1
|
Abou Daya F, Mandigo T, Patel D, Math S, Ober L, Maher M, Melkani G, Walker J, Saxena R. Drosophila Modeling Identifies Increased Sleep as a Link Between Insomnia and Cardiovascular Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647668. [PMID: 40291700 PMCID: PMC12026989 DOI: 10.1101/2025.04.07.647668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Insomnia is a common sleep disorder associated with negative long-term health outcomes. Mendelian randomization studies have found that insomnia significantly increases the risk of cardiovascular disease (CVD). To better understand the link between sleep and heart health, we identify genes associated with both insomnia and CVD. We model the disruption of the Drosophila melanogaster orthologs in neurons and cardiac tissue to characterize their cell-autonomous and non-cell-autonomous role in regulating sleep and cardiac physiology. We identify three genes that function in neurons and the heart to cell-autonomously regulate the function of each tissue. We find that the disruption of insomnia- and CVD-associated Drosophila orthologs in the heart most often lead to increased nighttime sleep. Inversely, disruptions in neurons that lead to increased sleep most often result in an elevated heart rate. To confirm the association between increased sleep and cardiac function, we performed a genetic correlation analysis from human data between long sleep-related traits and adverse cardiac outcomes. Significant correlations were found between most long sleep traits and heart failure, coronary artery disease, or myocardial infarction, reinforcing our findings in the fly linking increased or excessive sleep and altered cardiac health.
Collapse
|
2
|
Neuropeptide diuretic hormone 31 mediates memory and sleep via distinct neural pathways in Drosophila. Neurosci Res 2023:S0168-0102(23)00037-8. [PMID: 36780946 DOI: 10.1016/j.neures.2023.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/13/2023]
Abstract
Memory formation and sleep regulation are critical for brain functions in animals from invertebrates to humans. Neuropeptides play a pivotal role in regulating physiological behaviors, including memory formation and sleep. However, the detailed mechanisms by which neuropeptides regulate these physiological behaviors remains unclear. Herein, we report that neuropeptide diuretic hormone 31 (DH31) positively regulates memory formation and sleep in Drosophila melanogaster. The expression of DH31 in the dorsal and ventral fan-shaped body (dFB and vFB) neurons of the central complex and ventral lateral clock neurons (LNvs) in the brain was responsive to sleep regulation. In addition, the expression of membrane-tethered DH31 in dFB neurons rescued sleep defects in Dh31 mutants, suggesting that DH31 secreted from dFB, vFB, and LNvs acts on the DH31 receptor in the dFB to regulate sleep partly in an autoregulatory feedback loop. Moreover, the expression of DH31 in octopaminergic neurons, but not in the dFB neurons, is involved in forming intermediate-term memory. Our results suggest that DH31 regulates memory formation and sleep through distinct neural pathways.
Collapse
|
3
|
Riva S, Ispizua JI, Breide MT, Polcowñuk S, Lobera JR, Ceriani MF, Risau-Gusman S, Franco DL. Mating disrupts morning anticipation in Drosophila melanogaster females. PLoS Genet 2022; 18:e1010258. [PMID: 36548223 PMCID: PMC9779042 DOI: 10.1371/journal.pgen.1010258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 11/04/2022] [Indexed: 12/24/2022] Open
Abstract
After mating, the physiology of Drosophila females undergo several important changes, some of which are reflected in their rest-activity cycles. To explore the hypothesis that mating modifies the temporal organization of locomotor activity patterns, we recorded fly activity by a video tracking method. Monitoring rest-activity patterns under light/dark (LD) cycles indicated that mated females lose their ability to anticipate the night-day transition, in stark contrast to males and virgins. This postmating response is mediated by the activation of the sex peptide receptor (SPR) mainly on pickpocket (ppk) expressing neurons, since reducing expression of this receptor in these neurons restores the ability to anticipate the LD transition in mated females. Furthermore, we provide evidence of connectivity between ppk+ neurons and the pigment-dispersing factor (PDF)-positive ventral lateral neurons (sLNv), which play a central role in the temporal organization of daily activity. Since PDF has been associated to the generation of the morning activity peak, we hypothesized that the mating signal could modulate PDF levels. Indeed, we confirm that mated females have reduced PDF levels at the dorsal protocerebrum; moreover, SPR downregulation in ppk+ neurons mimics PDF levels observed in males. In sum, our results are consistent with a model whereby mating-triggered signals reach clock neurons in the fly central nervous system to modulate the temporal organization of circadian behavior according to the needs of the new status.
Collapse
Affiliation(s)
- Sabrina Riva
- Medical Physics Department, Bariloche Atomic Center, Comisión Nacional de Energía Atómica (CNEA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
| | - Juan Ignacio Ispizua
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir—IIBBA—CONICET, Buenos Aires, Argentina
| | - María Trinidad Breide
- Medical Physics Department, Bariloche Atomic Center, Comisión Nacional de Energía Atómica (CNEA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
| | - Sofía Polcowñuk
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir—IIBBA—CONICET, Buenos Aires, Argentina
| | - José Ricardo Lobera
- Medical Physics Department, Bariloche Atomic Center, Comisión Nacional de Energía Atómica (CNEA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
| | - María Fernanda Ceriani
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir—IIBBA—CONICET, Buenos Aires, Argentina
| | - Sebastian Risau-Gusman
- Medical Physics Department, Bariloche Atomic Center, Comisión Nacional de Energía Atómica (CNEA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
- * E-mail: (SRG); (DLF)
| | - Diana Lorena Franco
- Medical Physics Department, Bariloche Atomic Center, Comisión Nacional de Energía Atómica (CNEA) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
- * E-mail: (SRG); (DLF)
| |
Collapse
|
4
|
Discovery of levodopa-induced dyskinesia-associated genes using genomic studies in patients and Drosophila behavioral analyses. Commun Biol 2022; 5:872. [PMID: 36008531 PMCID: PMC9411113 DOI: 10.1038/s42003-022-03830-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 08/11/2022] [Indexed: 11/11/2022] Open
Abstract
Although levodopa is the most effective medication for Parkinson’s disease, long-term levodopa treatment is largely compromised due to late motor complications, including levodopa-induced dyskinesia (LID). However, the genetic basis of LID pathogenesis has not been fully understood. Here, we discover genes pathogenic for LID using Drosophila genetics and behavioral analyses combined with genome-wide association studies on 578 patients clinically diagnosed with LID. Similar to the therapeutic effect of levodopa in patients, acute levodopa treatments restore the motor defect of Parkinson’s disease model flies, while prolonged treatments cause LID-related symptoms, such as increased yawing, freezing and abrupt acceleration of locomotion. These symptoms require dopamine 1-like receptor 1 and are induced by neuronal overexpression of the receptor. Among genes selected from our analyses in the patient genome, neuronal knockdown of adenylyl cyclase 2 suppresses the levodopa-induced phenotypes and the receptor overexpression-induced symptoms in Drosophila. Together, our study provides genetic insights for LID pathogenesis through the D1-like receptor-adenylyl cyclase 2 signaling axis. A combined research approach using GWAS on Parkinson's disease patients and a Drosophila model of L-DOPA-induced dyskinesia (LID) reveals that LID is linked to ADCY2 signaling.
Collapse
|
5
|
Li W, Trigg JS, Taghert PH. Regulation of PDF receptor signaling controlling daily locomotor rhythms in Drosophila. PLoS Genet 2022; 18:e1010013. [PMID: 35605015 PMCID: PMC9166358 DOI: 10.1371/journal.pgen.1010013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/03/2022] [Accepted: 04/27/2022] [Indexed: 11/19/2022] Open
Abstract
Each day and in conjunction with ambient daylight conditions, neuropeptide PDF regulates the phase and amplitude of locomotor activity rhythms in Drosophila through its receptor, PDFR, a Family B G protein-coupled receptor (GPCR). We studied the in vivo process by which PDFR signaling turns off, by converting as many as half of the 28 potential sites of phosphorylation in its C terminal tail to a non-phosphorylatable residue (alanine). We report that many such sites are conserved evolutionarily, and their conversion creates a specific behavioral syndrome opposite to loss-of-function phenotypes previously described for pdfr. That syndrome includes increases in the amplitudes of both Morning and Evening behavioral peaks, as well as multi-hour delays of the Evening phase. The precise behavioral effects were dependent on day-length, and most effects mapped to conversion of only a few, specific serine residues near the very end of the protein and specific to its A isoform. Behavioral phase delays of the Evening activity under entraining conditions predicted the phase of activity cycles under constant darkness. The behavioral phenotypes produced by the most severe PDFR variant were ligand-dependent in vivo, and not a consequence of changes to their pharmacological properties, nor of changes in their surface expression, as measured in vitro. The mechanisms underlying termination of PDFR signaling are complex, subject to regulation that is modified by season, and central to a better understanding of the peptidergic modulation of behavior. In multi-cellular organisms, circadian pacemakers create output as a series of phase markers across the 24 hour day to allow other cells to pattern diverse aspects of daily rhythmic physiology and behavior. Within circadian pacemaker circuits, neuropeptide signaling is essential to help promote coherent circadian outputs. In the fruit fly Drosophila 150 neurons are dedicated circadian clocks and they all tell the same time. In spite of such strong synchronization, they provide diverse phasic outputs in the form of their discrete, asynchronous neuronal activity patterns. Neuropeptide signaling breaks the clock-generated symmetry and drives many pacemakers away from their preferred activity period in the morning. Each day, neuropeptide PDF is released by Morning pacemakers and delays the phase of activity of specific other pacemakers to later parts of the day or night. When and how the PDF that is released in the morning stops acting is unknown. Furthermore, timing of signal termination is not fixed because day length changes each day, hence the modulatory delay exerted by PDF must itself be regulated. Here we test a canonical model of G protein-coupled receptor physiology to ask how PDF receptor signaling is normally de-activated. We use behavioral measures to define sequence elements of the receptor whose post-translational modifications (e.g., phosphorylation) may define the duration of receptor signaling.
Collapse
Affiliation(s)
- Weihua Li
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Jennifer S. Trigg
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Paul H. Taghert
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
6
|
Flyer-Adams JG, Rivera-Rodriguez EJ, Yu J, Mardovin JD, Reed ML, Griffith LC. Regulation of Olfactory Associative Memory by the Circadian Clock Output Signal Pigment-Dispersing Factor (PDF). J Neurosci 2020; 40:9066-9077. [PMID: 33106351 PMCID: PMC7673005 DOI: 10.1523/jneurosci.0782-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 08/10/2020] [Accepted: 09/07/2020] [Indexed: 01/19/2023] Open
Abstract
Dissociation between the output of the circadian clock and external environmental cues is a major cause of human cognitive dysfunction. While the effects of ablation of the molecular clock on memory have been studied in many systems, little has been done to test the role of specific clock circuit output signals. To address this gap, we examined the effects of mutations of Pigment-dispersing factor (Pdf) and its receptor, Pdfr, on associative memory in male and female Drosophila Loss of PDF signaling significantly decreases the ability to form associative memory. Appetitive short-term memory (STM), which in wild-type (WT) is time-of-day (TOD) independent, is decreased across the day by mutation of Pdf or Pdfr, but more substantially in the morning than in the evening. This defect is because of PDFR expression in adult neurons outside the core clock circuit and the mushroom body (MB) Kenyon cells (KCs). The acquisition of a TOD difference in mutants implies the existence of multiple oscillators that act to normalize memory formation across the day for appetitive processes. Interestingly, aversive STM requires PDF but not PDFR, suggesting that there are valence-specific pathways downstream of PDF that regulate memory formation. These data argue that the circadian clock uses circuit-specific and molecularly diverse output pathways to enhance the ability of animals to optimize responses to changing conditions.SIGNIFICANCE STATEMENT From humans to invertebrates, cognitive processes are influenced by organisms' internal circadian clocks, the pace of which is linked to the solar cycle. Disruption of this link is increasingly common (e.g., jetlag, social jetlag disorders) and causes cognitive impairments that are costly and long lasting. A detailed understanding of how the internal clock regulates cognition is critical for the development of therapeutic methods. Here, we show for the first time that olfactory associative memory in Drosophila requires signaling by Pigment-dispersing factor (PDF), a neuromodulatory signaling peptide produced only by circadian clock circuit neurons. We also find a novel role for the clock circuit in stabilizing appetitive sucrose/odor memory across the day.
Collapse
Affiliation(s)
- Johanna G Flyer-Adams
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454-9110
| | - Emmanuel J Rivera-Rodriguez
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454-9110
| | - Junwei Yu
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454-9110
| | - Jacob D Mardovin
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454-9110
| | - Martha L Reed
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454-9110
| | - Leslie C Griffith
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454-9110
| |
Collapse
|
7
|
Coupling Neuropeptide Levels to Structural Plasticity in Drosophila Clock Neurons. Curr Biol 2020; 30:3154-3166.e4. [PMID: 32619484 DOI: 10.1016/j.cub.2020.06.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/05/2020] [Accepted: 06/02/2020] [Indexed: 12/24/2022]
Abstract
We have previously reported that pigment dispersing factor (PDF) neurons, which are essential in the control of rest-activity cycles in Drosophila, undergo circadian remodeling of their axonal projections, a phenomenon called circadian structural plasticity. Axonal arborizations display higher complexity during the day and become simpler at night, and this remodeling involves changes in the degree of connectivity. This phenomenon depends on the clock present within the ventrolateral neurons (LNvs) as well as in glia. In this work, we characterize in detail the contribution of the PDF neuropeptide to structural plasticity at different times across the day. Using diverse genetic strategies to temporally restrict its downregulation, we demonstrate that even subtle alterations to PDF cycling at the dorsal protocerebrum correlate with impaired remodeling, underscoring its relevance for the characteristic morning spread; PDF released from the small LNvs (sLNvs) and the large LNvs (lLNvs) contribute to the process. Moreover, forced depolarization recruits activity-dependent mechanisms to mediate growth only at night, overcoming the restriction imposed by the clock on membrane excitability. Interestingly, the active process of terminal remodeling requires PDF receptor (PDFR) signaling acting locally through the cyclic-nucleotide-gated channel ion channel subunit A (CNGA). Thus, clock-dependent PDF signaling shapes the connectivity of these essential clock neurons on daily basis.
Collapse
|
8
|
Raphael KA, Sved JA, Pearce S, Oakeshott JG, Gilchrist AS, Sherwin WB, Frommer M. Differences in gene regulation in a tephritid model of prezygotic reproductive isolation. INSECT MOLECULAR BIOLOGY 2019; 28:689-702. [PMID: 30955213 DOI: 10.1111/imb.12583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The two tephritid fruit fly pests, Bactrocera tryoni and Bactrocera neohumeralis, are unusually well suited to the study of the genetics of reproductive isolating mechanisms. Sequence difference between the species is no greater than between a pair of conspecific Drosophila melanogaster populations. The two species exist in close sympatry, yet do not hybridize in the field, apparently kept separate by a strong premating isolation mechanism involving the time of day at which mating occurs. This spurred us to search for key genes for which time of day expression is regulated differently between the species. Using replicated, quantitative transcriptomes from head tissues of males of the two species, sampled in the day and night, we identified 141 transcripts whose abundance showed a significant interaction between species and time of day, indicating a difference in gene regulation. The brain transcripts showing this interaction were enriched for genes with a neurone function and 90% of these were more abundant at night than day in B. tryoni. Features of the expression patterns suggest that there may be a difference in the regulation of sleep-wake cycles between the species. In particular several genes, which in D. melanogaster are expressed in circadian pacemaker cells, are promising candidates to further explore the genetic differentiation involved in this prezygotic reproductive isolation mechanism.
Collapse
Affiliation(s)
- K A Raphael
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - J A Sved
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - S Pearce
- CSIRO Land & Water Flagship, Canberra, ACT, Australia
| | - J G Oakeshott
- CSIRO Land & Water Flagship, Canberra, ACT, Australia
| | - A S Gilchrist
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - W B Sherwin
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - M Frommer
- Evolution and Ecology Research Centre, School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
9
|
Schlichting M, Weidner P, Diaz M, Menegazzi P, Dalla Benetta E, Helfrich-Förster C, Rosbash M. Light-Mediated Circuit Switching in the Drosophila Neuronal Clock Network. Curr Biol 2019; 29:3266-3276.e3. [PMID: 31564496 DOI: 10.1016/j.cub.2019.08.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/28/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022]
Abstract
The circadian clock is a timekeeper but also helps adapt physiology to the outside world. This is because an essential feature of clocks is their ability to adjust (entrain) to the environment, with light being the most important signal. Whereas cryptochrome-mediated entrainment is well understood in Drosophila, integration of light information via the visual system lacks a neuronal or molecular mechanism. Here, we show that a single photoreceptor subtype is essential for long-day adaptation. These cells activate key circadian neurons, namely the large ventral-lateral neurons (lLNvs), which release the neuropeptide pigment-dispersing factor (PDF). RNAi and rescue experiments show that PDF from these cells is necessary and sufficient for delaying the timing of the evening (E) activity in long-day conditions. This contrasts to PDF that derives from the small ventral-lateral neurons (sLNvs), which are essential for constant darkness (DD) rhythmicity. Using a cell-specific CRISPR/Cas9 assay, we show that lLNv-derived PDF directly interacts with neurons important for E activity timing. Interestingly, this pathway is specific for long-day adaptation and appears to be dispensable in equinox or DD conditions. The results therefore indicate that external cues cause a rearrangement of neuronal hierarchy, which contributes to behavioral plasticity.
Collapse
Affiliation(s)
- Matthias Schlichting
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA.
| | - Patrick Weidner
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA; Department for Neurobiology and Genetics, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Madelen Diaz
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Pamela Menegazzi
- Department for Neurobiology and Genetics, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Elena Dalla Benetta
- Department for Neurobiology and Genetics, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | | | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| |
Collapse
|
10
|
Nagy D, Cusumano P, Andreatta G, Anduaga AM, Hermann-Luibl C, Reinhard N, Gesto J, Wegener C, Mazzotta G, Rosato E, Kyriacou CP, Helfrich-Förster C, Costa R. Peptidergic signaling from clock neurons regulates reproductive dormancy in Drosophila melanogaster. PLoS Genet 2019; 15:e1008158. [PMID: 31194738 PMCID: PMC6592559 DOI: 10.1371/journal.pgen.1008158] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/25/2019] [Accepted: 04/25/2019] [Indexed: 11/18/2022] Open
Abstract
With the approach of winter, many insects switch to an alternative protective developmental program called diapause. Drosophila melanogaster females overwinter as adults by inducing a reproductive arrest that is characterized by inhibition of ovarian development at previtellogenic stages. The insulin producing cells (IPCs) are key regulators of this process, since they produce and release insulin-like peptides that act as diapause-antagonizing hormones. Here we show that in D. melanogaster two neuropeptides, Pigment Dispersing Factor (PDF) and short Neuropeptide F (sNPF) inhibit reproductive arrest, likely through modulation of the IPCs. In particular, genetic manipulations of the PDF-expressing neurons, which include the sNPF-producing small ventral Lateral Neurons (s-LNvs), modulated the levels of reproductive dormancy, suggesting the involvement of both neuropeptides. We expressed a genetically encoded cAMP sensor in the IPCs and challenged brain explants with synthetic PDF and sNPF. Bath applications of both neuropeptides increased cAMP levels in the IPCs, even more so when they were applied together, suggesting a synergistic effect. Bath application of sNPF additionally increased Ca2+ levels in the IPCs. Our results indicate that PDF and sNPF inhibit reproductive dormancy by maintaining the IPCs in an active state.
Collapse
Affiliation(s)
- Dóra Nagy
- Department of Biology, University of Padova, Padova, Italy
| | - Paola Cusumano
- Department of Biology, University of Padova, Padova, Italy
| | | | - Ane Martin Anduaga
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christiane Hermann-Luibl
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Nils Reinhard
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - João Gesto
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christian Wegener
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | | | - Ezio Rosato
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Charalambos P. Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Rodolfo Costa
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
11
|
A Symphony of Signals: Intercellular and Intracellular Signaling Mechanisms Underlying Circadian Timekeeping in Mice and Flies. Int J Mol Sci 2019; 20:ijms20092363. [PMID: 31086044 PMCID: PMC6540063 DOI: 10.3390/ijms20092363] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 12/11/2022] Open
Abstract
The central pacemakers of circadian timekeeping systems are highly robust yet adaptable, providing the temporal coordination of rhythms in behavior and physiological processes in accordance with the demands imposed by environmental cycles. These features of the central pacemaker are achieved by a multi-oscillator network in which individual cellular oscillators are tightly coupled to the environmental day-night cycle, and to one another via intercellular coupling. In this review, we will summarize the roles of various neurotransmitters and neuropeptides in the regulation of circadian entrainment and synchrony within the mammalian and Drosophila central pacemakers. We will also describe the diverse functions of protein kinases in the relay of input signals to the core oscillator or the direct regulation of the molecular clock machinery.
Collapse
|
12
|
Song Q, Feng G, Zhang J, Xia X, Ji M, Lv L, Ping Y. NMDA Receptor-mediated Ca2+ Influx in the Absence of Mg2+ Block Disrupts Rest: Activity Rhythms in Drosophila. Sleep 2018; 40:4330652. [PMID: 29029290 DOI: 10.1093/sleep/zsx166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Study Objectives The correlated activation of pre- and postsynaptic neurons is essential for the NMDA receptor-mediated Ca2+ influx by removing Mg2+ from block site and NMDA receptors have been implicated in phase resetting of circadian clocks. So we assessed rest:activity rhythms in Mg2+ block defective animals. Methods Using Drosophila locomotor monitoring system, we checked circadian rest:activity rhythms of different mutants under constant darkness (DD) and light:dark (LD) conditions. We recorded NMDA receptor-mediated currents or Ca2+ increase in neurons using patch-clamp and Ca2+ imaging techniques. Results We found that Mg2+ block defective mutant flies were completely arrhythmic under DD. To further understand the role of Mg2+ block in daily circadian rest:activity, we observed the mutant files under LD cycles, and we found severely reduced morning anticipation and advanced evening peak compared to control flies. We also used tissue-specific expression of Mg2+ block defective NMDA receptors and demonstrated pigment-dispersing factor receptor (PDFR)-expressing circadian neurons were implicated in mediating the circadian rest:activity deficits. Endogenous functional NMDA receptors are expressed in most Drosophila neurons, including in a subgroup of dorsal neurons (DN1s). Subsequently, we determined that the uncorrelated extra Ca2+ influx may act in part through Ca2+/Calmodulin (CaM)-stimulated PDE1c pathway leading to morning behavior phenotypes. Conclusions These results demonstrate that Mg2+ block of NMDA receptors at resting potential is essential for the daily circadian rest:activity rhythms and we propose that Mg2+ block functions to suppress CaM-stimulated PDE1c activation at resting potential, thus regulating Ca2+ and cyclic AMP oscillations in circadian and sleep circuits.
Collapse
Affiliation(s)
- Qian Song
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Ge Feng
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaxing Zhang
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Xuechun Xia
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Min Ji
- Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Lei Lv
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yong Ping
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders (No.13dz2260500), Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
13
|
Wakefulness Is Promoted during Day Time by PDFR Signalling to Dopaminergic Neurons in Drosophila melanogaster. eNeuro 2018; 5:eN-NWR-0129-18. [PMID: 30131970 PMCID: PMC6102377 DOI: 10.1523/eneuro.0129-18.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/25/2018] [Accepted: 06/18/2018] [Indexed: 11/27/2022] Open
Abstract
Circadian clocks modulate timing of sleep/wake cycles in animals; however, the underlying mechanisms remain poorly understood. In Drosophila melanogaster, large ventral lateral neurons (l-LNv) are known to promote wakefulness through the action of the neuropeptide pigment dispersing factor (PDF), but the downstream targets of PDF signalling remain elusive. In a screen using downregulation or overexpression (OEX) of the gene encoding PDF receptor (pdfr), we found that a subset of dopaminergic neurons responds to PDF to promote wakefulness during the day. Moreover, we found that small LNv (s-LNv) and dopaminergic neurons form synaptic contacts, and PDFR signalling inhibited dopaminergic neurons specifically during day time. We propose that these dopaminergic neurons that respond to PDFR signalling are sleep-promoting and that during the day when PDF levels are high, they are inhibited, thereby promoting wakefulness. Thus, we identify a novel circadian clock pathway that mediates wake promotion specifically during day time.
Collapse
|
14
|
Franco DL, Frenkel L, Ceriani MF. The Underlying Genetics of Drosophila Circadian Behaviors. Physiology (Bethesda) 2018; 33:50-62. [PMID: 29212892 DOI: 10.1152/physiol.00020.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/20/2017] [Accepted: 09/25/2017] [Indexed: 01/22/2023] Open
Abstract
Life is shaped by circadian clocks. This review focuses on how behavioral genetics in the fruit fly unveiled what is known today about circadian physiology. We will briefly summarize basic properties of the clock and focus on some clock-controlled behaviors to highlight how communication between central and peripheral oscillators defines their properties.
Collapse
Affiliation(s)
- D Lorena Franco
- Departamento de Física Médica, Centro Atómico Bariloche and Instituto Balseiro, CONICET, San Carlos de Bariloche, Río Negro, Argentina; and
| | - Lia Frenkel
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir (FIL)-Instituto de Investigaciones Bioquímicas-IIBBA-CONICET, Buenos Aires, Argentina
| | - M Fernanda Ceriani
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir (FIL)-Instituto de Investigaciones Bioquímicas-IIBBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
15
|
Calcium and cAMP directly modulate the speed of the Drosophila circadian clock. PLoS Genet 2018; 14:e1007433. [PMID: 29879123 PMCID: PMC6007936 DOI: 10.1371/journal.pgen.1007433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/19/2018] [Accepted: 05/18/2018] [Indexed: 01/08/2023] Open
Abstract
Circadian clocks impose daily periodicities to animal behavior and physiology. At their core, circadian rhythms are produced by intracellular transcriptional/translational feedback loops (TTFL). TTFLs may be altered by extracellular signals whose actions are mediated intracellularly by calcium and cAMP. In mammals these messengers act directly on TTFLs via the calcium/cAMP-dependent transcription factor, CREB. In the fruit fly, Drosophila melanogaster, calcium and cAMP also regulate the periodicity of circadian locomotor activity rhythmicity, but whether this is due to direct actions on the TTFLs themselves or are a consequence of changes induced to the complex interrelationship between different classes of central pacemaker neurons is unclear. Here we investigated this question focusing on the peripheral clock housed in the non-neuronal prothoracic gland (PG), which, together with the central pacemaker in the brain, controls the timing of adult emergence. We show that genetic manipulations that increased and decreased the levels of calcium and cAMP in the PG caused, respectively, a shortening and a lengthening of the periodicity of emergence. Importantly, knockdown of CREB in the PG caused an arrhythmic pattern of eclosion. Interestingly, the same manipulations directed at central pacemaker neurons caused arrhythmicity of eclosion and of adult locomotor activity, suggesting a common mechanism. Our results reveal that the calcium and cAMP pathways can alter the functioning of the clock itself. In the PG, these messengers, acting as outputs of the clock or as second messengers for stimuli external to the PG, could also contribute to the circadian gating of adult emergence. Circadian clocks impose daily periodicities to animal behavior and physiology. At their core, circadian rhythms are produced by intracellular transcriptional/translational feedback loops (TTFL). TTFLs may be altered by extracellular signals whose actions are mediated intracellularly by calcium and cAMP. In Drosophila, calcium and cAMP levels affect the periodicity of Drosophila circadian rhythms, but whether this is due to direct actions on the TTFLs themselves or is a consequence of changes induced to the complex interrelationship between different classes of central pacemaker neurons is unclear. Here we used the non-neuronal circadian clock located in the prothoracic gland (PG) to show that these messengers affect the speed of the circadian clock that controls the timing of adult emergence and suggest that these actions are mediated by CREB. Importantly, since calcium and cAMP are also output signals of the clock, they may contribute to the mechanism that imposes a circadian gating to the timing of adult emergence.
Collapse
|
16
|
Drosophila DH31 Neuropeptide and PDF Receptor Regulate Night-Onset Temperature Preference. J Neurosci 2017; 36:11739-11754. [PMID: 27852781 DOI: 10.1523/jneurosci.0964-16.2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 08/24/2016] [Accepted: 09/22/2016] [Indexed: 12/25/2022] Open
Abstract
Body temperature exhibits rhythmic fluctuations over a 24 h period (Refinetti and Menaker, 1992) and decreases during the night, which is associated with sleep initiation (Gilbert et al., 2004; Kräuchi, 2007a,b). However, the underlying mechanism of this temperature decrease is largely unknown. We have previously shown that Drosophila exhibit a daily temperature preference rhythm (TPR), in which their preferred temperatures increase during the daytime and then decrease at the transition from day to night (night-onset) (Kaneko et al., 2012). Because Drosophila are small ectotherms, their body temperature is very close to that of the ambient temperature (Stevenson, 1985), suggesting that their TPR generates their body temperature rhythm. Here, we demonstrate that the neuropeptide diuretic hormone 31 (DH31) and pigment-dispersing factor receptor (PDFR) contribute to regulate the preferred temperature decrease at night-onset. We show that PDFR and tethered-DH31 expression in dorsal neurons 2 (DN2s) restore the preferred temperature decrease at night-onset, suggesting that DH31 acts on PDFR in DN2s. Notably, we previously showed that the molecular clock in DN2s is important for TPR. Although PDF (another ligand of PDFR) is a critical factor for locomotor activity rhythms, Pdf mutants exhibit normal preferred temperature decreases at night-onset. This suggests that DH31-PDFR signaling specifically regulates a preferred temperature decrease at night-onset. Thus, we propose that night-onset TPR and locomotor activity rhythms are differentially controlled not only by clock neurons but also by neuropeptide signaling in the brain. SIGNIFICANCE STATEMENT Body temperature rhythm (BTR) is fundamental for the maintenance of functions essential for homeostasis, such as generating metabolic energy and sleep. One major unsolved question is how body temperature decreases dramatically during the night. Previously, we demonstrated that a BTR-like mechanism, referred to as temperature preference rhythm (TPR), exists in Drosophila Here, we demonstrate that the diuretic hormone 31 (DH31) neuropeptide and pigment-dispersing factor receptor (PDFR) regulate preferred temperature decreases at night-onset via dorsal neurons 2. This is the first in vivo evidence that DH31 could function as a ligand of PDFR. Although both DH31 and PDF are ligands of PDFR, we show that DH31 regulates night-onset TPR, but PDF does not, suggesting that night-onset TPR and locomotor activity rhythms are controlled by different neuropeptides via different clock cells.
Collapse
|
17
|
Moose DL, Haase SJ, Aldrich BT, Lear BC. The Narrow Abdomen Ion Channel Complex Is Highly Stable and Persists from Development into Adult Stages to Promote Behavioral Rhythmicity. Front Cell Neurosci 2017. [PMID: 28634443 PMCID: PMC5459923 DOI: 10.3389/fncel.2017.00159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The sodium leak channel NARROW ABDOMEN (NA)/ NALCN is an important component of circadian pacemaker neuronal output. In Drosophila, rhythmic expression of the NA channel regulator Nlf-1 in a subset of adult pacemaker neurons has been proposed to contribute to circadian regulation of channel localization or activity. Here we have restricted expression of Drosophila NA channel subunits or the Nlf-1 regulator to either development or adulthood using the temperature-inducible tubulin-GAL80ts system. Surprisingly, we find that developmental expression of endogenous channel subunits and Nlf-1 is sufficient to promote robust rhythmic behavior in adults. Moreover, we find that channel complex proteins produced during development persist in the Drosophila head with little decay for at least 5-7 days in adults. In contrast, restricting either endogenous or transgenic gene expression to adult stages produces only limited amounts of the functional channel complex. These data indicate that much of the NA channel complex that functions in adult circadian neurons is normally produced during development, and that the channel complex is very stable in most neurons in the Drosophila brain. Based on these findings, we propose that circadian regulation of NA channel function in adult pacemaker neurons is mediated primarily by post-translational mechanisms that are independent of Nlf-1.
Collapse
Affiliation(s)
- Devon L Moose
- Department of Biology, University of Iowa, Iowa CityIA, United States
| | - Stephanie J Haase
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa CityIA, United States
| | | | - Bridget C Lear
- Department of Biology, University of Iowa, Iowa CityIA, United States.,Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa CityIA, United States
| |
Collapse
|
18
|
Allen CN, Nitabach MN, Colwell CS. Membrane Currents, Gene Expression, and Circadian Clocks. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027714. [PMID: 28246182 DOI: 10.1101/cshperspect.a027714] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neuronal circadian oscillators in the mammalian and Drosophila brain express a circadian clock comprised of interlocking gene transcription feedback loops. The genetic clock regulates the membrane electrical activity by poorly understood signaling pathways to generate a circadian pattern of action potential firing. During the day, Na+ channels contribute an excitatory drive for the spontaneous activity of circadian clock neurons. Multiple types of K+ channels regulate the action potential firing pattern and the nightly reduction in neuronal activity. The membrane electrical activity possibly signaling by changes in intracellular Ca2+ and cyclic adenosine monophosphate (cAMP) regulates the activity of the gene clock. A decline in the signaling pathways that link the gene clock and neural activity during aging and disease may weaken the circadian output and generate significant impacts on human health.
Collapse
Affiliation(s)
- Charles N Allen
- Oregon Institute of Occupational Health Sciences and Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Michael N Nitabach
- Department of Cellular and Molecular Physiology and Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06520
| | - Christopher S Colwell
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, California 90024
| |
Collapse
|
19
|
Castro L, Yapo C, Vincent P. [Physiopathology of cAMP/PKA signaling in neurons]. Biol Aujourdhui 2017; 210:191-203. [PMID: 28327278 DOI: 10.1051/jbio/2017005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Indexed: 11/15/2022]
Abstract
Cyclic adenosine monophosphate (cAMP) and the cyclic-AMP dependent protein kinase (PKA) regulate a plethora of cellular functions in virtually all eukaryotic cells. In neurons, the cAMP/PKA signaling cascade controls a number of biological properties such as axonal growth, synaptic transmission, regulation of excitability or long term changes in the nucleus. Genetically-encoded optical biosensors for cAMP or PKA considerably improved our understanding of these processes by providing a real-time measurement in living neurons. In this review, we describe the recent progresses made in the creation of biosensors for cAMP or PKA activity. These biosensors revealed profound differences in the amplitude of the cAMP signal evoked by neuromodulators between various neuronal preparations. These responses can be resolved at the level of individual neurons, also revealing differences related to the neuronal type. At the subcellular level, biosensors reported different signal dynamics in domains like dendrites, cell body, nucleus and axon. Combining this imaging approach with pharmacology or genetical models points at phosphodiesterases and phosphatases as critical regulatory proteins. Biosensor imaging will certainly help understand the mechanism of action of current drugs as well as help in devising novel therapeutic strategies for neuropsychiatric diseases.
Collapse
|
20
|
Fluorescence circadian imaging reveals a PDF-dependent transcriptional regulation of the Drosophila molecular clock. Sci Rep 2017; 7:41560. [PMID: 28134281 PMCID: PMC5278502 DOI: 10.1038/srep41560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/21/2016] [Indexed: 01/17/2023] Open
Abstract
Circadian locomotor behaviour is controlled by a pacemaker circuit composed of clock-containing neurons. To interrogate the mechanistic relationship between the molecular clockwork and network communication critical to the operation of the Drosophila circadian pacemaker circuit, we established new fluorescent circadian reporters that permit single-cell recording of transcriptional and post-transcriptional rhythms in brain explants and cultured neurons. Live-imaging experiments combined with pharmacological and genetic manipulations demonstrate that the neuropeptide pigment-dispersing factor (PDF) amplifies the molecular rhythms via time-of-day- and activity-dependent upregulation of transcription from E-box-containing clock gene promoters within key pacemaker neurons. The effect of PDF on clock gene transcription and the known role of PDF in enhancing PER/TIM stability occur via independent pathways downstream of the PDF receptor, the former through a cAMP-independent mechanism and the latter through a cAMP-PKA dependent mechanism. These results confirm and extend the mechanistic understanding of the role of PDF in controlling the synchrony of the pacemaker neurons. More broadly, our results establish the utility of the new live-imaging tools for the study of molecular-neural interactions important for the operation of the circadian pacemaker circuit.
Collapse
|
21
|
Dissel S, Klose M, Donlea J, Cao L, English D, Winsky-Sommerer R, van Swinderen B, Shaw PJ. Enhanced sleep reverses memory deficits and underlying pathology in Drosophila models of Alzheimer's disease. Neurobiol Sleep Circadian Rhythms 2016; 2:15-26. [PMID: 29094110 PMCID: PMC5662006 DOI: 10.1016/j.nbscr.2016.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
To test the hypothesis that sleep can reverse cognitive impairment during Alzheimer's disease, we enhanced sleep in flies either co-expressing human amyloid precursor protein and Beta-secretase (APP:BACE), or in flies expressing human tau. The ubiquitous expression of APP:BACE or human tau disrupted sleep. The sleep deficits could be reversed and sleep could be enhanced when flies were administered the GABA-A agonist 4,5,6,7-tetrahydroisoxazolo-[5,4-c]pyridine-3-ol (THIP). Expressing APP:BACE disrupted both Short-term memory (STM) and Long-term memory (LTM) as assessed using Aversive Phototaxic Suppression (APS) and courtship conditioning. Flies expressing APP:BACE also showed reduced levels of the synaptic protein discs large (DLG). Enhancing sleep in memory-impaired APP:BACE flies fully restored both STM and LTM and restored DLG levels. Sleep also restored STM to flies expressing human tau. Using live-brain imaging of individual clock neurons expressing both tau and the cAMP sensor Epac1-camps, we found that tau disrupted cAMP signaling. Importantly, enhancing sleep in flies expressing human tau restored proper cAMP signaling. Thus, we demonstrate that sleep can be used as a therapeutic to reverse deficits that accrue during the expression of toxic peptides associated with Alzheimer's disease. THIP can be used to enhance sleep in two Drosophila models of Alzheimer's disease. Enhanced sleep reverses memory deficits in fly's expressing human APP:BACE and tau. Enhanced sleep restores cAMP levels in clock neurons expressing tau. Sleep can be used as a therapeutic to reverse Alzheimer's disease related deficits.
Collapse
Affiliation(s)
- Stephane Dissel
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Markus Klose
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Jeff Donlea
- Department of Neurobiology, University of California: Los Angeles Los Angeles, California, U.S.A
| | - Lijuan Cao
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Denis English
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| | - Raphaelle Winsky-Sommerer
- Surrey Sleep Research Centre, Faculty of Health and Medical Sciences University of Surrey Guildford Surrey, GU2 7XH, United Kingdom
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, Brisbane Qld 4072 Australia
| | - Paul J Shaw
- Department of Neuroscience, Washington University in St. Louis, 660 S. Euclid Ave, St. Louis, Missouri, U.S.A
| |
Collapse
|
22
|
Maurer C, Winter T, Chen S, Hung HC, Weber F. The CREB-binding protein affects the circadian regulation of behaviour. FEBS Lett 2016; 590:3213-20. [DOI: 10.1002/1873-3468.12336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Christian Maurer
- Biochemistry Center Heidelberg; University of Heidelberg; Germany
| | - Tobias Winter
- Biochemistry Center Heidelberg; University of Heidelberg; Germany
| | - Siwei Chen
- Biochemistry Center Heidelberg; University of Heidelberg; Germany
| | - Hsiu-Cheng Hung
- Biochemistry Center Heidelberg; University of Heidelberg; Germany
| | - Frank Weber
- Biochemistry Center Heidelberg; University of Heidelberg; Germany
| |
Collapse
|
23
|
Klose M, Duvall L, Li W, Liang X, Ren C, Steinbach JH, Taghert PH. Functional PDF Signaling in the Drosophila Circadian Neural Circuit Is Gated by Ral A-Dependent Modulation. Neuron 2016; 90:781-794. [PMID: 27161526 DOI: 10.1016/j.neuron.2016.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/13/2016] [Accepted: 03/20/2016] [Indexed: 12/18/2022]
Abstract
The neuropeptide PDF promotes the normal sequencing of circadian behavioral rhythms in Drosophila, but its signaling mechanisms are not well understood. We report daily rhythmicity in responsiveness to PDF in critical pacemakers called small LNvs. There is a daily change in potency, as great as 10-fold higher, around dawn. The rhythm persists in constant darkness and does not require endogenous ligand (PDF) signaling or rhythmic receptor gene transcription. Furthermore, rhythmic responsiveness reflects the properties of the pacemaker cell type, not the receptor. Dopamine responsiveness also cycles, in phase with that of PDF, in the same pacemakers, but does not cycle in large LNv. The activity of RalA GTPase in s-LNv regulates PDF responsiveness and behavioral locomotor rhythms. Additionally, cell-autonomous PDF signaling reversed the circadian behavioral effects of lowered RalA activity. Thus, RalA activity confers high PDF responsiveness, providing a daily gate around the dawn hours to promote functional PDF signaling.
Collapse
Affiliation(s)
- Markus Klose
- Dept. of Neuroscience, Washington University Medical School, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Laura Duvall
- Dept. of Neuroscience, Washington University Medical School, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Weihua Li
- Dept. of Neuroscience, Washington University Medical School, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Xitong Liang
- Dept. of Neuroscience, Washington University Medical School, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Chi Ren
- Dept. of Neuroscience, Washington University Medical School, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Joe Henry Steinbach
- Dept. of Anesthesiology, Washington University Medical School, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Paul H Taghert
- Dept. of Neuroscience, Washington University Medical School, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| |
Collapse
|
24
|
Miazzi F, Hansson BS, Wicher D. Odor-induced cAMP production in Drosophila melanogaster olfactory sensory neurons. ACTA ACUST UNITED AC 2016; 219:1798-803. [PMID: 27045092 DOI: 10.1242/jeb.137901] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/30/2016] [Indexed: 12/22/2022]
Abstract
Insect odorant receptors are seven transmembrane domain proteins that form cation channels, whose functional properties such as receptor sensitivity are subject to regulation by intracellular signaling cascades. Here, we used the cAMP fluorescent indicator Epac1-camps to investigate the occurrence of odor-induced cAMP production in olfactory sensory neurons (OSNs) of Drosophila melanogaster We show that stimulation of the receptor complex with an odor mixture or with the synthetic agonist VUAA1 induces a cAMP response. Moreover, we show that while the intracellular Ca(2+) concentration influences cAMP production, the OSN-specific receptor OrX is necessary to elicit cAMP responses in Ca(2+)-free conditions. These results provide direct evidence of a relationship between odorant receptor stimulation and cAMP production in olfactory sensory neurons in the fruit fly antenna and show that this method can be used to further investigate the role that this second messenger plays in insect olfaction.
Collapse
Affiliation(s)
- Fabio Miazzi
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knöll-Str. 8, Jena D-07745, Germany
| | - Bill S Hansson
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knöll-Str. 8, Jena D-07745, Germany
| | - Dieter Wicher
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knöll-Str. 8, Jena D-07745, Germany
| |
Collapse
|
25
|
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
|
26
|
Langenhan T, Barr MM, Bruchas MR, Ewer J, Griffith LC, Maiellaro I, Taghert PH, White BH, Monk KR. Model Organisms in G Protein-Coupled Receptor Research. Mol Pharmacol 2015; 88:596-603. [PMID: 25979002 PMCID: PMC4551050 DOI: 10.1124/mol.115.098764] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/14/2015] [Indexed: 12/19/2022] Open
Abstract
The study of G protein-coupled receptors (GPCRs) has benefited greatly from experimental approaches that interrogate their functions in controlled, artificial environments. Working in vitro, GPCR receptorologists discovered the basic biologic mechanisms by which GPCRs operate, including their eponymous capacity to couple to G proteins; their molecular makeup, including the famed serpentine transmembrane unit; and ultimately, their three-dimensional structure. Although the insights gained from working outside the native environments of GPCRs have allowed for the collection of low-noise data, such approaches cannot directly address a receptor's native (in vivo) functions. An in vivo approach can complement the rigor of in vitro approaches: as studied in model organisms, it imposes physiologic constraints on receptor action and thus allows investigators to deduce the most salient features of receptor function. Here, we briefly discuss specific examples in which model organisms have successfully contributed to the elucidation of signals controlled through GPCRs and other surface receptor systems. We list recent examples that have served either in the initial discovery of GPCR signaling concepts or in their fuller definition. Furthermore, we selectively highlight experimental advantages, shortcomings, and tools of each model organism.
Collapse
Affiliation(s)
- Tobias Langenhan
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Maureen M Barr
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Michael R Bruchas
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - John Ewer
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Leslie C Griffith
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Isabella Maiellaro
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Paul H Taghert
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Benjamin H White
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Kelly R Monk
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| |
Collapse
|
27
|
Krishnan HC, Lyons LC. Synchrony and desynchrony in circadian clocks: impacts on learning and memory. ACTA ACUST UNITED AC 2015; 22:426-37. [PMID: 26286653 PMCID: PMC4561405 DOI: 10.1101/lm.038877.115] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/29/2015] [Indexed: 12/11/2022]
Abstract
Circadian clocks evolved under conditions of environmental variation, primarily alternating light dark cycles, to enable organisms to anticipate daily environmental events and coordinate metabolic, physiological, and behavioral activities. However, modern lifestyle and advances in technology have increased the percentage of individuals working in phases misaligned with natural circadian activity rhythms. Endogenous circadian oscillators modulate alertness, the acquisition of learning, memory formation, and the recall of memory with examples of circadian modulation of memory observed across phyla from invertebrates to humans. Cognitive performance and memory are significantly diminished when occurring out of phase with natural circadian rhythms. Disruptions in circadian regulation can lead to impairment in the formation of memories and manifestation of other cognitive deficits. This review explores the types of interactions through which the circadian clock modulates cognition, highlights recent progress in identifying mechanistic interactions between the circadian system and the processes involved in memory formation, and outlines methods used to remediate circadian perturbations and reinforce circadian adaptation.
Collapse
Affiliation(s)
- Harini C Krishnan
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, Florida 32306, USA
| | - Lisa C Lyons
- Department of Biological Science, Program in Neuroscience, Florida State University, Tallahassee, Florida 32306, USA
| |
Collapse
|
28
|
Patel N, Gold MG. The genetically encoded tool set for investigating cAMP: more than the sum of its parts. Front Pharmacol 2015; 6:164. [PMID: 26300778 PMCID: PMC4526808 DOI: 10.3389/fphar.2015.00164] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/24/2015] [Indexed: 11/13/2022] Open
Abstract
Intracellular fluctuations of the second messenger cyclic AMP (cAMP) are regulated with spatial and temporal precision. This regulation is supported by the sophisticated arrangement of cyclases, phosphodiesterases, anchoring proteins, and receptors for cAMP. Discovery of these nuances to cAMP signaling has been facilitated by the development of genetically encodable tools for monitoring and manipulating cAMP and the proteins that support cAMP signaling. In this review, we discuss the state-of-the-art in development of different genetically encoded tools for sensing cAMP and the activity of its primary intracellular receptor protein kinase A (PKA). We introduce sequences for encoding adenylyl cyclases that enable cAMP levels to be artificially elevated within cells. We chart the evolution of sequences for selectively modifying protein-protein interactions that support cAMP signaling, and for driving cAMP sensors and manipulators to different subcellular locations. Importantly, these different genetically encoded tools can be applied synergistically, and we highlight notable instances that take advantage of this property. Finally, we consider prospects for extending the utility of the tool set to support further insights into the role of cAMP in health and disease.
Collapse
Affiliation(s)
- Neha Patel
- Department of Neuroscience, Physiology and Pharmacology, University College London London, UK
| | - Matthew G Gold
- Department of Neuroscience, Physiology and Pharmacology, University College London London, UK
| |
Collapse
|
29
|
Hermann-Luibl C, Helfrich-Förster C. Clock network in Drosophila. CURRENT OPINION IN INSECT SCIENCE 2015; 7:65-70. [PMID: 32846682 DOI: 10.1016/j.cois.2014.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 11/14/2014] [Accepted: 11/19/2014] [Indexed: 06/11/2023]
Abstract
The circadian clock consists of a network of peptidergic neurons in the brain of all animals. The function of this peptidergic network has been largely revealed in the fruit fly Drosophila melanogaster due to the relatively few well characterized clock neurons and because these neurons can be genetically manipulated. Here we review the neuronal organization of the circadian network and the role of individual clock neurons and neuropeptides in it.
Collapse
Affiliation(s)
- Christiane Hermann-Luibl
- Department of Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, Germany
| | - Charlotte Helfrich-Förster
- Department of Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, Germany.
| |
Collapse
|
30
|
Beckwith EJ, Ceriani MF. Experimental assessment of the network properties of the Drosophila circadian clock. J Comp Neurol 2015; 523:982-96. [PMID: 25504089 DOI: 10.1002/cne.23728] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 01/03/2023]
Abstract
Circadian rhythms are conserved across kingdoms and coordinate physiology and behavior for appropriate time-keeping. The neuronal populations that govern circadian rhythms are described in many animal models, and the current challenge is to understand how they interact to control overt rhythms, remaining plastic enough to respond and adapt to a changing environment. In Drosophila melanogaster, the circadian network comprises about 150 neurons, and the main synchronizer is the neuropeptide pigment-dispersing factor (PDF), released by the well-characterized central pacemaker neurons, the small ventral lateral neurons (sLNvs). However, the rules and properties governing the communication and coupling between this central pacemaker and downstream clusters are not fully elucidated. Here we genetically manipulate the speed of the molecular clock specifically in the central pacemaker neurons of Drosophila and provide experimental evidence of their restricted ability to synchronize downstream clusters. We also demonstrate that the sLNv-controlled clusters have an asymmetric entrainment range and were able to experimentally assess it. Our data imply that different clusters are subjected to different coupling strengths, and display independent endogenous periods. Finally, the manipulation employed here establishes a suitable paradigm to test other network properties as well as the cell-autonomous mechanisms running in different circadian-relevant clusters.
Collapse
Affiliation(s)
- Esteban J Beckwith
- Behavioral Genetics Laboratory, Fundación Instituto Leloir, Institute for Biochemical Research, Buenos Aires-Argentine Research Council, Buenos Aires, 1405BWE, Argentina
| | | |
Collapse
|
31
|
Flourakis M, Allada R. Patch-clamp electrophysiology in Drosophila circadian pacemaker neurons. Methods Enzymol 2014; 552:23-44. [PMID: 25707271 DOI: 10.1016/bs.mie.2014.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Circadian clocks modulate the action potential firing frequency of pacemaker neurons. This daily variation in membrane excitability has been described in multiple species: from mollusks to fruit flies and mammals. Here, we provide an overview of the Drosophila pacemaker neural network, how circadian clocks drive neuronal activity within this network and we will present electrophysiological methods that we have applied to directly measure neuronal activity and reveal signal transduction pathways.
Collapse
Affiliation(s)
- Matthieu Flourakis
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA.
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA.
| |
Collapse
|
32
|
Risau-Gusman S, Gleiser PM. A mathematical model of communication between groups of circadian neurons in Drosophila melanogaster. J Biol Rhythms 2014; 29:401-10. [PMID: 25416595 DOI: 10.1177/0748730414557865] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In the fruit fly, circadian behavior is controlled by a small number of specialized neurons, whose molecular clocks are relatively well known. However, much less is known about how these neurons communicate among themselves. In particular, only 1 circadian neuropeptide, pigment-dispersing factor (PDF), has been identified, and most aspects of its interaction with the molecular clock remain to be elucidated. Furthermore, it is speculated that many other peptides should contribute to circadian communication. We have developed a relatively detailed model of the 2 main groups of circadian pacemaker neurons (sLNvs and LNds) to investigate these issues. We have proposed many possible mechanisms for the interaction between the synchronization factors and the molecular clock, and we have compared the outputs with the experimental results reported in the literature both for the wild-type and PDF-null mutant. We have studied how different the properties of each neuron should be to account for the observations reported for the sLNvs in the mutant. We have found that only a few mechanisms, mostly related to the slowing down of nuclear entry of a circadian protein, can synchronize neurons that present these differences. Detailed immunofluorescent recordings have suggested that, whereas in the mutant, LNd neurons are synchronized, in the wild-type, a subset of the LNds oscillate faster than the rest. With our model, we find that a more likely explanation for the same observations is that this subset is being driven outside its synchronization range and displays therefore a complex pattern of oscillation.
Collapse
|
33
|
Kunst M, Tso MCF, Ghosh DD, Herzog ED, Nitabach MN. Rhythmic control of activity and sleep by class B1 GPCRs. Crit Rev Biochem Mol Biol 2014; 50:18-30. [PMID: 25410535 DOI: 10.3109/10409238.2014.985815] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Members of the class B1 family of G-protein coupled receptors (GPCRs) whose ligands are neuropeptides have been implicated in regulation of circadian rhythms and sleep in diverse metazoan clades. This review discusses the cellular and molecular mechanisms by which class B1 GPCRs, especially the mammalian VPAC2 receptor and its functional homologue PDFR in Drosophila and C. elegans, regulate arousal and daily rhythms of sleep and wake. There are remarkable parallels in the cellular and molecular roles played by class B1 intercellular signaling pathways in coordinating arousal and circadian timekeeping across multiple cells and tissues in these very different genetic model organisms.
Collapse
Affiliation(s)
- Michael Kunst
- Department of Cellular and Molecular Physiology, Yale University School of Medicine , New Haven, CT , USA and
| | | | | | | | | |
Collapse
|
34
|
Depetris-Chauvin A, Fernández-Gamba Á, Gorostiza EA, Herrero A, Castaño EM, Ceriani MF. Mmp1 processing of the PDF neuropeptide regulates circadian structural plasticity of pacemaker neurons. PLoS Genet 2014; 10:e1004700. [PMID: 25356918 PMCID: PMC4214601 DOI: 10.1371/journal.pgen.1004700] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 08/22/2014] [Indexed: 11/19/2022] Open
Abstract
In the Drosophila brain, the neuropeptide PIGMENT DISPERSING FACTOR (PDF) is expressed in the small and large Lateral ventral neurons (LNvs) and regulates circadian locomotor behavior. Interestingly, PDF immunoreactivity at the dorsal terminals changes across the day as synaptic contacts do as a result of a remarkable remodeling of sLNv projections. Despite the relevance of this phenomenon to circuit plasticity and behavior, the underlying mechanisms remain poorly understood. In this work we provide evidence that PDF along with matrix metalloproteinases (Mmp1 and 2) are key in the control of circadian structural remodeling. Adult-specific downregulation of PDF levels per se hampers circadian axonal remodeling, as it does altering Mmp1 or Mmp2 levels within PDF neurons post-developmentally. However, only Mmp1 affects PDF immunoreactivity at the dorsal terminals and exerts a clear effect on overt behavior. In vitro analysis demonstrated that PDF is hydrolyzed by Mmp1, thereby suggesting that Mmp1 could directly terminate its biological activity. These data demonstrate that Mmp1 modulates PDF processing, which leads to daily structural remodeling and circadian behavior. Circadian clocks have evolved as mechanisms that allow organisms to adapt to the day/night cyclical changes, a direct consequence of the rotation of the Earth. In the last two decades, and due to its amazing repertoire of genetic tools, Drosophila has been at the leading front in the discovery of genes that account for how the clock operates at a single cell level, which are conserved throughout the animal kingdom. Although the biochemical components underlying these molecular clocks have been characterized in certain detail, the mechanisms used by clock neurons to convey information to downstream pathways controlling behavior remain elusive. In the fruit fly, a subset of circadian neurons called the small ventral lateral neurons (sLNvs) are capable of synchronizing other clock cells relying on a neuropeptide named pigment dispersing factor (PDF). In addition, a number of years ago we described another mechanism as a possible candidate for contributing to the transmission of information downstream of the sLNvs, involving adult-specific remodeling of the axonal terminals of these circadian neurons. In this manuscript we describe some of the molecular events that lead to this striking form of structural plasticity on a daily basis.
Collapse
Affiliation(s)
- Ana Depetris-Chauvin
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Buenos Aires, Argentina
| | - Ágata Fernández-Gamba
- Laboratorio de Amiloidosis y Neurodegeneración, Fundación Instituto Leloir, IIB-BA-CONICET, Buenos Aires, Argentina
| | - E. Axel Gorostiza
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Buenos Aires, Argentina
| | - Anastasia Herrero
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Buenos Aires, Argentina
| | - Eduardo M. Castaño
- Laboratorio de Amiloidosis y Neurodegeneración, Fundación Instituto Leloir, IIB-BA-CONICET, Buenos Aires, Argentina
| | - M. Fernanda Ceriani
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
35
|
Calebiro D, Maiellaro I. cAMP signaling microdomains and their observation by optical methods. Front Cell Neurosci 2014; 8:350. [PMID: 25389388 PMCID: PMC4211404 DOI: 10.3389/fncel.2014.00350] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/07/2014] [Indexed: 11/22/2022] Open
Abstract
The second messenger cyclic AMP (cAMP) is a major intracellular mediator of many hormones and neurotransmitters and regulates a myriad of cell functions, including synaptic plasticity in neurons. Whereas cAMP can freely diffuse in the cytosol, a growing body of evidence suggests the formation of cAMP gradients and microdomains near the sites of cAMP production, where cAMP signals remain apparently confined. The mechanisms responsible for the formation of such microdomains are subject of intensive investigation. The development of optical methods based on fluorescence resonance energy transfer (FRET), which allow a direct observation of cAMP signaling with high temporal and spatial resolution, is playing a fundamental role in elucidating the nature of such microdomains. Here, we will review the optical methods used for monitoring cAMP and protein kinase A (PKA) signaling in living cells, providing some examples of their application in neurons, and will discuss the major hypotheses on the formation of cAMP/PKA microdomains.
Collapse
Affiliation(s)
- Davide Calebiro
- Institute of Pharmacology and Toxicology, University of Würzburg Würzburg, Germany ; Bio-Imaging Center/Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg Würzburg, Germany
| | - Isabella Maiellaro
- Institute of Pharmacology and Toxicology, University of Würzburg Würzburg, Germany ; Bio-Imaging Center/Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg Würzburg, Germany
| |
Collapse
|
36
|
Wei H, Yasar H, Funk NW, Giese M, Baz ES, Stengl M. Signaling of pigment-dispersing factor (PDF) in the Madeira cockroach Rhyparobia maderae. PLoS One 2014; 9:e108757. [PMID: 25269074 PMCID: PMC4182629 DOI: 10.1371/journal.pone.0108757] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 08/05/2014] [Indexed: 11/19/2022] Open
Abstract
The insect neuropeptide pigment-dispersing factor (PDF) is a functional ortholog of vasoactive intestinal polypeptide, the coupling factor of the mammalian circadian pacemaker. Despite of PDF's importance for synchronized circadian locomotor activity rhythms its signaling is not well understood. We studied PDF signaling in primary cell cultures of the accessory medulla, the circadian pacemaker of the Madeira cockroach. In Ca²⁺ imaging studies four types of PDF-responses were distinguished. In regularly bursting type 1 pacemakers PDF application resulted in dose-dependent long-lasting increases in Ca²⁺ baseline concentration and frequency of oscillating Ca²⁺ transients. Adenylyl cyclase antagonists prevented PDF-responses in type 1 cells, indicating that PDF signaled via elevation of intracellular cAMP levels. In contrast, in type 2 pacemakers PDF transiently raised intracellular Ca²⁺ levels even after blocking adenylyl cyclase activity. In patch clamp experiments the previously characterized types 1-4 could not be identified. Instead, PDF-responses were categorized according to ion channels affected. Application of PDF inhibited outward potassium or inward sodium currents, sometimes in the same neuron. In a comparison of Ca²⁺ imaging and patch clamp experiments we hypothesized that in type 1 cells PDF-dependent rises in cAMP concentrations block primarily outward K⁺ currents. Possibly, this PDF-dependent depolarization underlies PDF-dependent phase advances of pacemakers. Finally, we propose that PDF-dependent concomitant modulation of K⁺ and Na⁺ channels in coupled pacemakers causes ultradian membrane potential oscillations as prerequisite to efficient synchronization via resonance.
Collapse
Affiliation(s)
- Hongying Wei
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Hanzey Yasar
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Nico W. Funk
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Maria Giese
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - El-Sayed Baz
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
| | - Monika Stengl
- University of Kassel, FB 10, Biology, Animal Physiology, Kassel, Germany
- * E-mail:
| |
Collapse
|
37
|
Collins B, Kaplan HS, Cavey M, Lelito KR, Bahle AH, Zhu Z, Macara AM, Roman G, Shafer OT, Blau J. Differentially timed extracellular signals synchronize pacemaker neuron clocks. PLoS Biol 2014; 12:e1001959. [PMID: 25268747 PMCID: PMC4181961 DOI: 10.1371/journal.pbio.1001959] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 08/20/2014] [Indexed: 12/22/2022] Open
Abstract
Circadian pacemaker neurons in Drosophila are regulated by two synchronizing signals that are released at opposite times of day, generating a rhythm in intracellular cyclic AMP. Synchronized neuronal activity is vital for complex processes like behavior. Circadian pacemaker neurons offer an unusual opportunity to study synchrony as their molecular clocks oscillate in phase over an extended timeframe (24 h). To identify where, when, and how synchronizing signals are perceived, we first studied the minimal clock neural circuit in Drosophila larvae, manipulating either the four master pacemaker neurons (LNvs) or two dorsal clock neurons (DN1s). Unexpectedly, we found that the PDF Receptor (PdfR) is required in both LNvs and DN1s to maintain synchronized LNv clocks. We also found that glutamate is a second synchronizing signal that is released from DN1s and perceived in LNvs via the metabotropic glutamate receptor (mGluRA). Because simultaneously reducing Pdfr and mGluRA expression in LNvs severely dampened Timeless clock protein oscillations, we conclude that the master pacemaker LNvs require extracellular signals to function normally. These two synchronizing signals are released at opposite times of day and drive cAMP oscillations in LNvs. Finally we found that PdfR and mGluRA also help synchronize Timeless oscillations in adult s-LNvs. We propose that differentially timed signals that drive cAMP oscillations and synchronize pacemaker neurons in circadian neural circuits will be conserved across species. Circadian molecular clocks are essential for daily cycles in animal behavior and we have a good understanding of how these clocks work in individual pacemaker neurons. However, the accuracy of these individual clocks is meaningless unless they are synchronized with one another. In this study we show that synchronizing the principal pacemaker LNv neurons in Drosophila larvae require two extracellular signals that are received at opposite times of day: namely, the neuropeptide PDF released from LNvs themselves at dawn and glutamate released from dorsal clock neurons at dusk. LNvs perceive both PDF and glutamate via G-protein coupled receptors that increase or decrease intracellular cAMP, respectively. The alternating phases of PDF and glutamate release generate oscillations in intracellular cyclic AMP. In addition to maintaining synchrony between LNvs, this rhythm is also required for molecular clock oscillations in individual larval LNvs. We show that disruption of PDF and glutamate signaling also reduces synchrony in adult LNvs. This impairs the oscillations of clock proteins and flies have delayed onset of sleep. Our data highlight the importance of intercellular signaling in ensuring synchrony between clock neurons within the circadian network. Our findings help extend the conservation of clock properties between Drosophila and mammals beyond clock genes to include clock circuitry.
Collapse
Affiliation(s)
- Ben Collins
- Department of Biology, New York University, New York, New York, United States of America
| | - Harris S. Kaplan
- Department of Biology, New York University, New York, New York, United States of America
| | - Matthieu Cavey
- Department of Biology, New York University, New York, New York, United States of America
| | - Katherine R. Lelito
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrew H. Bahle
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Zhonghua Zhu
- Department of Biology, New York University, New York, New York, United States of America
| | - Ann Marie Macara
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Gregg Roman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, United States of America
| | - Orie T. Shafer
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Justin Blau
- Department of Biology, New York University, New York, New York, United States of America
- Center for Genomics & Systems Biology, New York University Abu Dhabi Institute, Abu Dhabi, United Arab Emirates
- Program in Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- * E-mail:
| |
Collapse
|
38
|
Schendzielorz J, Schendzielorz T, Arendt A, Stengl M. Bimodal oscillations of cyclic nucleotide concentrations in the circadian system of the Madeira cockroach Rhyparobia maderae. J Biol Rhythms 2014; 29:318-31. [PMID: 25231947 DOI: 10.1177/0748730414546133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pigment-dispersing factor (PDF) is the most important coupling factor of the circadian system in insects, comparable to its functional ortholog vasoactive intestinal polypeptide of the mammalian circadian clock. In Drosophila melanogaster, PDF signals via activation of adenylyl cyclases, controlling circadian locomotor activity rhythms at dusk and dawn. In addition, PDF mediates circadian rhythms of the visual system and is involved in entrainment to different photoperiods. We examined whether PDF daytime-dependently elevates cAMP levels in the Madeira cockroach Rhyparobia maderae and whether cAMP mimics PDF effects on locomotor activity rhythms. To determine time windows of PDF release, we searched for circadian rhythms in concentrations of cAMP and its functional opponent cGMP in the accessory medulla (AMe), the insect circadian pacemaker controlling locomotor activity rhythms, and in the optic lobes, as the major input and output area of the circadian clock. Enzyme-linked immunosorbent assays detected PDF-dependent increases of cAMP in optic lobes and daytime-dependent oscillations of cAMP and cGMP baseline levels in the AMe, both with maxima at dusk and dawn. Although these rhythms disappeared at the first day in constant conditions (DD1), cAMP but not cGMP oscillations returned at the second day in constant conditions (DD2). Whereas in light-dark cycles the cAMP baseline level remained constant in other optic lobe neuropils, it oscillated in phase with the AMe at DD2. To determine whether cAMP and cGMP mimic PDF-dependent control of locomotor activity rhythms, both cyclic nucleotides were injected at different times of the circadian day using running-wheel assays. Whereas cAMP injections generated delays at dusk and advances at dawn, cGMP only delayed locomotor activity at dusk. For the first time we found PDF-dependent phase advances at dawn in addition to previously described phase delays at dusk. Thus, we hypothesize that PDF release at dusk and dawn controls locomotor activity rhythms and visual system processing cAMP-dependently.
Collapse
Affiliation(s)
- Julia Schendzielorz
- Department of Biology, Animal Physiology, University of Kassel, Kassel, Germany
| | | | - Andreas Arendt
- Department of Biology, Animal Physiology, University of Kassel, Kassel, Germany
| | - Monika Stengl
- Department of Biology, Animal Physiology, University of Kassel, Kassel, Germany
| |
Collapse
|
39
|
Egekwu N, Sonenshine DE, Bissinger BW, Roe RM. Transcriptome of the female synganglion of the black-legged tick Ixodes scapularis (Acari: Ixodidae) with comparison between Illumina and 454 systems. PLoS One 2014; 9:e102667. [PMID: 25075967 PMCID: PMC4116169 DOI: 10.1371/journal.pone.0102667] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 06/23/2014] [Indexed: 11/26/2022] Open
Abstract
Illumina and 454 pyrosequencing were used to characterize genes from the synganglion of female Ixodes scapularis. GO term searching success for biological processes was similar for samples sequenced by both methods. However, for molecular processes, it was more successful for the Illumina samples than for 454 samples. Functional assignments of transcripts predicting neuropeptides, neuropeptide receptors, neurotransmitter receptors and other genes of interest was done, supported by strong e-values (<-6), and high consensus sequence alignments. Transcripts predicting 15 putative neuropeptide prepropeptides ((allatostatin, allatotropin, bursicon α, corticotropin releasing factor (CRF), CRF-binding protein, eclosion hormone, FMRFamide, glycoprotein A, insulin-like peptide, ion transport peptide, myoinhibitory peptide, inotocin ( = neurophysin-oxytocin), Neuropeptide F, sulfakinin and SIFamide)) and transcripts predicting receptors for 14 neuropeptides (allatostatin, calcitonin, cardioacceleratory peptide, corazonin, CRF, eclosion hormone, gonadotropin-releasing hormone/AKH-like, insulin-like peptide, neuropeptide F, proctolin, pyrokinin, SIFamide, sulfakinin and tachykinin) are reported. Similar to Dermacentor variabilis, we found transcripts matching pro-protein convertase, essential for converting neuropeptide hormones to their mature form. Additionally, transcripts predicting 6 neurotransmitter/neuromodulator receptors (acetylcholine, GABA, dopamine, glutamate, octopamine and serotonin) and 3 neurotransmitter transporters (GABA transporter, noradrenalin-norepinephrine transporter and Na+-neurotransmitter/symporter) are described. Further, we found transcripts predicting genes for pheromone odorant receptor, gustatory receptor, novel GPCR messages, ecdysone nuclear receptor, JH esterase binding protein, steroidogenic activating protein, chitin synthase, chitinase, and other genes of interest. Also found were transcripts predicting genes for spermatogenesis-associated protein, major sperm protein, spermidine oxidase and spermidine synthase, genes not normally expressed in the female CNS of other invertebrates. The diversity of messages predicting important genes identified in this study offers a valuable resource useful for understanding how the tick synganglion regulates important physiological functions.
Collapse
Affiliation(s)
- Noble Egekwu
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
| | - Daniel E. Sonenshine
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
| | | | - R. Michael Roe
- Department of Entomology, North Carolina State University, Raleigh, North Carolina, United States of America
| |
Collapse
|
40
|
Shafer OT, Yao Z. Pigment-Dispersing Factor Signaling and Circadian Rhythms in Insect Locomotor Activity. CURRENT OPINION IN INSECT SCIENCE 2014; 1:73-80. [PMID: 25386391 PMCID: PMC4224320 DOI: 10.1016/j.cois.2014.05.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Though expressed in relatively few neurons in insect nervous systems, pigment-dispersing factor (PDF) plays many roles in the control of behavior and physiology. PDF's role in circadian timekeeping is its best-understood function and the focus of this review. Here we recount the isolation and characterization of insect PDFs, review the evidence that PDF acts as a circadian clock output factor, and discuss emerging models of how PDF functions within circadian clock neuron network of Drosophila, the species in which this peptide's circadian roles are best understood.
Collapse
|
41
|
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
|
42
|
Synergistic interactions between the molecular and neuronal circadian networks drive robust behavioral circadian rhythms in Drosophila melanogaster. PLoS Genet 2014; 10:e1004252. [PMID: 24698952 PMCID: PMC3974645 DOI: 10.1371/journal.pgen.1004252] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 02/05/2014] [Indexed: 01/04/2023] Open
Abstract
Most organisms use 24-hr circadian clocks to keep temporal order and anticipate daily environmental changes. In Drosophila melanogaster CLOCK (CLK) and CYCLE (CYC) initiates the circadian system by promoting rhythmic transcription of hundreds of genes. However, it is still not clear whether high amplitude transcriptional oscillations are essential for circadian timekeeping. In order to address this issue, we generated flies in which the amplitude of CLK-driven transcription can be reduced partially (approx. 60%) or strongly (90%) without affecting the average levels of CLK-target genes. The impaired transcriptional oscillations lead to low amplitude protein oscillations that were not sufficient to drive outputs of peripheral oscillators. However, circadian rhythms in locomotor activity were resistant to partial reduction in transcriptional and protein oscillations. We found that the resilience of the brain oscillator is depending on the neuronal communication among circadian neurons in the brain. Indeed, the capacity of the brain oscillator to overcome low amplitude transcriptional oscillations depends on the action of the neuropeptide PDF and on the pdf-expressing cells having equal or higher amplitude of molecular rhythms than the rest of the circadian neuronal groups in the fly brain. Therefore, our work reveals the importance of high amplitude transcriptional oscillations for cell-autonomous circadian timekeeping. Moreover, we demonstrate that the circadian neuronal network is an essential buffering system that protects against changes in circadian transcription in the brain. Circadian clocks allow organisms to predict daily environmental changes. These clocks time the sleep/wake cycles and many other physiological and cellular pathways to 24hs rhythms. The current model states that circadian clocks keep time by the use of biochemical feedback loops. These feedback loops are responsible for the generation of high amplitude oscillations in gene expression. Abolishment of circadian transcriptional oscillations has been shown to abolish circadian function. Previous studies addressing this issue utilize manipulations in which the abolishment of the transcriptional oscillations is very dramatic and involves strong up or down-regulation of circadian genes. In this study we generated fruit flies in which we diminished the amplitude of circadian oscillations in a controlled way. We found that a decrease of more than 50% in the amplitude of circadian oscillations leads to impaired function of circadian physiological outputs in the periphery but does not significantly affect circadian behavior. This suggests that the clock in the brain has a specific compensatory mechanism. Moreover, we found that flies with reduced oscillation and impaired circadian neuronal communication display aberrant circadian rhythms. These finding support the idea of network buffering mechanisms that allows the brain to produce circadian rhythms even with low amplitude molecular oscillations.
Collapse
|
43
|
Seluzicki A, Flourakis M, Kula-Eversole E, Zhang L, Kilman V, Allada R. Dual PDF signaling pathways reset clocks via TIMELESS and acutely excite target neurons to control circadian behavior. PLoS Biol 2014; 12:e1001810. [PMID: 24643294 PMCID: PMC3958333 DOI: 10.1371/journal.pbio.1001810] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 02/05/2014] [Indexed: 12/20/2022] Open
Abstract
Molecular circadian clocks are interconnected via neural networks. In Drosophila, PIGMENT-DISPERSING FACTOR (PDF) acts as a master network regulator with dual functions in synchronizing molecular oscillations between disparate PDF(+) and PDF(-) circadian pacemaker neurons and controlling pacemaker neuron output. Yet the mechanisms by which PDF functions are not clear. We demonstrate that genetic inhibition of protein kinase A (PKA) in PDF(-) clock neurons can phenocopy PDF mutants while activated PKA can partially rescue PDF receptor mutants. PKA subunit transcripts are also under clock control in non-PDF DN1p neurons. To address the core clock target of PDF, we rescued per in PDF neurons of arrhythmic per⁰¹ mutants. PDF neuron rescue induced high amplitude rhythms in the clock component TIMELESS (TIM) in per-less DN1p neurons. Complete loss of PDF or PKA inhibition also results in reduced TIM levels in non-PDF neurons of per⁰¹ flies. To address how PDF impacts pacemaker neuron output, we focally applied PDF to DN1p neurons and found that it acutely depolarizes and increases firing rates of DN1p neurons. Surprisingly, these effects are reduced in the presence of an adenylate cyclase inhibitor, yet persist in the presence of PKA inhibition. We have provided evidence for a signaling mechanism (PKA) and a molecular target (TIM) by which PDF resets and synchronizes clocks and demonstrates an acute direct excitatory effect of PDF on target neurons to control neuronal output. The identification of TIM as a target of PDF signaling suggests it is a multimodal integrator of cell autonomous clock, environmental light, and neural network signaling. Moreover, these data reveal a bifurcation of PKA-dependent clock effects and PKA-independent output effects. Taken together, our results provide a molecular and cellular basis for the dual functions of PDF in clock resetting and pacemaker output.
Collapse
Affiliation(s)
- Adam Seluzicki
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Matthieu Flourakis
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Elzbieta Kula-Eversole
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Luoying Zhang
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Valerie Kilman
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
| | - Ravi Allada
- Department of Neurobiology, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|
44
|
Vecsey CG, Pírez N, Griffith LC. The Drosophila neuropeptides PDF and sNPF have opposing electrophysiological and molecular effects on central neurons. J Neurophysiol 2014; 111:1033-45. [PMID: 24353297 PMCID: PMC3949227 DOI: 10.1152/jn.00712.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 12/12/2013] [Indexed: 12/26/2022] Open
Abstract
Neuropeptides have widespread effects on behavior, but how these molecules alter the activity of their target cells is poorly understood. We employed a new model system in Drosophila melanogaster to assess the electrophysiological and molecular effects of neuropeptides, recording in situ from larval motor neurons, which transgenically express a receptor of choice. We focused on two neuropeptides, pigment-dispersing factor (PDF) and small neuropeptide F (sNPF), which play important roles in sleep/rhythms and feeding/metabolism. PDF treatment depolarized motor neurons expressing the PDF receptor (PDFR), increasing excitability. sNPF treatment had the opposite effect, hyperpolarizing neurons expressing the sNPF receptor (sNPFR). Live optical imaging using a genetically encoded fluorescence resonance energy transfer (FRET)-based sensor for cyclic AMP (cAMP) showed that PDF induced a large increase in cAMP, whereas sNPF caused a small but significant decrease in cAMP. Coexpression of pertussis toxin or RNAi interference to disrupt the G-protein Gαo blocked the electrophysiological responses to sNPF, showing that sNPFR acts via Gαo signaling. Using a fluorescent sensor for intracellular calcium, we observed that sNPF-induced hyperpolarization blocked spontaneous waves of activity propagating along the ventral nerve cord, demonstrating that the electrical effects of sNPF can cause profound changes in natural network activity in the brain. This new model system provides a platform for mechanistic analysis of how neuropeptides can affect target cells at the electrical and molecular level, allowing for predictions of how they regulate brain circuits that control behaviors such as sleep and feeding.
Collapse
Affiliation(s)
- Christopher G Vecsey
- National Center for Behavioral Genomics, Volen National Center for Complex Systems and Department of Biology, Brandeis University, Waltham, Massachusetts
| | | | | |
Collapse
|
45
|
Castro LRV, Guiot E, Polito M, Paupardin-Tritsch D, Vincent P. Decoding spatial and temporal features of neuronal cAMP/PKA signaling with FRET biosensors. Biotechnol J 2014; 9:192-202. [PMID: 24478276 DOI: 10.1002/biot.201300202] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 12/02/2013] [Accepted: 01/08/2014] [Indexed: 11/11/2022]
Abstract
Cyclic adenosine monophosphate (cAMP) and the cyclic-AMP-dependent protein kinase (PKA) regulate a plethora of cellular functions in virtually all eukaryotic cells. In neurons, the cAMP/PKA signaling cascade controls a number of biological properties such as axonal growth, pathfinding, efficacy of synaptic transmission, regulation of excitability, or long term changes. Genetically encoded optical biosensors for cAMP or PKA are considerably improving our understanding of these processes by providing a real-time measurement in living neurons. In this review, we describe the recent progress made in the creation of biosensors for cAMP or PKA activity. These biosensors revealed profound differences in the amplitude of the cAMP signal evoked by neuromodulators between various neuronal preparations. These responses can be resolved at the level of individual neurons, also revealing differences related to the neuronal type. At the sub-cellular level, biosensors reported different signal dynamics in domains like dendrites, cell body, nucleus, and axon. Combining this imaging approach with pharmacology or genetic models points at phosphodiesterases and phosphatases as critical regulatory proteins. Biosensor imaging will certainly emerge as a forefront tool to decipher the subtle mechanics of intracellular signaling. This will certainly help us to understand the mechanism of action of current drugs and foster the development of novel molecules for neuropsychiatric diseases.
Collapse
Affiliation(s)
- Liliana R V Castro
- CNRS UMR7102, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR7102, Paris, France
| | | | | | | | | |
Collapse
|
46
|
Duvall LB, Taghert PH. E and M circadian pacemaker neurons use different PDF receptor signalosome components in drosophila. J Biol Rhythms 2013; 28:239-48. [PMID: 23929551 DOI: 10.1177/0748730413497179] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We used real-time imaging to detect cAMP levels in neurons of intact fly brains to study the mechanisms of circadian pacemaker synchronization by the neuropeptide pigment dispersing factor (PDF) in Drosophila. PDF receptor (PDF-R) is expressed by both M (sLNv) and E (LNd) pacemaker subclasses and is coupled to G(sα) in both cases. We previously reported that PDF-R in M pacemakers elevates cAMP levels by activating the ortholog of mammalian adenylate cyclase 3 (AC3) but that AC3 disruptions had no effect on E pacemaker sensitivity to PDF. Here, we show that PDF-R in E pacemakers activates a different AC isoform, AC78C, an ortholog of mammalian AC8. Knockdown of AC78C by transgenic RNAi substantially reduces, but does not completely abrogate, PDF responses in these E pacemakers. The knockdown effect is intact when restricted to mature stages, suggesting a physiological and not a development role for AC78C in E pacemakers. The AC78C phenotype is rescued by the overexpression of AC78C but not by overexpression of the rutabaga AC. AC78C overexpression does not disrupt PDF responses in these E pacemakers, and neither AC78C knockdown nor its overexpression disrupted locomotor rhythms. Finally, knockdown of 2 AKAPs, nervy and AKAP200, partially reduces LNd PDF responses. These findings begin to identify the components of E pacemaker PDF-R signalosomes and indicate that they are distinct from PDF-R signalosomes in M pacemakers: we propose they contain AC78C and at least 1 other AC.
Collapse
Affiliation(s)
- Laura B Duvall
- Department of Anatomy & Neurobiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
47
|
Damulewicz M, Rosato E, Pyza E. Circadian regulation of the Na+/K+-ATPase alpha subunit in the visual system is mediated by the pacemaker and by retina photoreceptors in Drosophila melanogaster. PLoS One 2013; 8:e73690. [PMID: 24040028 PMCID: PMC3769360 DOI: 10.1371/journal.pone.0073690] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 07/29/2013] [Indexed: 12/20/2022] Open
Abstract
We investigated the diurnal oscillation in abundance of the catalytic α subunit of the sodium/potassium pump (ATPα) in the brain of Drosophila melanogaster. This rhythm is bimodal and is particularly robust in the glia cells of the lamina, the first optic neuropil. We observed loss of ATPα cycling in lamina glia in behaviourally arrhythmic per01 and tim01 mutants and in flies overexpressing the pro-apoptotic gene hid in the PDF-positive clock neurons. Moreover, the rhythm of ATPα abundance was altered in cry01 and Pdf0 mutants, in flies with a weakened clock mechanism in retina photoreceptor cells and in those subject to downregulation of the neuropeptide ITP by RNAi. This complex, rhythmic regulation of the α subunit suggests that the sodium/potassium pump may be a key target of the circadian pacemaker to impose daily control on brain activities, such as rhythmic changes in neuronal plasticity, which are best observed in the visual system.
Collapse
Affiliation(s)
- Milena Damulewicz
- Department of Cell Biology and Imaging, Institute of Zoology, Jagiellonian University, Kraków, Poland
| | - Ezio Rosato
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology, Jagiellonian University, Kraków, Poland
- * E-mail:
| |
Collapse
|
48
|
Zhang Y, Emery P. GW182 controls Drosophila circadian behavior and PDF-receptor signaling. Neuron 2013; 78:152-65. [PMID: 23583112 DOI: 10.1016/j.neuron.2013.01.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2013] [Indexed: 12/19/2022]
Abstract
The neuropeptide PDF is crucial for Drosophila circadian behavior: it keeps circadian neurons synchronized. Here, we identify GW182 as a key regulator of PDF signaling. Indeed, GW182 downregulation results in phenotypes similar to those of Pdf and Pdf-receptor (Pdfr) mutants. gw182 genetically interacts with Pdfr and cAMP signaling, which is essential for PDFR function. GW182 mediates miRNA-dependent gene silencing through its interaction with AGO1. Consistently, GW182's AGO1 interaction domain is required for GW182's circadian function. Moreover, our results indicate that GW182 modulates PDFR signaling by silencing the expression of the cAMP phosphodiesterase DUNCE. Importantly, this repression is under photic control, and GW182 activity level--which is limiting in circadian neurons--influences the responses of the circadian neural network to light. We propose that GW182's gene silencing activity functions as a rheostat for PDFR signaling and thus profoundly impacts the circadian neural network and its response to environmental inputs.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Neurobiology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA
| | | |
Collapse
|
49
|
Muraro NI, Pírez N, Ceriani MF. The circadian system: plasticity at many levels. Neuroscience 2013; 247:280-93. [PMID: 23727010 DOI: 10.1016/j.neuroscience.2013.05.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/17/2013] [Accepted: 05/20/2013] [Indexed: 11/16/2022]
Abstract
Over the years it has become crystal clear that a variety of processes encode time-of-day information, ranging from gene expression, protein stability, or subcellular localization of key proteins, to the fine tuning of network properties and modulation of input signals, ultimately ensuring that physiology and behavior are properly synchronized to a changing environment. The purpose of this review is to put forward examples (as opposed to generate a comprehensive revision of all the available literature) in which the circadian system displays a remarkable degree of plasticity, from cell autonomous to circuit-based levels. In the literature, the term circadian plasticity has been used to refer to different concepts. The obvious one, more literally, refers to any change that follows a circadian (circa=around, diem=day) pattern, i.e. a daily change of a given parameter. The discovery of daily remodeling of neuronal structures will be referred herein as structural circadian plasticity, and represents an additional and novel phenomenon modified daily. Finally, any plasticity that has to do with a circadian parameter would represent a type of circadian plasticity; as an example, adjustments that allow organisms to adapt their daily behavior to the annual changes in photoperiod is a form of circadian plasticity at a higher organizational level, which is an emergent property of the whole circadian system. Throughout this work we will revisit these types of changes by reviewing recent literature delving around circadian control of clock outputs, from the most immediate ones within pacemaker neurons to the circadian modulation of rest-activity cycles.
Collapse
Affiliation(s)
- N I Muraro
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir, IIB-BA-CONICET, Buenos Aires, Argentina
| | | | | |
Collapse
|
50
|
Pírez N, Christmann BL, Griffith LC. Daily rhythms in locomotor circuits in Drosophila involve PDF. J Neurophysiol 2013; 110:700-8. [PMID: 23678016 DOI: 10.1152/jn.00126.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The neuropeptide pigment-dispersing factor (PDF) has been studied extensively in Drosophila, and its role in circadian time-keeping has been firmly established. The role of PDF outside of the clock circuit, however, is poorly understood. A recent study suggested that PDF may act on the ellipsoid body (EB) to link the clock and sleep/activity circuits. We performed whole brain optical imaging with the fluorescence resonance energy transfer (FRET)-based cAMP sensor Epac1-camps expressed under control of the pdfR promoter to address how the clock and sleep deprivation affect the physiology of these cells. Basal cAMP levels in EB were regulated both by PDF and synaptic inputs that are controlled by the circadian clock. Acute application of PDF to the brain caused a significant, and PDF-receptor-dependent, increase in cAMP in EB cells. Application of TTX to block circuit-mediated effects of PDF increased the morning response but not the response at night, implying the existence of a temporally regulated, PDF-stimulated input that blocks cAMP generation. ACh produced both direct (TTX-insensitive) and indirect (TTX-sensitive) increases in cAMP during the day but was totally TTX-insensitive at night, indicating that ACh-stimulated inputs to the EB are suppressed at night. Sleep deprivation did not affect the cAMP responses of these cells to either PDF or ACh. These results suggest a novel role for PDF as a modulator of activity outside of the clock circuit. By elucidating the mechanisms by which the neuropeptide PDF act on its target cells, our work contributes to our understating of how the central clock coordinates activity and sleep.
Collapse
Affiliation(s)
- Nicolás Pírez
- Volen Center for Complex Systems and National Center for Behavioral Genomics, Department of Biology, Brandeis University, Waltham, MA 02454-9110, USA
| | | | | |
Collapse
|