1
|
Helfrich-Förster C. The Never Given 2022 Pittendrigh/Aschoff Lecture: The Clock Network in the Brain-Insights From Insects. J Biol Rhythms 2025; 40:120-142. [PMID: 39529231 PMCID: PMC11915775 DOI: 10.1177/07487304241290861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
My journey into chronobiology began in 1977 with lectures and internships with Wolfgang Engelmann and Hans Erkert at the University of Tübingen in Germany. At that time, the only known animal clock gene was Period, and the location and organization of the master circadian clock in the brain was completely unknown for the model insect Drosophila melanogaster. I was thus privileged to witness and participate in the research that led us from discovering the first clock gene to identifying the clock network in the fly brain and the putative pathways linking it to behavior and physiology. This article highlights my role in these developments and also shows how the successful use of D. melanogaster for studies of circadian rhythms has contributed to the understanding of clock networks in other animals. I also report on my experiences in the German scientific system and hope that my story will be of interest to some of you.
Collapse
Affiliation(s)
- Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
2
|
Damulewicz M, Mazzotta GM. A one-day journey to the suburbs: circadian clock in the Drosophila visual system. FEBS J 2025; 292:727-739. [PMID: 39484992 PMCID: PMC11839939 DOI: 10.1111/febs.17317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/17/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
Living organisms, which are constantly exposed to cyclical variations in their environment, need a high degree of plasticity in their visual system to respond to daily and seasonal fluctuations in lighting conditions. In Drosophila melanogaster, the visual system is a complex tissue comprising different photoreception structures that exhibit daily rhythms in gene expression, cell morphology, and synaptic plasticity, regulated by both the central and peripheral clocks. In this review, we briefly summarize the structure of the circadian clock and the visual system in Drosophila and comprehensively describe circadian oscillations in visual structures, from molecules to behaviors, which are fundamental for the fine-tuning of visual sensitivity. We also compare some features of the rhythmicity in the visual system with that of the central pacemaker and hypothesize about the differences in the regulatory signals and mechanisms that control these two clocks.
Collapse
Affiliation(s)
- Milena Damulewicz
- Department of Cell Biology and ImagingJagiellonian UniversityKrakówPoland
| | | |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Iiams SE, Wan G, Zhang J, Lugena AB, Zhang Y, Hayden AN, Merlin C. Loss of functional cryptochrome 1 reduces robustness of 24-hour behavioral rhythms in monarch butterflies. iScience 2024; 27:108980. [PMID: 38333697 PMCID: PMC10850777 DOI: 10.1016/j.isci.2024.108980] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 12/05/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
Light is one of the strongest cues for entrainment of circadian clocks. While some insect species rely only on visual input, others like Drosophila melanogaster use both the visual system and the deep-brain blue-light photoreceptor cryptochrome for entraining circadian rhythms. Here, we used the monarch butterfly Danaus plexippus (dp), which possesses a light-sensitive cryptochrome 1 (dpCry1), to test the conservation of mechanisms of clock entrainment. We showed that loss of functional dpCry1 reduced the amplitude and altered the phase of adult eclosion rhythms, and disrupted brain molecular circadian rhythms. Robust rhythms could be restored by entrainment to temperature cycles, indicating a likely functional core circadian clock in dpCry1 mutants. We also showed that rhythmic flight activity was less robust in dpCry1 mutants, and that visual impairment in dpNinaB1 mutants impacted flight suppression at night. Our data suggest that dpCRY1 is a major photoreceptor for light-entrainment of the monarch circadian clock.
Collapse
Affiliation(s)
- Samantha E. Iiams
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
- Genetics Interdisciplinary Program, Texas A&M University, College Station, TX 77843, USA
| | - Guijun Wan
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
- Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiwei Zhang
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
| | - Aldrin B. Lugena
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
| | - Ying Zhang
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
| | - Ashley N. Hayden
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
| | - Christine Merlin
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77843, USA
- Genetics Interdisciplinary Program, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
6
|
Xu S, Momin M, Ahmed S, Hossain A, Veeramuthu L, Pandiyan A, Kuo CC, Zhou T. Illuminating the Brain: Advances and Perspectives in Optoelectronics for Neural Activity Monitoring and Modulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303267. [PMID: 37726261 DOI: 10.1002/adma.202303267] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/30/2023] [Indexed: 09/21/2023]
Abstract
Optogenetic modulation of brain neural activity that combines optical and electrical modes in a unitary neural system has recently gained robust momentum. Controlling illumination spatial coverage, designing light-activated modulators, and developing wireless light delivery and data transmission are crucial for maximizing the use of optical neuromodulation. To this end, biocompatible electrodes with enhanced optoelectrical performance, device integration for multiplexed addressing, wireless transmission, and multimodal operation in soft systems have been developed. This review provides an outlook for uniformly illuminating large brain areas while spatiotemporally imaging the neural responses upon optoelectrical stimulation with little artifacts. Representative concepts and important breakthroughs, such as head-mounted illumination, multiple implanted optical fibers, and micro-light-delivery devices, are discussed. Examples of techniques that incorporate electrophysiological monitoring and optoelectrical stimulation are presented. Challenges and perspectives are posed for further research efforts toward high-density optoelectrical neural interface modulation, with the potential for nonpharmacological neurological disease treatments and wireless optoelectrical stimulation.
Collapse
Affiliation(s)
- Shumao Xu
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Marzia Momin
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Salahuddin Ahmed
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Arafat Hossain
- Department of Electrical Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| | - Loganathan Veeramuthu
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Archana Pandiyan
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Chi-Ching Kuo
- Department of Molecular Science and Engineering, National Taipei University of Technology, Taipei, 10608, Republic of China
| | - Tao Zhou
- Department of Engineering Science and Mechanics, Center for Neural Engineering, The Pennsylvania State University, Pennsylvania, 16802, USA
| |
Collapse
|
7
|
Au DD, Liu JC, Park SJ, Nguyen TH, Dimalanta M, Foden AJ, Holmes TC. Drosophila photoreceptor systems converge in arousal neurons and confer light responsive robustness. Front Neurosci 2023; 17:1160353. [PMID: 37274190 PMCID: PMC10235467 DOI: 10.3389/fnins.2023.1160353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023] Open
Abstract
Lateral ventral neurons (LNvs) in the fly circadian neural circuit mediate behaviors other than clock resetting, including light-activated acute arousal. Converging sensory inputs often confer functional redundancy. The LNvs have three distinct light input pathways: (1) cell autonomously expressed cryptochrome (CRY), (2) rhodopsin 7 (Rh7), and (3) synaptic inputs from the eyes and other external photoreceptors that express opsins and CRY. We explored the relative photoelectrical and behavioral input contributions of these three photoreceptor systems to determine their functional impact in flies. Patch-clamp electrophysiology measuring light evoked firing frequency (FF) was performed on large LNvs (l-LNvs) in response to UV (365 nm), violet (405 nm), blue (450 nm), or red (635 nm) LED light stimulation, testing controls versus mutants that lack photoreceptor inputs gl60j, cry-null, rh7-null, and double mutant gl60j-cry-null flies. For UV, violet, and blue short wavelength light inputs, all photoreceptor mutants show significantly attenuated action potential FF responses measured in the l-LNv. In contrast, red light FF responses are only significantly attenuated in double mutant gl60j-cry-null flies. We used a light-pulse arousal assay to compare behavioral responses to UV, violet, blue and red light of control and light input mutants, measuring the awakening arousal response of flies during subjective nighttime at two different intensities to capture potential threshold differences (10 and 400 μW/cm2). The light arousal behavioral results are similar to the electrophysiological results, showing significant attenuation of behavioral light responses for mutants compared to control. These results show that the different LNv convergent photoreceptor systems are integrated and together confer functional redundancy for light evoked behavioral arousal.
Collapse
Affiliation(s)
- David D. Au
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Jenny C. Liu
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Soo Jee Park
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Thanh H. Nguyen
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Mia Dimalanta
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Alexander J. Foden
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Todd C. Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
8
|
Hirata K, Shiga S. Bolwig Organ and Its Role in the Photoperiodic Response of Sarcophaga similis Larvae. INSECTS 2023; 14:115. [PMID: 36835683 PMCID: PMC9959995 DOI: 10.3390/insects14020115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Flesh-fly Sarcophaga similis larvae exhibit a photoperiodic response, in which short days induce pupal diapause for seasonal adaptation. Although the spectral sensitivity of photoperiodic photoreception is known, the photoreceptor organ remains unclear. We morphologically identified the Bolwig organ, a larval-photoreceptor identified in several other fly species, and examined the effects of its removal on the photoperiodic response in S. similis. Backfill-staining and embryonic-lethal-abnormal-vision (ELAV) immunohistochemical-staining identified ~34 and 38 cells, respectively, in a spherical body at the ocular depression of the cephalopharyngeal skeleton, suggesting that the spherical body is the Bolwig organ in S. similis. Forward-fill and immunohistochemistry revealed that Bolwig-organ neurons terminate in the vicinity of the dendritic fibres of pigment-dispersing factor-immunoreactive and potential circadian-clock neurons in the brain. After surgical removal of the Bolwig-organ regions, diapause incidence was not significantly different between short and long days, and was similar to that in the insects with an intact organ, under constant darkness. However, diapause incidence was not significantly different between the control and Bolwig-organ-removed insects for each photoperiod. These results suggest that the Bolwig organ contributes partially to photoperiodic photoreception, and that other photoreceptors may also be involved.
Collapse
Affiliation(s)
- Kazuné Hirata
- Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka 560-0043, Osaka, Japan
- Center for Ecological Research, Kyoto University, Otsu 520-2133, Shiga, Japan
| | - Sakiko Shiga
- Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka 560-0043, Osaka, Japan
| |
Collapse
|
9
|
Jean‐Guillaume CB, Kumar JP. Development of the ocellar visual system in Drosophila melanogaster. FEBS J 2022; 289:7411-7427. [PMID: 35490409 PMCID: PMC9805374 DOI: 10.1111/febs.16468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/28/2022] [Accepted: 04/29/2022] [Indexed: 01/14/2023]
Abstract
The adult visual system of the fruit fly, Drosophila melanogaster, contains seven eyes-two compound eyes, a pair of Hofbauer-Buchner eyelets, and three ocelli. Each of these eye types has a specialized and essential role to play in visual and/or circadian behavior. As such, understanding how each is specified, patterned, and wired is of primary importance to vision biologists. Since the fruit fly is amenable to manipulation by an enormous array of genetic and molecular tools, its development is one of the best and most studied model systems. After more than a century of experimental investigations, our understanding of how each eye type is specified and patterned is grossly uneven. The compound eye has been the subject of several thousand studies; thus, our knowledge of its development is the deepest. By comparison, very little is known about the specification and patterning of the other two visual systems. In this Viewpoint article, we will describe what is known about the function and development of the Drosophila ocelli.
Collapse
|
10
|
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
|
11
|
Ikeda K, Kataoka M, Tanaka NK. Nonsynaptic Transmission Mediates Light Context-Dependent Odor Responses in Drosophila melanogaster. J Neurosci 2022; 42:8621-8628. [PMID: 36180227 PMCID: PMC9671575 DOI: 10.1523/jneurosci.1106-22.2022] [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: 06/06/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Recent connectome analyses of the entire synaptic circuit in the nervous system have provided tremendous insights into how neural processing occurs through the synaptic relay of neural information. Conversely, the extent to which ephaptic transmission which does not depend on the synapses contributes to the relay of neural information, especially beyond a distance between adjacent neurons and to neural processing remains unclear. We show that ephaptic transmission mediated by extracellular potential changes in female Drosophila melanogaster can reach >200 µm, equivalent to the depth of its brain. Furthermore, ephaptic transmission driven by retinal photoreceptor cells mediates light-evoked firing rate increases in olfactory sensory neurons. These results indicate that ephaptic transmission contributes to sensory responses that can change momentarily in a context-dependent manner.SIGNIFICANCE STATEMENT Although extracellular field potential activities are commonly observed in many nervous systems, this activity has been generally considered as a side effect of synchronized spiking of neurons. This study, however, shows that field potential changes in retinae evoked by a sensory stimulus can control the excitability of distant neurons in vivo and mediates multimodal sensory integration in Drosophila melanogaster As such ephaptic transmission is more effective at a short distance, the ephaptic transmission from the retinae may contribute significantly to firing rate changes in downstream neurons of the photoreceptor cells in the optic lobe.
Collapse
Affiliation(s)
- Kazuaki Ikeda
- Division of Biology, Department of Biological Sciences, School of Science, Hokkaido University, Sapporo, 060-0810, Japan
- Graduate School of Life Sciences, Hokkaido University, Sapporo, 060-0810, Japan
| | - Masaki Kataoka
- Division of Biology, Department of Biological Sciences, School of Science, Hokkaido University, Sapporo, 060-0810, Japan
- Graduate School of Life Sciences, Hokkaido University, Sapporo, 060-0810, Japan
| | - Nobuaki K Tanaka
- Division of Biology, Department of Biological Sciences, School of Science, Hokkaido University, Sapporo, 060-0810, Japan
- Graduate School of Life Sciences, Hokkaido University, Sapporo, 060-0810, Japan
- Faculty of Science, Hokkaido University, Sapporo, 060-0810, Japan
| |
Collapse
|
12
|
Shafer OT, Gutierrez GJ, Li K, Mildenhall A, Spira D, Marty J, Lazar AA, Fernandez MDLP. ---Connectomic analysis of the Drosophila lateral neuron clock cells reveals the synaptic basis of functional pacemaker classes. eLife 2022; 11:79139. [PMID: 35766361 PMCID: PMC9365390 DOI: 10.7554/elife.79139] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
The circadian clock orchestrates daily changes in physiology and behavior to ensure internal temporal order and optimal timing across the day. In animals, a central brain clock coordinates circadian rhythms throughout the body and is characterized by a remarkable robustness that depends on synaptic connections between constituent neurons. The clock neuron network of Drosophila, which shares network motifs with clock networks in the mammalian brain yet is built of many fewer neurons, offers a powerful model for understanding the network properties of circadian timekeeping. Here, we report an assessment of synaptic connectivity within a clock network, focusing on the critical lateral neuron (LN) clock neuron classes within the Janelia hemibrain dataset. Our results reveal that previously identified anatomical and functional subclasses of LNs represent distinct connectomic types. Moreover, we identify a small number of non-clock cell subtypes representing highly synaptically coupled nodes within the clock neuron network. This suggests that neurons lacking molecular timekeeping likely play integral roles within the circadian timekeeping network. To our knowledge, this represents the first comprehensive connectomic analysis of a circadian neuronal network. Most organisms on Earth possess an internal timekeeping system which ensures that bodily processes such as sleep, wakefulness or digestion take place at the right time. These precise daily rhythms are kept in check by a master clock in the brain. There, thousands of neurons – some of which carrying an internal ‘molecular clock’ – connect to each other through structures known as synapses. Exactly how the resulting network is organised to support circadian timekeeping remains unclear. To explore this question, Shafer, Gutierrez et al. focused on fruit flies, as recent efforts have systematically mapped every neuron and synaptic connection in the brain of this model organism. Analysing available data from the hemibrain connectome project at Janelia revealed that that the neurons with the most important timekeeping roles were in fact forming the fewest synapses within the network. In addition, neurons without internal molecular clocks mediated strong synaptic connections between those that did, suggesting that ‘clockless’ cells still play an integral role in circadian timekeeping. With this research, Shafer, Gutierrez et al. provide unexpected insights into the organisation of the master body clock. Better understanding the networks that underpin circadian rhythms will help to grasp how and why these are disrupted in obesity, depression and Alzheimer’s disease.
Collapse
Affiliation(s)
- Orie T Shafer
- Advanced Science Research Center, City University of New York, New York, United States
| | - Gabrielle J Gutierrez
- Center for Theoretical Neuroscience, Columbia University, New York City, United States
| | - Kimberly Li
- Department of Neuroscience and Behavior, Barnard College, New York, United States
| | - Amber Mildenhall
- Department of Neuroscience and Behavior, Barnard College, New York, United States
| | - Daphna Spira
- Center for Theoretical Neuroscience, Columbia University, New York City, United States
| | - Jonathan Marty
- Department of Electrical Engineering, Columbia University, New York, United States
| | - Aurel A Lazar
- Department of Electrical Engineering, Columbia University, New York, United States
| | | |
Collapse
|
13
|
Montell C. Drosophila sensory receptors-a set of molecular Swiss Army Knives. Genetics 2021; 217:1-34. [PMID: 33683373 DOI: 10.1093/genetics/iyaa011] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/17/2020] [Indexed: 01/01/2023] Open
Abstract
Genetic approaches in the fruit fly, Drosophila melanogaster, have led to a major triumph in the field of sensory biology-the discovery of multiple large families of sensory receptors and channels. Some of these families, such as transient receptor potential channels, are conserved from animals ranging from worms to humans, while others, such as "gustatory receptors," "olfactory receptors," and "ionotropic receptors," are restricted to invertebrates. Prior to the identification of sensory receptors in flies, it was widely assumed that these proteins function in just one modality such as vision, smell, taste, hearing, and somatosensation, which includes thermosensation, light, and noxious mechanical touch. By employing a vast combination of genetic, behavioral, electrophysiological, and other approaches in flies, a major concept to emerge is that many sensory receptors are multitaskers. The earliest example of this idea was the discovery that individual transient receptor potential channels function in multiple senses. It is now clear that multitasking is exhibited by other large receptor families including gustatory receptors, ionotropic receptors, epithelial Na+ channels (also referred to as Pickpockets), and even opsins, which were formerly thought to function exclusively as light sensors. Genetic characterizations of these Drosophila receptors and the neurons that express them also reveal the mechanisms through which flies can accurately differentiate between different stimuli even when they activate the same receptor, as well as mechanisms of adaptation, amplification, and sensory integration. The insights gleaned from studies in flies have been highly influential in directing investigations in many other animal models.
Collapse
Affiliation(s)
- Craig Montell
- Department of Molecular, Cellular, and Developmental Biology, The Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
14
|
Poe AR, Mace KD, Kayser MS. Getting into rhythm: developmental emergence of circadian clocks and behaviors. FEBS J 2021; 289:6576-6588. [PMID: 34375504 DOI: 10.1111/febs.16157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/30/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022]
Abstract
Circadian clocks keep time to coordinate diverse behaviors and physiological functions. While molecular circadian rhythms are evident during early development, most behavioral rhythms, such as sleep-wake, do not emerge until far later. Here, we examine the development of circadian clocks, outputs, and behaviors across phylogeny, with a particular focus on Drosophila. We explore potential mechanisms for how central clocks and circadian output loci establish communication, and discuss why from an evolutionary perspective sleep-wake and other behavioral rhythms emerge long after central clocks begin keeping time.
Collapse
Affiliation(s)
- Amy R Poe
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kyla D Mace
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew S Kayser
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
15
|
Dapergola E, Menegazzi P, Raabe T, Hovhanyan A. Light Stimuli and Circadian Clock Affect Neural Development in Drosophila melanogaster. Front Cell Dev Biol 2021; 9:595754. [PMID: 33763414 PMCID: PMC7982892 DOI: 10.3389/fcell.2021.595754] [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: 08/17/2020] [Accepted: 02/02/2021] [Indexed: 11/13/2022] Open
Abstract
Endogenous clocks enable organisms to adapt cellular processes, physiology, and behavior to daily variation in environmental conditions. Metabolic processes in cyanobacteria to humans are under the influence of the circadian clock, and dysregulation of the circadian clock causes metabolic disorders. In mouse and Drosophila, the circadian clock influences translation of factors involved in ribosome biogenesis and synchronizes protein synthesis. Notably, nutrition signals are mediated by the insulin receptor/target of rapamycin (InR/TOR) pathways to regulate cellular metabolism and growth. However, the role of the circadian clock in Drosophila brain development and the potential impact of clock impairment on neural circuit formation and function is less understood. Here we demonstrate that changes in light stimuli or disruption of the molecular circadian clock cause a defect in neural stem cell growth and proliferation. Moreover, we show that disturbed cell growth and proliferation are accompanied by reduced nucleolar size indicative of impaired ribosomal biogenesis. Further, we define that light and clock independently affect the InR/TOR growth regulatory pathway due to the effect on regulators of protein biosynthesis. Altogether, these data suggest that alterations in InR/TOR signaling induced by changes in light conditions or disruption of the molecular clock have an impact on growth and proliferation properties of neural stem cells in the developing Drosophila brain.
Collapse
Affiliation(s)
- Eleni Dapergola
- Institute of Medical Radiation and Cell Research, Biozentrum, University of Würzburg, Würzburg, Germany
| | - Pamela Menegazzi
- Neurobiology and Genetics, Theodor-Boveri Institute, Biozentrum, University of Würzburg, Würzburg, Germany
| | - Thomas Raabe
- Institute of Medical Radiation and Cell Research, Biozentrum, University of Würzburg, Würzburg, Germany
| | - Anna Hovhanyan
- Institute of Medical Radiation and Cell Research, Biozentrum, University of Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Schlichting M. Entrainment of the Drosophila clock by the visual system. Neurosci Insights 2020; 15:2633105520903708. [PMID: 35174330 PMCID: PMC8842342 DOI: 10.1177/2633105520903708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/08/2020] [Indexed: 12/27/2022] Open
Abstract
Circadian clocks evolved as an adaptation to the cyclic change of day and night. To precisely adapt to this environment, the endogenous period has to be adjusted every day to exactly 24 hours by a process called entrainment. Organisms can use several external cues, called zeitgebers, to adapt. These include changes in temperature, humidity, or light. The latter is the most powerful signal to synchronize the clock in animals. Research shows that a complex visual system and circadian photoreceptors work together to adjust animal physiology to the outside world. This review will focus on the importance of the visual system for clock synchronization in the fruit fly Drosophila melanogaster. It will cover behavioral and physiological evidence that supports the importance of the visual system in light entrainment.
Collapse
|
17
|
Yin J, Gibbs M, Long C, Rosenthal J, Kim HS, Kim A, Sheng C, Ding P, Javed U, Yuan Q. Transcriptional Regulation of Lipophorin Receptors Supports Neuronal Adaptation to Chronic Elevations of Activity. Cell Rep 2019; 25:1181-1192.e4. [PMID: 30380410 PMCID: PMC6294312 DOI: 10.1016/j.celrep.2018.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/20/2018] [Accepted: 10/03/2018] [Indexed: 12/18/2022] Open
Abstract
Activity-dependent modifications strongly influence neural development. However, molecular programs underlying their context and circuit-specific effects are not well understood. To study global transcriptional changes associated with chronic elevation of synaptic activity, we performed cell-type-specific transcriptome profiling of Drosophila ventral lateral neurons (LNvs) in the developing visual circuit and identified activity-modified transcripts that are enriched in neuron morphogenesis, circadian regulation, and lipid metabolism and trafficking. Using bioinformatics and genetic analyses, we validated activity-induced isoform-specific upregulation of Drosophila lipophorin receptors LpR1 and LpR2, the homologs of mammalian low-density lipoprotein receptor (LDLR) family proteins. Furthermore, our morphological and physiological studies uncovered critical functions of neuronal lipophorin receptors (LpRs) in maintaining the structural and functional integrities in neurons challenged by chronic elevations of activity. Together, our findings identify LpRs as molecular targets for activity-dependent transcriptional regulation and reveal the functional significance of cell-type-specific regulation of neuronal lipid uptake in experience-dependent plasticity and adaptive responses. Yin et al. highlight Drosophila lipophorin receptors (LpRs) as molecular targets for activity-dependent transcriptional regulation and reveal the functional significance of cell-type-specific regulation of neuronal lipid uptake in experience-dependent plasticity and adaptive responses.
Collapse
Affiliation(s)
- Jun Yin
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Mary Gibbs
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Caixia Long
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Justin Rosenthal
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Hyong S Kim
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Anna Kim
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Chengyu Sheng
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Peng Ding
- Neurobiology Department, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Uzma Javed
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Quan Yuan
- Dendrite Morphogenesis and Plasticity Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
18
|
Baik LS, Recinos Y, Chevez JA, Au DD, Holmes TC. Multiple Phototransduction Inputs Integrate to Mediate UV Light-evoked Avoidance/Attraction Behavior in Drosophila. J Biol Rhythms 2019; 34:391-400. [PMID: 31140349 DOI: 10.1177/0748730419847339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Short-wavelength light guides many behaviors that are crucial for an insect's survival. In Drosophila melanogaster, short-wavelength light induces both attraction and avoidance behaviors. How light cues evoke two opposite valences of behavioral responses remains unclear. Here, we comprehensively examine the effects of (1) light intensity, (2) timing of light (duration of exposure, circadian time of day), and (3) phototransduction mechanisms processing light information that determine avoidance versus attraction behavior assayed at high spatiotemporal resolution in Drosophila. External opsin-based photoreceptors signal for attraction behavior in response to low-intensity ultraviolet (UV) light. In contrast, the cell-autonomous neuronal photoreceptors, CRYPTOCHROME (CRY) and RHODOPSIN 7 (RH7), signal avoidance responses to high-intensity UV light. In addition to binary attraction versus avoidance behavioral responses to UV light, flies show distinct clock-dependent spatial preference within a light environment coded by different light input channels.
Collapse
Affiliation(s)
- Lisa Soyeon Baik
- Department of Physiology and Biophysics, School of Medicine, University of California at Irvine, Irvine, California
| | - Yocelyn Recinos
- Department of Physiology and Biophysics, School of Medicine, University of California at Irvine, Irvine, California
| | - Joshua A Chevez
- Department of Physiology and Biophysics, School of Medicine, University of California at Irvine, Irvine, California
| | - David D Au
- Department of Physiology and Biophysics, School of Medicine, University of California at Irvine, Irvine, California
| | - Todd C Holmes
- Department of Physiology and Biophysics, School of Medicine, University of California at Irvine, Irvine, California
| |
Collapse
|
19
|
Zhao J, Warman GR, Stanewsky R, Cheeseman JF. Development of the Molecular Circadian Clock and Its Light Sensitivity in Drosophila Melanogaster. J Biol Rhythms 2019; 34:272-282. [PMID: 30879378 DOI: 10.1177/0748730419836818] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The importance of the circadian clock for the control of behavior and physiology is well established but how and when it develops is not fully understood. Here the initial expression pattern of the key clock gene period was recorded in Drosophila from embryos in vivo, using transgenic luciferase reporters. PERIOD expression in the presumptive central-clock dorsal neurons started to oscillate in the embryo in constant darkness. In behavioral experiments, a single 12-h light pulse given during the embryonic stage synchronized adult activity rhythms, implying the early development of entrainment mechanisms. These findings suggest that the central clock is functional already during embryogenesis. In contrast to central brain expression, PERIOD in the peripheral cells or their precursors increased during the embryonic stage and peaked during the pupal stage without showing circadian oscillations. Its rhythmic expression only initiated in the adult. We conclude that cyclic expression of PERIOD in the central-clock neurons starts in the embryo, presumably in the dorsal neurons or their precursors. It is not until shortly after eclosion when cyclic and synchronized expression of PERIOD in peripheral tissues commences throughout the animal.
Collapse
Affiliation(s)
- Jia Zhao
- Department of Anaesthesiology, School of Medicine, University of Auckland, Auckland, 1142 New Zealand
| | - Guy Robert Warman
- Department of Anaesthesiology, School of Medicine, University of Auckland, Auckland, 1142 New Zealand
| | - Ralf Stanewsky
- Institute for Neuro- and Behavioral Biology, Westfälische Wilhelms University, 48149 Münster, Germany
| | - James Frederick Cheeseman
- Department of Anaesthesiology, School of Medicine, University of Auckland, Auckland, 1142 New Zealand
| |
Collapse
|
20
|
Alejevski F, Saint-Charles A, Michard-Vanhée C, Martin B, Galant S, Vasiliauskas D, Rouyer F. The HisCl1 histamine receptor acts in photoreceptors to synchronize Drosophila behavioral rhythms with light-dark cycles. Nat Commun 2019; 10:252. [PMID: 30651542 PMCID: PMC6335465 DOI: 10.1038/s41467-018-08116-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 12/19/2018] [Indexed: 01/09/2023] Open
Abstract
In Drosophila, the clock that controls rest-activity rhythms synchronizes with light-dark cycles through either the blue-light sensitive cryptochrome (Cry) located in most clock neurons, or rhodopsin-expressing histaminergic photoreceptors. Here we show that, in the absence of Cry, each of the two histamine receptors Ort and HisCl1 contribute to entrain the clock whereas no entrainment occurs in the absence of the two receptors. In contrast to Ort, HisCl1 does not restore entrainment when expressed in the optic lobe interneurons. Indeed, HisCl1 is expressed in wild-type photoreceptors and entrainment is strongly impaired in flies with photoreceptors mutant for HisCl1. Rescuing HisCl1 expression in the Rh6-expressing photoreceptors restores entrainment but it does not in other photoreceptors, which send histaminergic inputs to Rh6-expressing photoreceptors. Our results thus show that Rh6-expressing neurons contribute to circadian entrainment as both photoreceptors and interneurons, recalling the dual function of melanopsin-expressing ganglion cells in the mammalian retina.
Collapse
Affiliation(s)
- Faredin Alejevski
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Alexandra Saint-Charles
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
- Institut de la Vision, Univ. P. & M. Curie, INSERM, CNRS, Sorbonne Université, Paris, 75012, France
| | - Christine Michard-Vanhée
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Béatrice Martin
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Sonya Galant
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Daniel Vasiliauskas
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - François Rouyer
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190, Gif-sur-Yvette, France.
| |
Collapse
|
21
|
Chatterjee A, Lamaze A, De J, Mena W, Chélot E, Martin B, Hardin P, Kadener S, Emery P, Rouyer F. Reconfiguration of a Multi-oscillator Network by Light in the Drosophila Circadian Clock. Curr Biol 2018; 28:2007-2017.e4. [PMID: 29910074 PMCID: PMC6039274 DOI: 10.1016/j.cub.2018.04.064] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/28/2018] [Accepted: 04/18/2018] [Indexed: 01/02/2023]
Abstract
The brain clock that drives circadian rhythms of locomotor activity relies on a multi-oscillator neuronal network. In addition to synchronizing the clock with day-night cycles, light also reformats the clock-driven daily activity pattern. How changes in lighting conditions modify the contribution of the different oscillators to remodel the daily activity pattern remains largely unknown. Our data in Drosophila indicate that light readjusts the interactions between oscillators through two different modes. We show that a morning s-LNv > DN1p circuit works in series, whereas two parallel evening circuits are contributed by LNds and other DN1ps. Based on the photic context, the master pacemaker in the s-LNv neurons swaps its enslaved partner-oscillator-LNd in the presence of light or DN1p in the absence of light-to always link up with the most influential phase-determining oscillator. When exposure to light further increases, the light-activated LNd pacemaker becomes independent by decoupling from the s-LNvs. The calibration of coupling by light is layered on a clock-independent network interaction wherein light upregulates the expression of the PDF neuropeptide in the s-LNvs, which inhibits the behavioral output of the DN1p evening oscillator. Thus, light modifies inter-oscillator coupling and clock-independent output-gating to achieve flexibility in the network. It is likely that the light-induced changes in the Drosophila brain circadian network could reveal general principles of adapting to varying environmental cues in any neuronal multi-oscillator system.
Collapse
Affiliation(s)
- Abhishek Chatterjee
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Angélique Lamaze
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Joydeep De
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Wilson Mena
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Elisabeth Chélot
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Béatrice Martin
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Paul Hardin
- Department of Biology and Center for Biological Clocks Research, Texas A&M University, College Station, TX 77845-3258, USA
| | | | - Patrick Emery
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - François Rouyer
- Institut des Neurosciences Paris-Saclay, Univ. Paris Sud, CNRS, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
22
|
Top D, Young MW. Coordination between Differentially Regulated Circadian Clocks Generates Rhythmic Behavior. Cold Spring Harb Perspect Biol 2018; 10:a033589. [PMID: 28893860 PMCID: PMC6028074 DOI: 10.1101/cshperspect.a033589] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Specialized groups of neurons in the brain are key mediators of circadian rhythms, receiving daily environmental cues and communicating those signals to other tissues in the organism for entrainment and to organize circadian physiology. In Drosophila, the "circadian clock" is housed in seven neuronal clusters, which are defined by their expression of the main circadian proteins, Period, Timeless, Clock, and Cycle. These clusters are distributed across the fly brain and are thereby subject to the respective environments associated with their anatomical locations. While these core components are universally expressed in all neurons of the circadian network, additional regulatory proteins that act on these components are differentially expressed, giving rise to "local clocks" within the network that nonetheless converge to regulate coherent behavioral rhythms. In this review, we describe the communication between the neurons of the circadian network and the molecular differences within neurons of this network. We focus on differences in protein-expression patterns and discuss how such variation can impart functional differences in each local clock. Finally, we summarize our current understanding of how communication within the circadian network intersects with intracellular biochemical mechanisms to ultimately specify behavioral rhythms. We propose that additional efforts are required to identify regulatory mechanisms within each neuronal cluster to understand the molecular basis of circadian behavior.
Collapse
Affiliation(s)
- Deniz Top
- Laboratory of Genetics, The Rockefeller University, New York, New York 10065
| | - Michael W Young
- Laboratory of Genetics, The Rockefeller University, New York, New York 10065
| |
Collapse
|
23
|
Sugie A, Marchetti G, Tavosanis G. Structural aspects of plasticity in the nervous system of Drosophila. Neural Dev 2018; 13:14. [PMID: 29960596 PMCID: PMC6026517 DOI: 10.1186/s13064-018-0111-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Neurons extend and retract dynamically their neurites during development to form complex morphologies and to reach out to their appropriate synaptic partners. Their capacity to undergo structural rearrangements is in part maintained during adult life when it supports the animal's ability to adapt to a changing environment or to form lasting memories. Nonetheless, the signals triggering structural plasticity and the mechanisms that support it are not yet fully understood at the molecular level. Here, we focus on the nervous system of the fruit fly to ask to which extent activity modulates neuronal morphology and connectivity during development. Further, we summarize the evidence indicating that the adult nervous system of flies retains some capacity for structural plasticity at the synaptic or circuit level. For simplicity, we selected examples mostly derived from studies on the visual system and on the mushroom body, two regions of the fly brain with extensively studied neuroanatomy.
Collapse
Affiliation(s)
- Atsushi Sugie
- Center for Transdisciplinary Research, Niigata University, Niigata, 951-8585 Japan
- Brain Research Institute, Niigata University, Niigata, 951-8585 Japan
| | | | - Gaia Tavosanis
- Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| |
Collapse
|
24
|
Humberg TH, Bruegger P, Afonso B, Zlatic M, Truman JW, Gershow M, Samuel A, Sprecher SG. Dedicated photoreceptor pathways in Drosophila larvae mediate navigation by processing either spatial or temporal cues. Nat Commun 2018; 9:1260. [PMID: 29593252 PMCID: PMC5871836 DOI: 10.1038/s41467-018-03520-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 02/21/2018] [Indexed: 11/09/2022] Open
Abstract
To integrate changing environmental cues with high spatial and temporal resolution is critical for animals to orient themselves. Drosophila larvae show an effective motor program to navigate away from light sources. How the larval visual circuit processes light stimuli to control navigational decision remains unknown. The larval visual system is composed of two sensory input channels, Rhodopsin5 (Rh5) and Rhodopsin6 (Rh6) expressing photoreceptors (PRs). We here characterize how spatial and temporal information are used to control navigation. Rh6-PRs are required to perceive temporal changes of light intensity during head casts, while Rh5-PRs are required to control behaviors that allow navigation in response to spatial cues. We characterize how distinct behaviors are modulated and identify parallel acting and converging features of the visual circuit. Functional features of the larval visual circuit highlight the principle of how early in a sensory circuit distinct behaviors may be computed by partly overlapping sensory pathways.
Collapse
Affiliation(s)
| | - Pascal Bruegger
- Department of Biology, University of Fribourg, 1700, Fribourg, Switzerland
| | - Bruno Afonso
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, 20147, VA, USA
| | - Marta Zlatic
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, 20147, VA, USA.,Department of Zoology, University of Cambridge, CB2 3EJ, Cambridge, UK
| | - James W Truman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, 20147, VA, USA
| | - Marc Gershow
- Department of Physics and Center for Neural Science, New York University, New York, 10003, NY, USA
| | - Aravinthan Samuel
- Department of Physics and Center for Brain Science, Harvard University, Cambridge, 02138, MA, USA
| | - Simon G Sprecher
- Department of Biology, University of Fribourg, 1700, Fribourg, Switzerland.
| |
Collapse
|
25
|
Kistenpfennig C, Grebler R, Ogueta M, Hermann-Luibl C, Schlichting M, Stanewsky R, Senthilan PR, Helfrich-Förster C. A New Rhodopsin Influences Light-dependent Daily Activity Patterns of Fruit Flies. J Biol Rhythms 2017; 32:406-422. [PMID: 28840790 DOI: 10.1177/0748730417721826] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rhodopsin 7 ( Rh7), a new invertebrate Rhodopsin gene, was discovered in the genome of Drosophila melanogaster in 2000, but its function has remained elusive. We generated an Rh7 null mutant ( Rh70) by P element-mediated mutagenesis and found that an absence of Rh7 had significant effects on fly activity patterns during light-dark (LD) cycles: Rh70 mutants exhibited less morning activity and a longer siesta than wild-type controls. Consistent with these results, we found that Rh7 appears to be expressed in a few dorsal clock neurons that have been previously implicated in the control of the siesta. We also found putative Rh7 expression in R8 photoreceptor cells of the compound eyes and in the Hofbauer-Buchner eyelets, which have been shown to control the precise timing of locomotor activity. The absence of Rh7 alone impaired neither the flies' responses to constant white light nor the ability to follow phase shifts of white LD cycles. However, in blue light (470 nm), Rh70 mutants needed significantly longer to synchronize than wild-type controls, suggesting that Rh7 is a blue light-sensitive photopigment with a minor contribution to circadian clock synchronization. In combination with mutants that lacked additionally cryptochrome-based and/or eye-based light input to the circadian clock, the absence of Rh7 provoked slightly stronger effects.
Collapse
Affiliation(s)
- Christa Kistenpfennig
- Neurobiology and Genetics, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany.,1. Oxitec Ltd, 71 Innovation Drive, Milton Park, Abingdon, OX14 4RQ, UK
| | - Rudi Grebler
- Neurobiology and Genetics, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Maite Ogueta
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Christiane Hermann-Luibl
- Neurobiology and Genetics, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Matthias Schlichting
- Neurobiology and Genetics, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany.,2. Howard Hughes Medical Institute and National Center for Behavioral Genomics, Department of Biology, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Ralf Stanewsky
- Department of Cell and Developmental Biology, University College London, London, UK.,3. Institute for Neuro- and Behavioral Biology, Westfälische Wilhelms University, Badestraße 9/13, 48149 Münster, Germany
| | - Pingkalai R Senthilan
- Neurobiology and Genetics, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
26
|
Larderet I, Fritsch PM, Gendre N, Neagu-Maier GL, Fetter RD, Schneider-Mizell CM, Truman JW, Zlatic M, Cardona A, Sprecher SG. Organization of the Drosophila larval visual circuit. eLife 2017; 6:28387. [PMID: 30726702 PMCID: PMC5577918 DOI: 10.7554/elife.28387] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/07/2017] [Indexed: 11/20/2022] Open
Abstract
Visual systems transduce, process and transmit light-dependent environmental cues. Computation of visual features depends on photoreceptor neuron types (PR) present, organization of the eye and wiring of the underlying neural circuit. Here, we describe the circuit architecture of the visual system of Drosophila larvae by mapping the synaptic wiring diagram and neurotransmitters. By contacting different targets, the two larval PR-subtypes create two converging pathways potentially underlying the computation of ambient light intensity and temporal light changes already within this first visual processing center. Locally processed visual information then signals via dedicated projection interneurons to higher brain areas including the lateral horn and mushroom body. The stratified structure of the larval optic neuropil (LON) suggests common organizational principles with the adult fly and vertebrate visual systems. The complete synaptic wiring diagram of the LON paves the way to understanding how circuits with reduced numerical complexity control wide ranges of behaviors.
Collapse
Affiliation(s)
- Ivan Larderet
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Nanae Gendre
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Richard D Fetter
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | | | - James W Truman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Marta Zlatic
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Albert Cardona
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Simon G Sprecher
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
27
|
Haeme oxygenase protects against UV light DNA damages in the retina in clock-dependent manner. Sci Rep 2017; 7:5197. [PMID: 28701782 PMCID: PMC5507878 DOI: 10.1038/s41598-017-05418-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/30/2017] [Indexed: 11/09/2022] Open
Abstract
In the present study, we showed that in the retina of Drosophila, the expression of the ho gene, encoding haeme oxygenase (HO), is regulated by light but only at the beginning of the day. This timing must be set by the circadian clock as light pulses applied at other time points during the day do not increase the ho mRNA level. Moreover, light-induced activation of HO does not depend on the canonical phototransduction pathway but instead involves cryptochrome and is enhanced by ultraviolet (UV) light. Interestingly, the level of DNA damage in the retina after UV exposure was inversely related to the circadian oscillation of the ho mRNA level during the night, being the highest when the HO level was low and reversed during the day. Accordingly, induction of HO by hemin was associated with low DNA damage, while inhibition of HO activity by SnPPIX aggravated the damage. Our data suggest that HO acts in the retina to decrease oxidative DNA damage in photoreceptors caused by UV-rich light in the morning.
Collapse
|
28
|
Ni JD, Baik LS, Holmes TC, Montell C. A rhodopsin in the brain functions in circadian photoentrainment in Drosophila. Nature 2017; 545:340-344. [PMID: 28489826 PMCID: PMC5476302 DOI: 10.1038/nature22325] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 03/30/2017] [Indexed: 11/09/2022]
Abstract
Animals partition their daily activity rhythms through their internal circadian clocks, which are synchronized by oscillating day-night cycles of light. The fruit fly, Drosophila melanogaster, senses day/night cycles in part through rhodopsin-dependent light reception in the compound eye, and photoreceptor cells in the Hofbauer-Buchner (H-B) eyelet1. However, a more significant light entrainment pathway is mediated in central pacemaker neurons in the brain. The Drosophila circadian clock is extremely light sensitive. However, the only known light sensor in pacemaker neurons, the flavoprotein, cryptochrome (Cry)2,3, responds only to high levels of light in vitro4. These observations indicate the existence of an additional light-sensing pathway in fly pacemaker neurons5. Here, we identified an uncharacterized rhodopsin, Rh7, which functions in circadian light entrainment through circadian pacemaker neurons in the brain. The pacemaker neurons respond to violet light, which was dependent on Rh7. While loss of either cry or rh7 caused minor affects on photoentrainment, the defects in the double mutant were profound. The circadian photoresponse to constant light was impaired in the rh7 mutant, especially under dim light. The demonstration that Rh7 functions in circadian pacemaker neurons represents the first role for an opsin in the central brain.
Collapse
Affiliation(s)
- Jinfei D Ni
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California 93106, USA.,Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Lisa S Baik
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California 92697, USA
| | - Todd C Holmes
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, California 92697, USA.,Center for Circadian Biology, University of California, San Diego, La Jolla, California 92093, USA
| | - Craig Montell
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California 93106, USA
| |
Collapse
|
29
|
Humberg TH, Sprecher SG. Age- and Wavelength-Dependency of Drosophila Larval Phototaxis and Behavioral Responses to Natural Lighting Conditions. Front Behav Neurosci 2017; 11:66. [PMID: 28473759 PMCID: PMC5397426 DOI: 10.3389/fnbeh.2017.00066] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/03/2017] [Indexed: 12/14/2022] Open
Abstract
Animals use various environmental cues as key determinant for their behavioral decisions. Visual systems are hereby responsible to translate light-dependent stimuli into neuronal encoded information. Even though the larval eyes of the fruit fly Drosophila melanogaster are comparably simple, they comprise two types of photoreceptor neurons (PRs), defined by different Rhodopsin genes expressed. Recent findings support that for light avoidance Rhodopsin5 (Rh5) expressing photoreceptors are crucial, while Rhodopsin6 (Rh6) expressing photoreceptors are dispensable under laboratory conditions. However, it remains debated how animals change light preference during larval live. We show that larval negative phototaxis is age-independent as it persists in larvae from foraging to wandering developmental stages. Moreover, if spectrally different Rhodopsins are employed for the detection of different wavelength of light remains unexplored. We found that negative phototaxis can be elicit by light with wavelengths ranging from ultraviolet (UV) to green. This behavior is uniquely mediated by Rh5 expressing photoreceptors, and therefore suggest that this photoreceptor-type is able to perceive UV up to green light. In contrast to laboratory our field experiments revealed that Drosophila larvae uses both types of photoreceptors under natural lighting conditions. All our results, demonstrate that Drosophila larval eyes mediate avoidance of light stimuli with a wide, ecological relevant range of quantity (intensities) and quality (wavelengths). Thus, the two photoreceptor-types appear more likely to play a role in different aspects of phototaxis under natural lighting conditions, rather than color discrimination.
Collapse
Affiliation(s)
| | - Simon G Sprecher
- Department of Biology, University of FribourgFribourg, Switzerland
| |
Collapse
|
30
|
Abruzzi KC, Zadina A, Luo W, Wiyanto E, Rahman R, Guo F, Shafer O, Rosbash M. RNA-seq analysis of Drosophila clock and non-clock neurons reveals neuron-specific cycling and novel candidate neuropeptides. PLoS Genet 2017; 13:e1006613. [PMID: 28182648 PMCID: PMC5325595 DOI: 10.1371/journal.pgen.1006613] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/24/2017] [Accepted: 02/01/2017] [Indexed: 12/21/2022] Open
Abstract
Locomotor activity rhythms are controlled by a network of ~150 circadian neurons within the adult Drosophila brain. They are subdivided based on their anatomical locations and properties. We profiled transcripts “around the clock” from three key groups of circadian neurons with different functions. We also profiled a non-circadian outgroup, dopaminergic (TH) neurons. They have cycling transcripts but fewer than clock neurons as well as low expression and poor cycling of clock gene transcripts. This suggests that TH neurons do not have a canonical circadian clock and that their gene expression cycling is driven by brain systemic cues. The three circadian groups are surprisingly diverse in their cycling transcripts and overall gene expression patterns, which include known and putative novel neuropeptides. Even the overall phase distributions of cycling transcripts are distinct, indicating that different regulatory principles govern transcript oscillations. This surprising cell-type diversity parallels the functional heterogeneity of the different neurons. Organisms ranging from bacteria to humans contain circadian clocks. They keep internal time and also integrate environmental cues such as light to provide external time information for entrainment. In the fruit fly Drosophila melanogaster, ~150 brain neurons contain the circadian machinery and are critical for controlling behavior. Several subgroups of these clock neurons have been identified by their anatomical locations and specific functions. Our work aims to profile these neurons and to characterize their molecular contents: what to they contain and how do they differ? To this end, we have purified 3 important subgroups of clock neurons and identified their expressed genes at different times of day. Some are expressed at all times, whereas others are “cycling,” i.e., expressed more strongly at a particular time of day like the morning. Interestingly, each circadian subgroup is quite different. The data provide hints about what functions each group of neurons carries out and how they may work together to keep time. In addition, even a non-circadian group of neurons has cycling genes and has implications for the extent to which all cells have or do not have a functional circadian clock.
Collapse
Affiliation(s)
- Katharine C. Abruzzi
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Abigail Zadina
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Weifei Luo
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Evelyn Wiyanto
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Reazur Rahman
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Fang Guo
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Orie Shafer
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
| | - Michael Rosbash
- Howard Hughes Medical Institute and National Center for Behavioral Genomics,Department of Biology, Brandeis University, Waltham, United States of America
- * E-mail:
| |
Collapse
|
31
|
CRYPTOCHROME mediates behavioral executive choice in response to UV light. Proc Natl Acad Sci U S A 2017; 114:776-781. [PMID: 28062690 DOI: 10.1073/pnas.1607989114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Drosophila melanogaster CRYPTOCHROME (CRY) mediates behavioral and electrophysiological responses to blue light coded by circadian and arousal neurons. However, spectroscopic and biochemical assays of heterologously expressed CRY suggest that CRY may mediate functional responses to UV-A (ultraviolet A) light as well. To determine the relative contributions of distinct phototransduction systems, we tested mutants lacking CRY and mutants with disrupted opsin-based phototransduction for behavioral and electrophysiological responses to UV light. CRY and opsin-based external photoreceptor systems cooperate for UV light-evoked acute responses. CRY mediates behavioral avoidance responses related to executive choice, consistent with its expression in central brain neurons.
Collapse
|
32
|
H2O2-Sensitive Isoforms of Drosophila melanogaster TRPA1 Act in Bitter-Sensing Gustatory Neurons to Promote Avoidance of UV During Egg-Laying. Genetics 2016; 205:749-759. [PMID: 27932542 DOI: 10.1534/genetics.116.195172] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/27/2016] [Indexed: 01/12/2023] Open
Abstract
The evolutionarily conserved TRPA1 channel can sense various stimuli including temperatures and chemical irritants. Recent results have suggested that specific isoforms of Drosophila TRPA1 (dTRPA1) are UV-sensitive and that their UV sensitivity is due to H2O2 sensitivity. However, whether such UV sensitivity served any physiological purposes in animal behavior was unclear. Here, we demonstrate that H2O2-sensitive dTRPA1 isoforms promote avoidance of UV when adult Drosophila females are selecting sites for egg-laying. First, we show that blind/visionless females are still capable of sensing and avoiding UV during egg-laying when intensity of UV is high yet within the range of natural sunlight. Second, we show that such vision-independent UV avoidance is mediated by a group of bitter-sensing neurons on the proboscis that express H2O2-sensitive dTRPA1 isoforms. We show that these bitter-sensing neurons exhibit dTRPA1-dependent UV sensitivity. Importantly, inhibiting activities of these bitter-sensing neurons, reducing their dTRPA1 expression, or reducing their H2O2-sensitivity all significantly reduced blind females' UV avoidance, whereas selectively restoring a H2O2-sensitive isoform of dTRPA1 in these neurons restored UV avoidance. Lastly, we show that specifically expressing the red-shifted channelrhodopsin CsChrimson in these bitter-sensing neurons promotes egg-laying avoidance of red light, an otherwise neutral cue for egg-laying females. Together, these results demonstrate a physiological role of the UV-sensitive dTRPA1 isoforms, reveal that adult Drosophila possess at least two sensory systems for detecting UV, and uncover an unexpected role of bitter-sensing taste neurons in UV sensing.
Collapse
|
33
|
Malpel S, Klarsfeld A, Rouyer F. Circadian Synchronization and Rhythmicity in Larval Photoperception-Defective Mutants of Drosophila. J Biol Rhythms 2016; 19:10-21. [PMID: 14964700 DOI: 10.1177/0748730403260621] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A single light episode during the first larval stage can set the phase of adult Drosophila activity rhythms, showing that a light-sensitive circadian clock is functional in larvae and is capable of keeping time throughout development. These behavioral data are supported by the finding that neurons expressing clock proteins already exist in the larval brain and appear to be connected to the larval visual system. To define the photoreceptive pathways of the larval clock, the authors investigated circadian synchronization during larval stages in various visual systems and/or cryptochrome-defective strains. They show that adult activity rhythms cannot be entrained by light applied to larvae lacking both cryptochrome and the visual system, although such rhythms were entrained by larval stage-restricted temperature cycles. Larvae lacking either pathway alone were light entrainable, but the phase of the resulting adult rhythm was advanced relative to wild-type flies. Unexpectedly, adult behavioral rhythms of the glass60jand norpAP24visual system mutants that were entrained in the same conditions were found to be severely impaired, in contrast to those of the wild type. Extension of the entrainment until the adult stage restored close to wild-type behavioral rhythms in the mutants. The results show that both cryptochrome and the larval visual system participate to circadian photoreception in larvae and that mutations affecting the visual system can impair behavioral rhythmicity.
Collapse
Affiliation(s)
- Sébastien Malpel
- Institut de Neurobiologie Alfred Fessard, CNRS UPR 2216 (NGI), 91198 Gif-sur-Yvette, France
| | | | | |
Collapse
|
34
|
Kingston ACN, Cronin TW. Diverse Distributions of Extraocular Opsins in Crustaceans, Cephalopods, and Fish. Integr Comp Biol 2016; 56:820-833. [PMID: 27252200 DOI: 10.1093/icb/icw022] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Non-visual and extraocular photoreceptors are common among animals, but current understanding linking molecular pathways to physiological function of these receptors is lacking. Opsin diversity in extraocular tissues suggests that many putative extraocular photoreceptors utilize the "visual" phototransduction pathway-the same phototransduction pathway as photoreceptors within the retina dedicated to light detection for image sensing. Here, we provide a brief overview of the current understanding of non-visual and extraocular photoreceptors, and contribute a synopsis of several novel putative extraocular photoreceptors that use both visual and non-visual phototransduction pathways. Crayfish, cephalopods, and flat fish express opsins in diverse tissues, suggesting the presence of extraocular photoreceptors. In most cases, we find that these animals use the same phototransduction pathway that is utilized in the retinas for image-formation. However, we also find the presence of non-visual phototransduction components in the skin of flounders. Our evidence suggests that extraocular photoreceptors may employ a number of phototransduction pathways that do not appear to correlate with purpose or location of the photoreceptor.
Collapse
Affiliation(s)
- Alexandra C N Kingston
- Department of Biological Sciences, University of Maryland Baltimore County 1000 Hilltop Circle, Baltimore, MD 21250, USA
| | - Thomas W Cronin
- Department of Biological Sciences, University of Maryland Baltimore County 1000 Hilltop Circle, Baltimore, MD 21250, USA
| |
Collapse
|
35
|
Saint-Charles A, Michard-Vanhée C, Alejevski F, Chélot E, Boivin A, Rouyer F. Four of the six Drosophila rhodopsin-expressing photoreceptors can mediate circadian entrainment in low light. J Comp Neurol 2016; 524:2828-44. [PMID: 26972685 DOI: 10.1002/cne.23994] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 12/30/2022]
Abstract
Light is the major stimulus for the synchronization of circadian clocks with day-night cycles. The light-driven entrainment of the clock that controls rest-activity rhythms in Drosophila relies on different photoreceptive molecules. Cryptochrome (CRY) is expressed in most brain clock neurons, whereas six different rhodopsins (RH) are present in the light-sensing organs. The compound eye includes outer photoreceptors that express RH1 and inner photoreceptors that each express one of the four rhodopsins RH3-RH6. RH6 is also expressed in the extraretinal Hofbauer-Buchner eyelet, whereas RH2 is only found in the ocelli. In low light, the synchronization of behavioral rhythms relies on either CRY or the canonical rhodopsin phototransduction pathway, which requires the phospholipase C-β encoded by norpA (no receptor potential A). We used norpA(P24) cry(02) double mutants that are circadianly blind in low light and restored NORPA function in each of the six types of photoreceptors, defined as expressing a particular rhodopsin. We first show that the NORPA pathway is less efficient than CRY for synchronizing rest-activity rhythms with delayed light-dark cycles but is important for proper phasing, whereas the two light-sensing pathways can mediate efficient adjustments to phase advances. Four of the six rhodopsin-expressing photoreceptors can mediate circadian entrainment, and all are more efficient for advancing than for delaying the behavioral clock. In contrast, neither RH5-expressing retinal photoreceptors nor RH2-expressing ocellar photoreceptors are sufficient to mediate synchronization through the NORPA pathway. Our results thus reveal different contributions of rhodopsin-expressing photoreceptors and suggest the existence of several circuits for rhodopsin-dependent circadian entrainment. J. Comp. Neurol. 524:2828-2844, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alexandra Saint-Charles
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifque, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Christine Michard-Vanhée
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifque, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Faredin Alejevski
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifque, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Elisabeth Chélot
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifque, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - Antoine Boivin
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifque, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| | - François Rouyer
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifque, Université Paris-Saclay, 91190, Gif-sur-Yvette, France
| |
Collapse
|
36
|
Yoshii T, Hermann-Luibl C, Helfrich-Förster C. Circadian light-input pathways in Drosophila. Commun Integr Biol 2016; 9:e1102805. [PMID: 27066180 PMCID: PMC4802797 DOI: 10.1080/19420889.2015.1102805] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 09/25/2015] [Accepted: 09/25/2015] [Indexed: 12/02/2022] Open
Abstract
Light is the most important environmental cue to entrain the circadian clock in most animals. In the fruit fly Drosophila melanogaster, the light entrainment mechanisms of the clock have been well-studied. The Drosophila brain contains approximately 150 neurons that rhythmically express circadian clock genes. These neurons are called "clock neurons" and control behavioral activity rhythms. Many clock neurons express the Cryptochrome (CRY) protein, which is sensitive to UV and blue light, and thus enables clock neurons deep in the brain to directly perceive light. In addition to the CRY protein, external photoreceptors in the Drosophila eyes play an important role in circadian light-input pathways. Recent studies have provided new insights into the mechanisms that integrate these light inputs into the circadian network of the brain. In this review, we will summarize the current knowledge on the light entrainment pathways in the Drosophila circadian clock.
Collapse
Affiliation(s)
- Taishi Yoshii
- Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Christiane Hermann-Luibl
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, Am Hubland, Würzburg, Germany
| |
Collapse
|
37
|
Carvalho MJA, Mirth CK. Coordinating morphology with behavior during development: an integrative approach from a fly perspective. Front Ecol Evol 2015. [DOI: 10.3389/fevo.2015.00005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
38
|
Guo F, Cerullo I, Chen X, Rosbash M. PDF neuron firing phase-shifts key circadian activity neurons in Drosophila. eLife 2014; 3. [PMID: 24939987 PMCID: PMC4092873 DOI: 10.7554/elife.02780] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/16/2014] [Indexed: 01/06/2023] Open
Abstract
Our experiments address two long-standing models for the function of the Drosophila brain circadian network: a dual oscillator model, which emphasizes the primacy of PDF-containing neurons, and a cell-autonomous model for circadian phase adjustment. We identify five different circadian (E) neurons that are a major source of rhythmicity and locomotor activity. Brief firing of PDF cells at different times of day generates a phase response curve (PRC), which mimics a light-mediated PRC and requires PDF receptor expression in the five E neurons. Firing also resembles light by causing TIM degradation in downstream neurons. Unlike light however, firing-mediated phase-shifting is CRY-independent and exploits the E3 ligase component CUL-3 in the early night to degrade TIM. Our results suggest that PDF neurons integrate light information and then modulate the phase of E cell oscillations and behavioral rhythms. The results also explain how fly brain rhythms persist in constant darkness and without CRY. DOI:http://dx.doi.org/10.7554/eLife.02780.001 Most animals have daily rhythms of activity: some are awake during the day and asleep at night, whilst others are more active at night, or during the twilight hours around dawn and dusk. These cycles of activity are driven by an internal body clock, which is reset in response to external cues, like light and temperature, and which keeps the animal in sync with the day–night cycle. The fruit fly Drosophila has daily—or circadian—rhythms of behavior, which are controlled by a network of genes that are switched ‘on’ or ‘off’ at different times in every 24-hr period. These circadian genes encode various proteins, including PERIOD and TIMELESS. The levels of these two proteins increase during the day and into the night, until they reach a point at which they cause their own genes to be switched off. PERIOD and TIMELESS are then destroyed each morning, and the cycle begins anew. Most of these same proteins perform similar functions in mammals. In the fly brain, two groups of neurons express these key proteins and control the timings of activity or movement. One group, called M cells, regulates activity in the morning and also produces a small molecule called PDF. Another group, called E cells, controls evening activity, but is less well-defined. Since M cells can maintain circadian rhythms even in total darkness, these cells were also considered key ‘pacemaker neurons’. However, Guo et al. now challenge this view by identifying five E cells that are the major source of circadian activity. Blocking the release of signaling molecules from these neurons severely disrupted movement in both the morning and the evening. The E cells are also critical for timekeeping under a normal day–night cycle. Guo et al. found that the rhythm of the E cells is reset when the M cell neurons fire, which causes a release of PDF molecules. Further, PDF molecules reset the E cells by causing the degradation of the TIMELESS protein—which is similar to the effect of light, although light cause TIMELESS to be degraded via a different biochemical pathway. Guo et al. suggest that under normal light–dark conditions the E cells are important for driving the flies' activity as well as for overall timekeeping. The M cells, instead, appear to function primarily to integrate information about light and reset the E cell clock. Challenges moving forward will include understanding other ways in which the firing of neurons can affect timekeeping, as well as looking if there any differences between the five E cells. DOI:http://dx.doi.org/10.7554/eLife.02780.002
Collapse
Affiliation(s)
- Fang Guo
- Department of Biology, Brandeis University, Waltham, United States
| | - Isadora Cerullo
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Xiao Chen
- Department of Biology, Brandeis University, Waltham, United States
| | - Michael Rosbash
- Department of Biology, Brandeis University, Waltham, United States
| |
Collapse
|
39
|
Helfrich-Förster C. From neurogenetic studies in the fly brain to a concept in circadian biology. J Neurogenet 2014; 28:329-47. [PMID: 24655073 DOI: 10.3109/01677063.2014.905556] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This paper is dedicated to Karl-Friedrich Fischbach, who has always shared with me the interest in the function of the fly brain, especially that of its optic lobes. He has accompanied me during my first steps in scientific research. The paper tells the story how our first common attempts to localize the circadian clock in the fly brain finally helped in phrasing the two-oscillator principle of circadian clocks that seems to be valid far beyond the fly circadian system. I hope that Karl-Friedrich will take this story as praise for his generosity in supporting younger scientists outside his own lab, even without the reward of a common paper.
Collapse
Affiliation(s)
- Charlotte Helfrich-Förster
- Neurobiology and Genetics, Biocenter, Theodor-Boveri Institute, University of Würzburg , Würzburg , Germany
| |
Collapse
|
40
|
Chahad-Ehlers S, Gentile C, Lima JBP, Peixoto AA, Bruno RV. Analysis of cycle gene expression in Aedes aegypti brains by in situ hybridization. PLoS One 2013; 8:e52559. [PMID: 23300979 PMCID: PMC3534671 DOI: 10.1371/journal.pone.0052559] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/16/2012] [Indexed: 12/29/2022] Open
Abstract
Even though the blood-sucking mosquito Aedes aegypti is one of the most important disease vectors, relatively little is known about the molecular mechanisms underlying processes involved in the temporal pattern of its activity and host seeking behavior. In this study, we analyzed the expression of the cycle (cyc) gene, one of the core components of the circadian clock, in Ae. aegypti brains by in situ hybridization at two different time points in light-dark conditions and compared the results with those obtained using a quantitative PCR assay (qPCR). Within the brain, differential labeling was detected according to distinct areas empirically pre-defined. Six out of seven of these areas showed significantly higher staining at ZT3 (three hours after light-on) compared to ZT11 (one before light-off), which is consistent with the qPCR data. Predominant staining was observed in three of those areas which correspond to positions of the optical and antennal lobes, as well as the region where the neurons controlling activity rhythms are presumably localized.
Collapse
Affiliation(s)
- Samira Chahad-Ehlers
- Laboratório de Biologia Molecular de Insetos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Carla Gentile
- Laboratório de Biologia Molecular de Insetos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - José Bento Pereira Lima
- Laboratório de Fisiologia e Controle de Artrópodes Vetores, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratório de Entomologia, Instituto de Biologia do Exército, Rio de Janeiro, Brazil
- Instituto Nacional de Ciencia e Tecnologia em Entomologia Molecular, CNPq, Rio de Janeiro, Brazil
| | - Alexandre Afranio Peixoto
- Laboratório de Biologia Molecular de Insetos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Instituto Nacional de Ciencia e Tecnologia em Entomologia Molecular, CNPq, Rio de Janeiro, Brazil
| | - Rafaela Vieira Bruno
- Laboratório de Biologia Molecular de Insetos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Instituto Nacional de Ciencia e Tecnologia em Entomologia Molecular, CNPq, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
41
|
Beuchle D, Jaumouillé E, Nagoshi E. The nuclear receptor unfulfilled is required for free-running clocks in Drosophila pacemaker neurons. Curr Biol 2012; 22:1221-7. [PMID: 22658597 DOI: 10.1016/j.cub.2012.04.052] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/26/2012] [Accepted: 04/24/2012] [Indexed: 10/28/2022]
Abstract
An intricate neural circuit composed of multiple classes of clock neurons controls circadian locomotor rhythms in Drosophila. Evidence indicates that the small ventral lateral neurons (s-LNvs, M cells) are the dominant pacemaker neurons that synchronize the clocks throughout the circuit and drive free-running locomotor rhythms. Little is known, however, about the molecular underpinning of this unique function of the s-LNvs. Here, we show that the nuclear receptor gene unfulfilled (unf; DHR51) is required for the function of the s-LNvs. UNFULFILLED (UNF) is rhythmically expressed in the s-LNvs, and unf mutant flies are behaviorally arrhythmic. Knockdown of unf in developing LNvs irreversibly destroys the ability of adult s-LNvs to generate free-running rhythms, whereas depletion of UNF from adult LNvs dampens the rhythms of the s-LNvs only in constant darkness. These temporally controlled LNv-targeted unf knockdowns desynchronize circuit-wide molecular rhythms and disrupt behavioral rhythms. Therefore, UNF is a prerequisite for free-running clocks in the s-LNvs and for the function of the entire circadian circuit.
Collapse
Affiliation(s)
- Dirk Beuchle
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | | | | |
Collapse
|
42
|
Szular J, Sehadova H, Gentile C, Szabo G, Chou WH, Britt SG, Stanewsky R. Rhodopsin 5- and Rhodopsin 6-mediated clock synchronization in Drosophila melanogaster is independent of retinal phospholipase C-β signaling. J Biol Rhythms 2012; 27:25-36. [PMID: 22306971 DOI: 10.1177/0748730411431673] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Circadian clocks of most organisms are synchronized with the 24-hour solar day by the changes of light and dark. In Drosophila, both the visual photoreceptors in the compound eyes as well as the blue-light photoreceptor Cryptochrome expressed within the brain clock neurons contribute to this clock synchronization. A specialized photoreceptive structure located between the retina and the optic lobes, the Hofbauer-Buchner (H-B) eyelet, projects to the clock neurons in the brain and also participates in light synchronization. The compound eye photoreceptors and the H-B eyelet contain Rhodopsin photopigments, which activate the canonical invertebrate phototransduction cascade after being excited by light. We show here that 2 of the photopigments present in these photoreceptors, Rhodopsin 5 (Rh5) and Rhodopsin 6 (Rh6), contribute to light synchronization in a mutant (norpA(P41) ) that disrupts canonical phototransduction due to the absence of Phospholipase C-β (PLC-β). We reveal that norpA(P41) is a true loss-of-function allele, resulting in a truncated PLC-β protein that lacks the catalytic domain. Light reception mediated by Rh5 and Rh6 must therefore utilize either a different (nonretinal) PLC-β enzyme or alternative signaling mechanisms, at least in terms of clock-relevant photoreception. This novel signaling mode may distinguish Rhodopsin-mediated irradiance detection from image-forming vision in Drosophila.
Collapse
Affiliation(s)
- Joanna Szular
- School of Biological and Chemical Sciences, Queen Mary, University of London, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
43
|
Yao Z, Macara AM, Lelito KR, Minosyan TY, Shafer OT. Analysis of functional neuronal connectivity in the Drosophila brain. J Neurophysiol 2012; 108:684-96. [PMID: 22539819 DOI: 10.1152/jn.00110.2012] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Drosophila melanogaster is a valuable model system for the neural basis of complex behavior, but an inability to routinely interrogate physiologic connections within central neural networks of the fly brain remains a fundamental barrier to progress in the field. To address this problem, we have introduced a simple method of measuring functional connectivity based on the independent expression of the mammalian P2X2 purinoreceptor and genetically encoded Ca(2+) and cAMP sensors within separate genetically defined subsets of neurons in the adult brain. We show that such independent expression is capable of specifically rendering defined sets of neurons excitable by pulses of bath-applied ATP in a manner compatible with high-resolution Ca(2+) and cAMP imaging in putative follower neurons. Furthermore, we establish that this approach is sufficiently sensitive for the detection of excitatory and modulatory connections deep within larval and adult brains. This technically facile approach can now be used in wild-type and mutant genetic backgrounds to address functional connectivity within neuronal networks governing a wide range of complex behaviors in the fly. Furthermore, the effectiveness of this approach in the fly brain suggests that similar methods using appropriate heterologous receptors might be adopted for other widely used model systems.
Collapse
Affiliation(s)
- Zepeng Yao
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | | | | | | | | |
Collapse
|
44
|
Peripheral circadian rhythms and their regulatory mechanism in insects and some other arthropods: a review. J Comp Physiol B 2012; 182:729-40. [PMID: 22327195 DOI: 10.1007/s00360-012-0651-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 01/18/2012] [Accepted: 01/26/2012] [Indexed: 01/15/2023]
Abstract
Many physiological functions of insects show a rhythmic change to adapt to daily environmental cycles. These rhythms are controlled by a multi-clock system. A principal clock located in the brain usually organizes the overall behavioral rhythms, so that it is called the "central clock". However, the rhythms observed in a variety of peripheral tissues are often driven by clocks that reside in those tissues. Such autonomous rhythms can be found in sensory organs, digestive and reproductive systems. Using Drosophila melanogaster as a model organism, researchers have revealed that the peripheral clocks are self-sustained oscillators with a molecular machinery slightly different from that of the central clock. However, individual clocks normally run in harmony with each other to keep a coordinated temporal structure within an animal. How can this be achieved? What is the molecular mechanism underlying the oscillation? Also how are the peripheral clocks entrained by light-dark cycles? There are still many questions remaining in this research field. In the last several years, molecular techniques have become available in non-model insects so that the molecular oscillatory mechanisms are comparatively investigated among different insects, which give us more hints to understand the essential regulatory mechanism of the multi-oscillatory system across insects and other arthropods. Here we review current knowledge on arthropod's peripheral clocks and discuss their physiological roles and molecular mechanisms.
Collapse
|
45
|
Lelito KR, Shafer OT. Reciprocal cholinergic and GABAergic modulation of the small ventrolateral pacemaker neurons of Drosophila's circadian clock neuron network. J Neurophysiol 2012; 107:2096-108. [PMID: 22279191 DOI: 10.1152/jn.00931.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The relatively simple clock neuron network of Drosophila is a valuable model system for the neuronal basis of circadian timekeeping. Unfortunately, many key neuronal classes of this network are inaccessible to electrophysiological analysis. We have therefore adopted the use of genetically encoded sensors to address the physiology of the fly's circadian clock network. Using genetically encoded Ca(2+) and cAMP sensors, we have investigated the physiological responses of two specific classes of clock neuron, the large and small ventrolateral neurons (l- and s-LN(v)s), to two neurotransmitters implicated in their modulation: acetylcholine (ACh) and γ-aminobutyric acid (GABA). Live imaging of l-LN(v) cAMP and Ca(2+) dynamics in response to cholinergic agonist and GABA application were well aligned with published electrophysiological data, indicating that our sensors were capable of faithfully reporting acute physiological responses to these transmitters within single adult clock neuron soma. We extended these live imaging methods to s-LN(v)s, critical neuronal pacemakers whose physiological properties in the adult brain are largely unknown. Our s-LN(v) experiments revealed the predicted excitatory responses to bath-applied cholinergic agonists and the predicted inhibitory effects of GABA and established that the antagonism of ACh and GABA extends to their effects on cAMP signaling. These data support recently published but physiologically untested models of s-LN(v) modulation and lead to the prediction that cholinergic and GABAergic inputs to s-LN(v)s will have opposing effects on the phase and/or period of the molecular clock within these critical pacemaker neurons.
Collapse
Affiliation(s)
- Katherine R Lelito
- Dept. of Molecular, Cellular, and Developmental Biology, Univ. of Michigan, Ann Arbor, MI 48109-1048, USA
| | | |
Collapse
|
46
|
Abstract
TRP channels have emerged as key biological sensors in vision, taste, olfaction, hearing, and touch. Despite their importance, virtually nothing is known about the folding and transport of TRP channels during biosynthesis. Here, we identify XPORT (exit protein of rhodopsin and TRP) as a critical chaperone for TRP and its G protein-coupled receptor (GPCR), rhodopsin (Rh1). XPORT is a resident ER and secretory pathway protein that interacts with TRP and Rh1, as well as with Hsp27 and Hsp90. XPORT promotes the targeting of TRP to the membrane in Drosophila S2 cells, a finding that provides a critical first step toward solving a longstanding problem in the successful heterologous expression of TRP. Mutations in xport result in defective transport of TRP and Rh1, leading to retinal degeneration. Our results identify XPORT as a molecular chaperone and provide a mechanistic link between TRP channels and their GPCRs during biosynthesis and transport.
Collapse
|
47
|
Yoshii T, Rieger D, Helfrich-Förster C. Two clocks in the brain. PROGRESS IN BRAIN RESEARCH 2012; 199:59-82. [DOI: 10.1016/b978-0-444-59427-3.00027-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Yuan Q, Xiang Y, Yan Z, Han C, Jan LY, Jan YN. Light-induced structural and functional plasticity in Drosophila larval visual system. Science 2011; 333:1458-62. [PMID: 21903815 DOI: 10.1126/science.1207121] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
How to build and maintain a reliable yet flexible circuit is a fundamental question in neurobiology. The nervous system has the capacity for undergoing modifications to adapt to the changing environment while maintaining its stability through compensatory mechanisms, such as synaptic homeostasis. Here, we describe our findings in the Drosophila larval visual system, where the variation of sensory inputs induced substantial structural plasticity in dendritic arbors of the postsynaptic neuron and concomitant changes to its physiological output. Furthermore, our genetic analyses have identified the cyclic adenosine monophosphate (cAMP) pathway and a previously uncharacterized cell surface molecule as critical components in regulating experience-dependent modification of the postsynaptic dendrite morphology in Drosophila.
Collapse
Affiliation(s)
- Quan Yuan
- Howard Hughes Medical Institute, Department of Physiology and Biochemistry, University of California, San Francisco, 1550 4th Street, San Francisco, CA 94158, USA
| | | | | | | | | | | |
Collapse
|
49
|
Sprecher SG, Cardona A, Hartenstein V. The Drosophila larval visual system: high-resolution analysis of a simple visual neuropil. Dev Biol 2011; 358:33-43. [PMID: 21781960 DOI: 10.1016/j.ydbio.2011.07.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2011] [Revised: 06/30/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022]
Abstract
The task of the visual system is to translate light into neuronal encoded information. This translation of photons into neuronal signals is achieved by photoreceptor neurons (PRs), specialized sensory neurons, located in the eye. Upon perception of light the PRs will send a signal to target neurons, which represent a first station of visual processing. Increasing complexity of visual processing stems from the number of distinct PR subtypes and their various types of target neurons that are contacted. The visual system of the fruit fly larva represents a simple visual system (larval optic neuropil, LON) that consists of 12 PRs falling into two classes: blue-senstive PRs expressing Rhodopsin 5 (Rh5) and green-sensitive PRs expressing Rhodopsin 6 (Rh6). These afferents contact a small number of target neurons, including optic lobe pioneers (OLPs) and lateral clock neurons (LNs). We combine the use of genetic markers to label both PR subtypes and the distinct, identifiable sets of target neurons with a serial EM reconstruction to generate a high-resolution map of the larval optic neuropil. We find that the larval optic neuropil shows a clear bipartite organization consisting of one domain innervated by PRs and one devoid of PR axons. The topology of PR projections, in particular the relationship between Rh5 and Rh6 afferents, is maintained from the nerve entering the brain to the axon terminals. The target neurons can be subdivided according to neurotransmitter or neuropeptide they use as well as the location within the brain. We further track the larval optic neuropil through development from first larval instar to its location in the adult brain as the accessory medulla.
Collapse
Affiliation(s)
- Simon G Sprecher
- Institute of Developmental and Cell Biology, Department of Biology, University of Fribourg, Chemin du Musee 10, 1700, Fribourg, Switzerland
| | | | | |
Collapse
|
50
|
Yoshii T, Hermann C, Helfrich-Förster C. Cryptochrome-Positive and -Negative Clock Neurons in Drosophila Entrain Differentially to Light and Temperature. J Biol Rhythms 2010; 25:387-98. [DOI: 10.1177/0748730410381962] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The blue-light photoreceptive protein Cryptochrome (CRY) plays an important role in the light synchronization of the Drosophila circadian clock. Previously, we found that among the approximately 150 clock neurons, many but not all neurons express CRY. We speculated that the CRY-positive pacemaker neurons may be especially important for light entrainment, whereas the CRY-negative neurons may be important for other environmental cues, for example, temperature. To investigate this hypothesis, we tested the entrainability of the clock neurons to out-of-phase light and temperature cycles. When light-dark or light-dim light cycles were shifted by 12 h with respect to temperature cycles, behavioral rhythms of wild-type flies were re-entrained by the light cycles. In this condition, we found that TIMELESS (TIM) level was strongly influenced by the temperature cycles in many CRY-negative clock neurons, suggesting that the CRY-negative neurons have higher sensitivity to temperature. Under the same conditions, cry-null mutants entrained to the temperature cycles or very slowly re-entrained to light-dark cycles. Our results suggest that there are 2 types of clock neurons having differential sensitivities to light and temperature, and CRY is a key component for the preferential entrainment to light.
Collapse
Affiliation(s)
- Taishi Yoshii
- Institute of Zoology, University of Regensburg, Regensburg, Germany, Biozentrum, University of Würzburg, Würzburg, Germany,
| | | | - Charlotte Helfrich-Förster
- Institute of Zoology, University of Regensburg, Regensburg, Germany, Biozentrum, University of Würzburg, Würzburg, Germany
| |
Collapse
|