kinase 3 (SIK3) and AMPKα have previously been implicated in sleep regulation. We generated loss-of-function (LOF) mutants for Lkb1 in both Drosophila and mice. Sleep, but not circadian rhythms, was reduced in Lkb1-mutant flies and in flies with neuronal deletion of Lkb1. Genetic interactions between Lkb1 and Threonine to Alanine mutation at residue 184 of AMPK in Drosophila sleep or those between Lkb1 and Threonine to Glutamic Acid mutation at residue 196 of SIK3 in Drosophila viability have been observed. Sleep was reduced in mice after virally mediated reduction of Lkb1 in the brain. Electroencephalography (EEG) analysis showed that non-rapid eye movement (NREM) sleep and sleep need were both reduced in Lkb1-mutant mice. These results indicate that LKB1 plays a physiological role in sleep regulation conserved from flies to mice.
Collapse
Affiliation(s)
- Ziyi Liu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Lifen Jiang
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, Guangdong, China
| | - Chaoyi Li
- Shenzhen Bay Laboratory, Institute of Molecular Physiology, Shenzhen, Guangdong, China
| | - Chengang Li
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Jingqun Yang
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Jianjun Yu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Renbo Mao
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Yi Rao
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Chemistry and Molecular Engineering, School of Pharmaceutical Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing, China
- Capital Medical University, Beijing, China
- Changping Laboratory, Beijing, China
| |
Collapse
|
40
|
Schmalz F, El Jundi B, Rössler W, Strube-Bloss M. Categorizing Visual Information in Subpopulations of Honeybee Mushroom Body Output Neurons. Front Physiol 2022; 13:866807. [PMID: 35574496 PMCID: PMC9092450 DOI: 10.3389/fphys.2022.866807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022] Open
Abstract
Multisensory integration plays a central role in perception, as all behaviors usually require the input of different sensory signals. For instance, for a foraging honeybee the association of a food source includes the combination of olfactory and visual cues to be categorized as a flower. Moreover, homing after successful foraging using celestial cues and the panoramic scenery may be dominated by visual cues. Hence, dependent on the context, one modality might be leading and influence the processing of other modalities. To unravel the complex neural mechanisms behind this process we studied honeybee mushroom body output neurons (MBON). MBONs represent the first processing level after olfactory-visual convergence in the honeybee brain. This was physiologically confirmed in our previous study by characterizing a subpopulation of multisensory MBONs. These neurons categorize incoming sensory inputs into olfactory, visual, and olfactory-visual information. However, in addition to multisensory units a prominent population of MBONs was sensitive to visual cues only. Therefore, we asked which visual features might be represented at this high-order integration level. Using extracellular, multi-unit recordings in combination with visual and olfactory stimulation, we separated MBONs with multisensory responses from purely visually driven MBONs. Further analysis revealed, for the first time, that visually driven MBONs of both groups encode detailed aspects within this individual modality, such as light intensity and light identity. Moreover, we show that these features are separated by different MBON subpopulations, for example by extracting information about brightness and wavelength. Most interestingly, the latter MBON population was tuned to separate UV-light from other light stimuli, which were only poorly differentiated from each other. A third MBON subpopulation was neither tuned to brightness nor to wavelength and encoded the general presence of light. Taken together, our results support the view that the mushroom body, a high-order sensory integration, learning and memory center in the insect brain, categorizes sensory information by separating different behaviorally relevant aspects of the multisensory scenery and that these categories are channeled into distinct MBON subpopulations.
Collapse
Affiliation(s)
- Fabian Schmalz
- Behavioral Physiology and Sociobiology (Zoology II), Biozentrum, University of Würzburg, Würzburg, Germany
| | - Basil El Jundi
- Behavioral Physiology and Sociobiology (Zoology II), Biozentrum, University of Würzburg, Würzburg, Germany
| | - Wolfgang Rössler
- Behavioral Physiology and Sociobiology (Zoology II), Biozentrum, University of Würzburg, Würzburg, Germany
| | - Martin Strube-Bloss
- Department of Biological Cybernetics and Theoretical Biology, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
41
|
Burdina EV, Gruntenko NE. Physiological Aspects of Wolbachia pipientis–Drosophila melanogaster Relationship. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Rahul, Siddique YH. Drosophila: A Model to Study the Pathogenesis of Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:259-277. [PMID: 35040399 DOI: 10.2174/1871527320666210809120621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/15/2021] [Accepted: 06/13/2021] [Indexed: 12/12/2022]
Abstract
Human Central Nervous System (CNS) is the complex part of the human body, which regulates multiple cellular and molecular events taking place simultaneously. Parkinsons Disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease (AD). The pathological hallmarks of PD are loss of dopaminergic neurons in the substantianigra (SN) pars compacta (SNpc) and accumulation of misfolded α-synuclein, in intra-cytoplasmic inclusions called Lewy bodies (LBs). So far, there is no cure for PD, due to the complexities of molecular mechanisms and events taking place during the pathogenesis of PD. Drosophila melanogaster is an appropriate model organism to unravel the pathogenicity not only behind PD but also other NDs. In this context as numerous biological functions are preserved between Drosophila and humans. Apart from sharing 75% of human disease-causing genes homolog in Drosophila, behavioral responses like memory-based tests, negative geotaxis, courtship and mating are also well studied. The genetic, as well as environmental factors, can be studied in Drosophila to understand the geneenvironment interactions behind the disease condition. Through genetic manipulation, mutant flies can be generated harboring human orthologs, which can prove to be an excellent model to understand the effect of the mutant protein on the pathogenicity of NDs.
Collapse
Affiliation(s)
- Rahul
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh,India
| | - Yasir Hasan Siddique
- Drosophila Transgenic Laboratory, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh,India
| |
Collapse
|
43
|
Lei Z, Henderson K, Keleman K. A neural circuit linking learning and sleep in Drosophila long-term memory. Nat Commun 2022; 13:609. [PMID: 35105888 PMCID: PMC8807839 DOI: 10.1038/s41467-022-28256-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 01/07/2022] [Indexed: 12/02/2022] Open
Abstract
Animals retain some but not all experiences in long-term memory (LTM). Sleep supports LTM retention across animal species. It is well established that learning experiences enhance post-learning sleep. However, the underlying mechanisms of how learning mediates sleep for memory retention are not clear. Drosophila males display increased amounts of sleep after courtship learning. Courtship learning depends on Mushroom Body (MB) neurons, and post-learning sleep is mediated by the sleep-promoting ventral Fan-Shaped Body neurons (vFBs). We show that post-learning sleep is regulated by two opposing output neurons (MBONs) from the MB, which encode a measure of learning. Excitatory MBONs-γ2α'1 becomes increasingly active upon increasing time of learning, whereas inhibitory MBONs-β'2mp is activated only by a short learning experience. These MB outputs are integrated by SFS neurons, which excite vFBs to promote sleep after prolonged but not short training. This circuit may ensure that only longer or more intense learning experiences induce sleep and are thereby consolidated into LTM.
Collapse
Affiliation(s)
- Zhengchang Lei
- Janelia Research Campus, HHMI, 19700 Helix Dr, Ashburn, VA, 20147, USA
| | - Kristin Henderson
- Janelia Research Campus, HHMI, 19700 Helix Dr, Ashburn, VA, 20147, USA
| | - Krystyna Keleman
- Janelia Research Campus, HHMI, 19700 Helix Dr, Ashburn, VA, 20147, USA.
| |
Collapse
|
44
|
Karam CS, Williams BL, Jones SK, Javitch JA. The Role of the Dopamine Transporter in the Effects of Amphetamine on Sleep and Sleep Architecture in Drosophila. Neurochem Res 2022; 47:177-189. [PMID: 33630236 PMCID: PMC8384956 DOI: 10.1007/s11064-021-03275-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 12/26/2022]
Abstract
The dopamine transporter (DAT) mediates the inactivation of released dopamine (DA) through its reuptake, and thereby plays an important homeostatic role in dopaminergic neurotransmission. Amphetamines exert their stimulant effects by targeting DAT and inducing the reverse transport of DA, leading to a dramatic increase of extracellular DA. Animal models have proven critical to investigating the molecular and cellular mechanisms underlying transporter function and its modulation by psychostimulants such as amphetamine. Here we establish a behavioral model for amphetamine action using adult Drosophila melanogaster. We use it to characterize the effects of amphetamine on sleep and sleep architecture. Our data show that amphetamine induces hyperactivity and disrupts sleep in a DA-dependent manner. Flies that do not express a functional DAT (dDAT null mutants) have been shown to be hyperactive and to exhibit significantly reduced sleep at baseline. Our data show that, in contrast to its action in control flies, amphetamine decreases the locomotor activity of dDAT null mutants and restores their sleep by modulating distinct aspects of sleep structure. To begin to explore the circuitry involved in the actions of amphetamine on sleep, we also describe the localization of dDAT throughout the fly brain, particularly in neuropils known to regulate sleep. Together, our data establish Drosophila as a robust model for studying the regulatory mechanisms that govern DAT function and psychostimulant action.
Collapse
Affiliation(s)
- Caline S Karam
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Brenna L Williams
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Sandra K Jones
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Jonathan A Javitch
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA.
- Department of Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, 1051 Riverside Dr, Unit 19, New York, NY, 10032, USA.
| |
Collapse
|
45
|
Tomita J, Ban G, Kato YS, Kume K. Protocerebral Bridge Neurons That Regulate Sleep in Drosophila melanogaster. Front Neurosci 2021; 15:647117. [PMID: 34720844 PMCID: PMC8554056 DOI: 10.3389/fnins.2021.647117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
The central complex is one of the major brain regions that control sleep in Drosophila. However, the circuitry details of sleep regulation have not been elucidated yet. Here, we show a novel sleep-regulating neuronal circuit in the protocerebral bridge (PB) of the central complex. Activation of the PB interneurons labeled by the R59E08-Gal4 and the PB columnar neurons with R52B10-Gal4 promoted sleep and wakefulness, respectively. A targeted GFP reconstitution across synaptic partners (t-GRASP) analysis demonstrated synaptic contact between these two groups of sleep-regulating PB neurons. Furthermore, we found that activation of a pair of dopaminergic (DA) neurons projecting to the PB (T1 DA neurons) decreased sleep. The wake-promoting T1 DA neurons and the sleep-promoting PB interneurons formed close associations. Dopamine 2-like receptor (Dop2R) knockdown in the sleep-promoting PB interneurons increased sleep. These results indicated that the neuronal circuit in the PB, regulated by dopamine signaling, mediates sleep-wakefulness.
Collapse
Affiliation(s)
- Jun Tomita
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Gosuke Ban
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Yoshiaki S Kato
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kazuhiko Kume
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
46
|
Tian X. Enhancing mask activity in dopaminergic neurons extends lifespan in flies. Aging Cell 2021; 20:e13493. [PMID: 34626525 PMCID: PMC8590106 DOI: 10.1111/acel.13493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/05/2021] [Accepted: 09/19/2021] [Indexed: 12/11/2022] Open
Abstract
Dopaminergic neurons (DANs) are essential modulators for brain functions involving memory formation, reward processing, and decision‐making. Here I demonstrate a novel and important function of the DANs in regulating aging and longevity. Overexpressing the putative scaffolding protein Mask in two small groups of DANs in flies can significantly extend the lifespan in flies and sustain adult locomotor and fecundity at old ages. This Mask‐induced beneficial effect requires dopaminergic transmission but cannot be recapitulated by elevating dopamine production alone in the DANs. Independent activation of Gαs in the same two groups of DANs via the drug‐inducible DREADD system also extends fly lifespan, further indicating the connection of specific DANs to aging control. The Mask‐induced lifespan extension appears to depend on the function of Mask to regulate microtubule (MT) stability. A structure–function analysis demonstrated that the ankyrin repeats domain in the Mask protein is both necessary for regulating MT stability (when expressed in muscles and motor neurons) and sufficient to prolong longevity (when expressed in the two groups of DANs). Furthermore, DAN‐specific overexpression of Unc‐104 or knockdown of p150Glued, two independent interventions previously shown to impact MT dynamics, also extends lifespan in flies. Together, these data demonstrated a novel DANs‐dependent mechanism that, upon the tuning of their MT dynamics, modulates systemic aging and longevity in flies.
Collapse
Affiliation(s)
- Xiaolin Tian
- Neuroscience Center of Excellence Department of Cell Biology and Anatomy Louisiana State University Health Sciences Center New Orleans Louisiana USA
| |
Collapse
|
47
|
Jeong J, Lee J, Kim JH, Lim C. Metabolic flux from the Krebs cycle to glutamate transmission tunes a neural brake on seizure onset. PLoS Genet 2021; 17:e1009871. [PMID: 34714823 PMCID: PMC8555787 DOI: 10.1371/journal.pgen.1009871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 10/11/2021] [Indexed: 01/18/2023] Open
Abstract
Kohlschütter-Tönz syndrome (KTS) manifests as neurological dysfunctions, including early-onset seizures. Mutations in the citrate transporter SLC13A5 are associated with KTS, yet their underlying mechanisms remain elusive. Here, we report that a Drosophila SLC13A5 homolog, I'm not dead yet (Indy), constitutes a neurometabolic pathway that suppresses seizure. Loss of Indy function in glutamatergic neurons caused "bang-induced" seizure-like behaviors. In fact, glutamate biosynthesis from the citric acid cycle was limiting in Indy mutants for seizure-suppressing glutamate transmission. Oral administration of the rate-limiting α-ketoglutarate in the metabolic pathway rescued low glutamate levels in Indy mutants and ameliorated their seizure-like behaviors. This metabolic control of the seizure susceptibility was mapped to a pair of glutamatergic neurons, reversible by optogenetic controls of their activity, and further relayed onto fan-shaped body neurons via the ionotropic glutamate receptors. Accordingly, our findings reveal a micro-circuit that links neural metabolism to seizure, providing important clues to KTS-associated neurodevelopmental deficits.
Collapse
Affiliation(s)
- Jiwon Jeong
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Jongbin Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Ji-hyung Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Chunghun Lim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- * E-mail:
| |
Collapse
|
48
|
Hulse BK, Haberkern H, Franconville R, Turner-Evans D, Takemura SY, Wolff T, Noorman M, Dreher M, Dan C, Parekh R, Hermundstad AM, Rubin GM, Jayaraman V. A connectome of the Drosophila central complex reveals network motifs suitable for flexible navigation and context-dependent action selection. eLife 2021; 10:e66039. [PMID: 34696823 PMCID: PMC9477501 DOI: 10.7554/elife.66039] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
Flexible behaviors over long timescales are thought to engage recurrent neural networks in deep brain regions, which are experimentally challenging to study. In insects, recurrent circuit dynamics in a brain region called the central complex (CX) enable directed locomotion, sleep, and context- and experience-dependent spatial navigation. We describe the first complete electron microscopy-based connectome of the Drosophila CX, including all its neurons and circuits at synaptic resolution. We identified new CX neuron types, novel sensory and motor pathways, and network motifs that likely enable the CX to extract the fly's head direction, maintain it with attractor dynamics, and combine it with other sensorimotor information to perform vector-based navigational computations. We also identified numerous pathways that may facilitate the selection of CX-driven behavioral patterns by context and internal state. The CX connectome provides a comprehensive blueprint necessary for a detailed understanding of network dynamics underlying sleep, flexible navigation, and state-dependent action selection.
Collapse
Affiliation(s)
- Brad K Hulse
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Hannah Haberkern
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Romain Franconville
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Daniel Turner-Evans
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Shin-ya Takemura
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Tanya Wolff
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Marcella Noorman
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Marisa Dreher
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Chuntao Dan
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Ruchi Parekh
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Ann M Hermundstad
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Gerald M Rubin
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Vivek Jayaraman
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| |
Collapse
|
49
|
A pair of dopamine neurons mediate chronic stress signals to induce learning deficit in Drosophila melanogaster. Proc Natl Acad Sci U S A 2021; 118:2023674118. [PMID: 34654742 DOI: 10.1073/pnas.2023674118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic stress could induce severe cognitive impairments. Despite extensive investigations in mammalian models, the underlying mechanisms remain obscure. Here, we show that chronic stress could induce dramatic learning and memory deficits in Drosophila melanogaster The chronic stress-induced learning deficit (CSLD) is long lasting and associated with other depression-like behaviors. We demonstrated that excessive dopaminergic activity provokes susceptibility to CSLD. Remarkably, a pair of PPL1-γ1pedc dopaminergic neurons that project to the mushroom body (MB) γ1pedc compartment play a key role in regulating susceptibility to CSLD so that stress-induced PPL1-γ1pedc hyperactivity facilitates the development of CSLD. Consistently, the mushroom body output neurons (MBON) of the γ1pedc compartment, MBON-γ1pedc>α/β neurons, are important for modulating susceptibility to CSLD. Imaging studies showed that dopaminergic activity is necessary to provoke the development of chronic stress-induced maladaptations in the MB network. Together, our data support that PPL1-γ1pedc mediates chronic stress signals to drive allostatic maladaptations in the MB network that lead to CSLD.
Collapse
|
50
|
Driscoll M, Buchert SN, Coleman V, McLaughlin M, Nguyen A, Sitaraman D. Compartment specific regulation of sleep by mushroom body requires GABA and dopaminergic signaling. Sci Rep 2021; 11:20067. [PMID: 34625611 PMCID: PMC8501079 DOI: 10.1038/s41598-021-99531-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022] Open
Abstract
Sleep is a fundamental behavioral state important for survival and is universal in animals with sufficiently complex nervous systems. As a highly conserved neurobehavioral state, sleep has been described in species ranging from jellyfish to humans. Biogenic amines like dopamine, serotonin and norepinephrine have been shown to be critical for sleep regulation across species but the precise circuit mechanisms underlying how amines control persistence of sleep, arousal and wakefulness remain unclear. The fruit fly, Drosophila melanogaster, provides a powerful model system for the study of sleep and circuit mechanisms underlying state transitions and persistence of states to meet the organisms motivational and cognitive needs. In Drosophila, two neuropils in the central brain, the mushroom body (MB) and the central complex (CX) have been shown to influence sleep homeostasis and receive aminergic neuromodulator input critical to sleep–wake switch. Dopamine neurons (DANs) are prevalent neuromodulator inputs to the MB but the mechanisms by which they interact with and regulate sleep- and wake-promoting neurons within MB are unknown. Here we investigate the role of subsets of PAM-DANs that signal wakefulness and project to wake-promoting compartments of the MB. We find that PAM-DANs are GABA responsive and require GABAA-Rdl receptor in regulating sleep. In mapping the pathways downstream of PAM neurons innervating γ5 and β′2 MB compartments we find that wakefulness is regulated by both DopR1 and DopR2 receptors in downstream Kenyon cells (KCs) and mushroom body output neurons (MBONs). Taken together, we have identified and characterized a dopamine modulated sleep microcircuit within the mushroom body that has previously been shown to convey information about positive and negative valence critical for memory formation. These studies will pave way for understanding how flies balance sleep, wakefulness and arousal.
Collapse
Affiliation(s)
- Margaret Driscoll
- Department of Psychological Sciences, College of Arts and Sciences, University of San Diego, 5998 Alcala Park, San Diego, CA, 92110, USA
| | - Steven N Buchert
- Department of Psychology, College of Science, California State University- East Bay, 25800 Carlos Bee Blvd, Hayward, CA, 94542, USA
| | - Victoria Coleman
- Department of Psychological Sciences, College of Arts and Sciences, University of San Diego, 5998 Alcala Park, San Diego, CA, 92110, USA
| | - Morgan McLaughlin
- Department of Psychological Sciences, College of Arts and Sciences, University of San Diego, 5998 Alcala Park, San Diego, CA, 92110, USA
| | - Amanda Nguyen
- Department of Psychological Sciences, College of Arts and Sciences, University of San Diego, 5998 Alcala Park, San Diego, CA, 92110, USA
| | - Divya Sitaraman
- Department of Psychological Sciences, College of Arts and Sciences, University of San Diego, 5998 Alcala Park, San Diego, CA, 92110, USA. .,Department of Psychology, College of Science, California State University- East Bay, 25800 Carlos Bee Blvd, Hayward, CA, 94542, USA.
| |
Collapse
|