1
|
Force E, Debernard S. Endocrine regulation of reproductive behaviors in insects: a comprehensive review. CURRENT OPINION IN INSECT SCIENCE 2025; 69:101360. [PMID: 40058696 DOI: 10.1016/j.cois.2025.101360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
Insects use pheromones in a complex system of sexual communication for reproduction. Hormones, peptides, and biogenic amines are crucial regulators involved in reproductive behaviors. Despite knowledge gaps, this review shows how hormones and related molecules influence insect reproduction and highlights the intricate endocrine network that governs reproductive behaviors through diverse signaling pathways. In the future, it will be very interesting to explore not only endocrine regulation but also the impact of environmental changes on reproductive behaviors, deepening our understanding of insect reproductive processes and their adaptability.
Collapse
Affiliation(s)
- Evan Force
- Sorbonne Université, Université Paris-Est Créteil, INRAE, CNRS, IRD, Institute for Ecology and Environmental Sciences of Paris, iEES Paris, F-75005 Paris, France.
| | - Stéphane Debernard
- Sorbonne Université, Université Paris-Est Créteil, INRAE, CNRS, IRD, Institute for Ecology and Environmental Sciences of Paris, iEES Paris, F-75005 Paris, France.
| |
Collapse
|
2
|
Lee GG, Peterson AJ, Kim MJ, Shimell M, O’Connor MB, Park JH. Linking expression and function of Drosophila type-I TGF-β receptor baboon isoforms: Multiple roles of BaboA isoform in shaping of the adult central nervous system. PLoS One 2025; 20:e0318406. [PMID: 40445987 PMCID: PMC12124520 DOI: 10.1371/journal.pone.0318406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 05/05/2025] [Indexed: 06/02/2025] Open
Abstract
Evolutionarily conserved transforming growth factor β (TGF-β) signaling is used in both vertebrates and invertebrates to regulate a variety of developmental and cellular processes. The baboon (babo) gene encoding a Drosophila type-I TGF-β receptor produces three isoforms via alternative splicing: BaboA, BaboB, and BaboC. In this study, we generated three fly lines, each carrying an isoform-specific GFP tag, and another line with a GFP conjugated at the C-terminus common to all isoforms. Using these lines, we assessed (1) whether the tagged proteins function properly in rescue assays and (2) how the isoform expression is regulated in various tissues including the central nervous system (CNS). A Gal4 knock-in line in the babo locus was also characterized for reporter expression, mutant phenotypes, and isoform-specific knockdown phenotypes. We found that the C-terminal tag does not interrupt the subcellular targeting and functions of the tagged isoforms, but the internal isoform tags do so in a cell- and isoform-specific fashion. Nevertheless, our results demonstrated that these tags faithfully reflect endogenous expression of individual isoforms. Certain cell types express single or multiple isoforms at different levels, suggesting that alternative splicing could determine the isoform types and their levels depending on cell (or tissue) type. The larval CNS displays distinct patterns of two isoforms, BaboA and BaboC. BaboC is mostly expressed in neural cells originating during embryogenesis, while BaboA is broadly expressed in neural cells produced from both embryonic and postembryonic stages. Assays of both isoform-specific mutants and cell-specific knockdown of individual isoforms revealed broad roles played by BaboA in postembryonic neurogenesis and differentiation of precursor neurons, remodeling processes of persisting larval neurons, and metamorphic CNS reorganization, which are essential for establishing of the adult CNS. Taken together, this study demonstrates that the GFP-tagged lines permit visualization of endogenous expression of individual isoforms, which further provides clues about cell- and stage-specific functions played by each isoform.
Collapse
Affiliation(s)
- Gyunghee G. Lee
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Aidan J. Peterson
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - MaryJane Shimell
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Michael B. O’Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jae H. Park
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| |
Collapse
|
3
|
Asahina K, Zelikowsky M. Comparative Perspectives on Neuropeptide Function and Social Isolation. Biol Psychiatry 2025; 97:942-952. [PMID: 39892690 PMCID: PMC12048258 DOI: 10.1016/j.biopsych.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
Chronic social isolation alters behavior across animal species. Genetic model organisms such as mice and flies provide crucial insight into the molecular and physiological effects of social isolation on brain cells and circuits. Here, we comparatively review recent findings regarding the function of conserved neuropeptides in social isolation in mice and flies. Analogous functions of 3 classes of neuropeptides-tachykinins, cholecystokinins, and neuropeptide Y/F-in the two model organisms suggest that these molecules may be involved in modulating behavioral changes induced by social isolation across a wider range of species, including humans. Comparative approaches armed with tools to dissect neuropeptidergic function can lead to an integrated understanding of the impacts of social isolation on brain circuits and behavior.
Collapse
Affiliation(s)
- Kenta Asahina
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California.
| | - Moriel Zelikowsky
- Department of Neurobiology, School of Medicine, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
4
|
Ombuya A, Guo J, Liu W. Insect Mating Behaviors: A Review of the Regulatory Role of Neuropeptides. INSECTS 2025; 16:506. [PMID: 40429219 PMCID: PMC12112582 DOI: 10.3390/insects16050506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/19/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Insect mating behaviors are complex, diverse, and primarily regulated by neuropeptides and their receptors. Neuropeptides are peptide signaling molecules mainly secreted by insects' central nervous system (CNS) to reach target organs. A substantial body of research on the role of neuropeptides in regulating mating behaviors in insects has been undertaken. This review aims to (1) synthesize existing knowledge on insect mating behaviors, (2) elucidate the neuropeptidergic mechanisms governing these behaviors, and (3) identify knowledge gaps and propose future research directions. The mating process, covering mate attraction, courtship rituals, copulation, and post-mating behaviors, was elucidated with appropriate examples. Additionally, specific neuropeptides involved at each stage of the mating process, their functions, and mechanistic aspects were discussed as demonstrated in research. The review highlights that insects display behavioral dimorphism in the mating process driven by a complex underlying neuropeptidergic mechanism. While previous publications have generally addressed the role of neuropeptides in insect behavior, none has intensively and methodically examined their role in mating behaviors. In this review, we synthesized 18 neuropeptides that we found to regulate mating behaviors in insects. We note that some of the neuropeptides are malfunctional in their regulatory roles, while others are specific. We also note that these neuropeptides execute their regulatory functions through the G protein-coupled receptor (GPCR) signaling pathway but may take different routes and messengers downstream to effect behavioral change. Neuropeptides also interact with other regulatory systems, such as the endocrine system, to discharge their functions. Given their significance in mediating mating and reproduction, targeted manipulation of the signaling system of neuropeptides could serve as viable targets in the production of ecologically friendly pest management tools. Tools that could disrupt the mating process would be applied in crop production systems to reduce the population pressure of destructive pests, consequently reducing the urge to use chemical pesticides that are ecologically unfriendly. Our findings not only advance the understanding of neuropeptide-mediated mating regulation but also highlight their potential as eco-friendly pest control targets.
Collapse
Affiliation(s)
- Alfayo Ombuya
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China;
- Key Laboratory of Invasive Alien Species Control of Ministry of Agriculture and Rural Affairs, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Kenya Plant Health Inspectorate Service (KEPHIS), Mombasa Regional Office, Mombasa P.O. Box 80126-80100, Kenya
| | - Jianyang Guo
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China;
- Key Laboratory of Invasive Alien Species Control of Ministry of Agriculture and Rural Affairs, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Wanxue Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China;
- Key Laboratory of Invasive Alien Species Control of Ministry of Agriculture and Rural Affairs, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
5
|
Biswas P, Bako JA, Liston JB, Yu H, Wat LW, Miller CJ, Gordon MD, Huan T, Stanley M, Rideout EJ. Insulin/insulin-like growth factor signaling pathway promotes higher fat storage in Drosophila females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.623936. [PMID: 40342968 PMCID: PMC12060994 DOI: 10.1101/2024.11.18.623936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
In Drosophila , adult females store more fat than males. While the mechanisms that restrict body fat in males are becoming clearer, less is known about how females achieve higher fat storage. Here, we perform a detailed investigation of the mechanisms that promote higher fat storage in females. We show greater intake of dietary sugar supports higher fat storage due to female-biased remodeling of the fat body lipidome. Dietary sugar stimulates a female-specific increase in Drosophila insulin-like peptide 3 (Dilp3), which acts together with greater peripheral insulin sensitivity to augment insulin/insulin-like growth factor signaling pathway (IIS) activity in adult females. Indeed, Dilp3 overexpression prevented the female-biased decrease in body fat after removal of dietary sugar. Given that adult-specific IIS inhibition caused a female-biased decrease in body fat, our data reveal IIS as a key determinant of female fat storage.
Collapse
|
6
|
Iyer AR, Scholz-Carlson E, Bell E, Biondi G, Richhariya S, Fernandez MP. Circadian rhythms are more resilient to pacemaker neuron disruption in female Drosophila. PLoS Biol 2025; 23:e3003146. [PMID: 40327674 PMCID: PMC12080924 DOI: 10.1371/journal.pbio.3003146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 05/15/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
The circadian system regulates the timing of multiple molecular, physiological, metabolic, and behavioral phenomena. In Drosophila, as in other species, most of the research on how the timekeeping system in the brain controls the timing of behavioral outputs has been conducted in males, or sex has not been included as a biological variable. A critical set of circadian pacemaker neurons in Drosophila release the neuropeptide pigment-dispersing factor (PDF), which functions as a key output factor in the network with complex effects on other clock neurons. Lack of Pdf or its receptor, PdfR, results in most flies displaying arrhythmicity in activity-rest cycles under constant conditions. However, our results show that female circadian rhythms are less affected by mutations in both Pdf and PdfR. CRISPR-Cas9-mediated mutagenesis of Pdf, specifically in ventral lateral neurons (LNvs), also has a greater effect on male rhythms. We tested the influence of M-cells on the circadian network and showed that speeding up the molecular clock specifically in M-cells led to sexually dimorphic phenotypes, with a more pronounced effect on male rhythmic behavior. Our results suggest that the female circadian system is more resilient to manipulations of M-cells and the PDF pathway, suggesting that circadian timekeeping is more distributed across the clock neuron network in females.
Collapse
Affiliation(s)
- Aishwarya Ramakrishnan Iyer
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, United States of America
- Department of Neuroscience and Behavior, Barnard College, New York City, New York, United States of America
| | - Eva Scholz-Carlson
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, United States of America
- Department of Neuroscience and Behavior, Barnard College, New York City, New York, United States of America
| | - Evardra Bell
- Department of Neuroscience and Behavior, Barnard College, New York City, New York, United States of America
| | - Grace Biondi
- Department of Neuroscience and Behavior, Barnard College, New York City, New York, United States of America
| | - Shlesha Richhariya
- HHMI, Brandeis University, Waltham, Massachusetts, United States of America
| | - Maria P. Fernandez
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana, United States of America
- Department of Neuroscience and Behavior, Barnard College, New York City, New York, United States of America
| |
Collapse
|
7
|
Sabandal PR, Kim YC, Sabandal JM, Han KA. Social context and dopamine signaling converge in the mushroom body to drive impulsivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.21.639508. [PMID: 40027633 PMCID: PMC11870619 DOI: 10.1101/2025.02.21.639508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Organisms adapt their behaviors flexibly in response to various internal and environmental factors. However, how and where these factors converge in the brain to alter behavior is not well understood. In this study, we examine how social context interacts with dopamine activity to influence inhibitory control in Drosophila . We found that, regardless of social context-whether isolated or in groups-wild-type flies consistently showed strong movement suppression in a go/no-go task that measures action restraint. In contrast, flies with enhanced dopamine activity suppressed their movements when tested alone or with potential mates but exhibited impulsive behaviors when exposed to same-sex peers. This social-context-dependent impulsivity was shown to rely on dopamine-D1 receptor-cAMP signaling in mushroom body (MB) neurons. Remarkably, activating the MB was sufficient to induce impulsivity, even without dopamine input or a social context. Our findings highlight MB as a critical hub where social context and dopamine signaling converge to regulate impulsive behavior in Drosophila . Signficance statement This study demonstrates that impulsivity results from the interplay between elevated dopamine levels and social context, rather than dopamine alone, with the mushroom body (MB) serving as a key neural hub for integrating these signals in Drosophila . Social stimuli, such as the presence of same-sex peers, disrupt inhibitory control in a context-dependent manner, highlighting the importance of multimodal sensory inputs and MB activity. These findings challenge the isolation-focused approach in traditional impulsivity research and underscore the need to account for social influences when investigating cognitive processes and disorders like ADHD, autism, and substance use, where social settings often amplify symptoms. Classification Genetics / Neuroscience.
Collapse
|
8
|
Akpoghiran O, Strich AK, Koh K. Effects of sex, mating status, and genetic background on circadian behavior in Drosophila. Front Neurosci 2025; 18:1532868. [PMID: 39844849 PMCID: PMC11750873 DOI: 10.3389/fnins.2024.1532868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Circadian rhythms play a crucial role in regulating behavior, physiology, and health. Sexual dimorphism, a widespread phenomenon across species, influences circadian behaviors. Additionally, post-mating physiological changes in females are known to modulate various behaviors, yet their effects on circadian rhythms remain underexplored. Here, using Drosophila melanogaster, a powerful model for studying circadian mechanisms, we systematically assessed the impact of sex and mating status on circadian behavior. We measured circadian period length and rhythm strength in virgin and mated males and females, including females mated to males lacking Sex Peptide (SP), a key mediator of post-mating changes. Across four wild-type and control strains, we found that males consistently exhibited shorter circadian periods than females, regardless of mating status, suggesting that circadian period length is a robust sexually dimorphic trait. In contrast, rhythm strength was influenced by the interaction between sex and mating status, with female mating generally reducing rhythm strength in the presence of SP signaling. Notably, genetic background significantly modulated these effects on rhythm strength. Our findings demonstrate that while circadian period length is a stable sex-specific trait, rhythm strength is shaped by a complex interplay between sex, mating status, and genetic background. This study advances our understanding of how sex and mating influence circadian rhythms in Drosophila and provides a foundation for future research into sexually dimorphic mechanisms underlying human diseases associated with circadian disruptions.
Collapse
Affiliation(s)
| | | | - Kyunghee Koh
- Department of Neuroscience, Farber Institute for Neurosciences, Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
9
|
Akpoghiran O, Strich AK, Koh K. Effects of sex, mating status, and genetic background on circadian behavior in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624853. [PMID: 39605702 PMCID: PMC11601570 DOI: 10.1101/2024.11.22.624853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Circadian rhythms play a crucial role in regulating behavior, physiology, and health. Sexual dimorphism, a widespread phenomenon across species, influences circadian behaviors. Additionally, post-mating physiological changes in females are known to modulate various behaviors, yet their effects on circadian rhythms remain underexplored. Here, using Drosophila melanogaster, a powerful model for studying circadian mechanisms, we systematically assessed the impact of sex and mating status on circadian behavior. We measured circadian period length and rhythm strength in virgin and mated males and females, including females mated to males lacking Sex Peptide (SP), a key mediator of post-mating changes. Across four wild-type and control strains, we found that males consistently exhibited shorter circadian periods than females, regardless of mating status, suggesting that circadian period length is a robust sexually dimorphic trait. In contrast, rhythm strength was influenced by the interaction between sex and mating status, with female mating generally reducing rhythm strength in the presence of SP signaling. Notably, genetic background significantly modulated these effects on rhythm strength. Our findings demonstrate that while circadian period length is a stable sex-specific trait, rhythm strength is shaped by a complex interplay between sex, mating status, and genetic background. This study advances our understanding of how sex and mating influence circadian rhythms in Drosophila and provides a foundation for future research into sexually dimorphic mechanisms underlying human diseases associated with circadian disruptions.
Collapse
Affiliation(s)
- Oghenerukevwe Akpoghiran
- Department of Neuroscience, Farber Institute for Neurosciences, and Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexandra K. Strich
- Department of Neuroscience, Farber Institute for Neurosciences, and Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Kyunghee Koh
- Department of Neuroscience, Farber Institute for Neurosciences, and Synaptic Biology Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
10
|
Iyer AR, Scholz-Carlson E, Bell E, Biondi G, Richhariya S, Fernandez MP. The Circadian Neuropeptide PDF has Sexually Dimorphic Effects on Activity Rhythms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578273. [PMID: 38352594 PMCID: PMC10862788 DOI: 10.1101/2024.01.31.578273] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The circadian system regulates the timing of multiple molecular, physiological, metabolic, and behavioral phenomena. In Drosophila as in other species, most of the research on how the timekeeping system in the brain controls timing of behavioral outputs has been conducted in males, or sex was not included as a biological variable. The main circadian pacemaker neurons in Drosophila release the neuropeptide Pigment Dispersing Factor (PDF), which functions as a key synchronizing factor in the network with complex effects on other clock neurons. Lack of Pdf or its receptor, PdfR, results in most flies displaying arrhythmicity in activity-rest cycles under constant conditions. However, our results show that female circadian rhythms are less affected by mutations in both Pdf and PdfR. Crispr-Cas9 mutagenesis of Pdf specifically in the ventral lateral neurons (LNvs) also has a greater effect on male rhythms. We tested the influence of the M-cells over the circadian network and show that speeding up the molecular clock specifically in the M-cells leads to sexually dimorphic phenotypes, with a more pronounced effect on male rhythmic behavior. Our results suggest that the female circadian system is more resilient to manipulations of the PDF pathway and that circadian timekeeping is more distributed across the clock neuron network in females.
Collapse
|
11
|
Le JQ, Ma D, Dai X, Rosbash M. Light and dopamine impact two circadian neurons to promote morning wakefulness. Curr Biol 2024; 34:3941-3954.e4. [PMID: 39142287 PMCID: PMC11404089 DOI: 10.1016/j.cub.2024.07.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/13/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024]
Abstract
In both mammals and flies, circadian brain neurons orchestrate physiological oscillations and behaviors like wake and sleep-these neurons can be subdivided by morphology and by gene expression patterns. Recent single-cell sequencing studies identified 17 Drosophila circadian neuron groups. One of these includes only two lateral neurons (LNs), which are marked by the expression of the neuropeptide ion transport peptide (ITP). Although these two ITP+ LNs have long been grouped with five other circadian evening activity cells, inhibiting the two neurons alone strongly reduces morning activity, indicating that they also have a prominent morning function. As dopamine signaling promotes activity in Drosophila, like in mammals, we considered that dopamine might influence this morning activity function. Moreover, the ITP+ LNs express higher mRNA levels than other LNs of the type 1-like dopamine receptor Dop1R1. Consistent with the importance of Dop1R1, cell-specific CRISPR-Cas9 mutagenesis of this receptor in the two ITP+ LNs renders flies significantly less active in the morning, and ex vivo live imaging shows Dop1R1-dependent cyclic AMP (cAMP) responses to dopamine in these two neurons. Notably, the response is more robust in the morning, reflecting higher morning Dop1R1 mRNA levels in the two neurons. As mRNA levels are not elevated in constant darkness, this suggests light-dependent upregulation of morning Dop1R1 transcript levels. Taken together with the enhanced morning cAMP response to dopamine, the data indicate how light and dopamine promote morning wakefulness in flies, mimicking the important effect of light on morning wakefulness in humans.
Collapse
Affiliation(s)
- Jasmine Quynh Le
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Dingbang Ma
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA; Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xihuimin Dai
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02453, USA.
| |
Collapse
|
12
|
Dou X, Chen K, Brown MR, Strand MR. Reciprocal interactions between neuropeptide F and RYamide regulate host attraction in the mosquito Aedes aegypti. Proc Natl Acad Sci U S A 2024; 121:e2408072121. [PMID: 38950363 PMCID: PMC11252962 DOI: 10.1073/pnas.2408072121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 07/03/2024] Open
Abstract
Female mosquitoes produce eggs in gonadotrophic cycles that are divided between a previtellogenic and vitellogenic phase. Previtellogenic females consume water and sugar sources like nectar while also being attracted to hosts for blood feeding. Consumption of a blood meal activates the vitellogenic phase, which produces mature eggs and suppresses host attraction. In this study, we tested the hypothesis that neuropeptide Y-like hormones differentially modulate host attraction behavior in the mosquito Aedes aegypti. A series of experiments collectively indicated that enteroendocrine cells (EECs) in the posterior midgut produce and release neuropeptide F (NPF) into the hemolymph during the previtellogenic phase which stimulates attraction to humans and biting behavior. Consumption of a blood meal, which primarily consists of protein by dry weight, down-regulated NPF in EECs until mature eggs developed, which was associated with a decline in hemolymph titer. NPF depletion depended on protein digestion but was not associated with EEC loss. Other experiments showed that neurons in the terminal ganglion extend axons to the posterior midgut and produce RYamide, which showed evidence of increased secretion into circulation after a blood meal. Injection of RYamide-1 and -2 into previtellogenic females suppressed host attraction, while coinjection of RYamides with or without short NPF-2 also inhibited the host attraction activity of NPF. Overall, our results identify NPF and RYamide as gut-associated hormones in A. aegypti that link host attraction behavior to shifts in diet during sequential gonadotrophic cycles.
Collapse
Affiliation(s)
- Xiaoyi Dou
- Department of Entomology, University of Georgia, Athens, GA30602
| | - Kangkang Chen
- Department of Entomology, University of Georgia, Athens, GA30602
| | - Mark R. Brown
- Department of Entomology, University of Georgia, Athens, GA30602
| | | |
Collapse
|
13
|
Mao R, Yu J, Deng B, Dai X, Du Y, Du S, Zhang W, Rao Y. Conditional chemoconnectomics (cCCTomics) as a strategy for efficient and conditional targeting of chemical transmission. eLife 2024; 12:RP91927. [PMID: 38686992 PMCID: PMC11060718 DOI: 10.7554/elife.91927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
Dissection of neural circuitry underlying behaviors is a central theme in neurobiology. We have previously proposed the concept of chemoconnectome (CCT) to cover the entire chemical transmission between neurons and target cells in an organism and created tools for studying it (CCTomics) by targeting all genes related to the CCT in Drosophila. Here we have created lines targeting the CCT in a conditional manner after modifying GFP RNA interference, Flp-out, and CRISPR/Cas9 technologies. All three strategies have been validated to be highly effective, with the best using chromatin-peptide fused Cas9 variants and scaffold optimized sgRNAs. As a proof of principle, we conducted a comprehensive intersection analysis of CCT genes expression profiles in the clock neurons, uncovering 43 CCT genes present in clock neurons. Specific elimination of each from clock neurons revealed that loss of the neuropeptide CNMa in two posterior dorsal clock neurons (DN1ps) or its receptor (CNMaR) caused advanced morning activity, indicating a suppressive role of CNMa-CNMaR on morning anticipation, opposite to the promoting role of PDF-PDFR on morning anticipation. These results demonstrate the effectiveness of conditional CCTomics and its tools created here and establish an antagonistic relationship between CNMa-CNMaR and PDF-PDFR signaling in regulating morning anticipation.
Collapse
Affiliation(s)
- Renbo Mao
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
- National Institute of Biological Sciences, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Jianjun Yu
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| | - Bowen Deng
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| | - Xihuimin Dai
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| | - Yuyao Du
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| | - Sujie Du
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| | - Wenxia Zhang
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| | - Yi Rao
- Laboratory of Neurochemical Biology, Chinese Institute for Brain ResearchBeijingChina
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes for Medical Research, Capital Medical University; Changping LaboratoryChangpingChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
14
|
Le JQ, Ma D, Dai X, Rosbash M. Light and dopamine impact two circadian neurons to promote morning wakefulness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583333. [PMID: 38496661 PMCID: PMC10942368 DOI: 10.1101/2024.03.04.583333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
In both mammals and flies, circadian brain neurons orchestrate physiological oscillations and behaviors like wake and sleep; these neurons can be subdivided by morphology and by gene expression patterns. Recent single-cell sequencing studies identified 17 Drosophila circadian neuron groups. One of these include only two lateral neurons (LNs), which are marked by the expression of the neuropeptide ion transport peptide (ITP). Although these two ITP+ LNs have long been grouped with five other circadian evening activity cells, inhibiting the two neurons alone strongly reduces morning activity; this indicates that they are prominent morning neurons. As dopamine signaling promotes activity in Drosophila like in mammals, we considered that dopamine might influence this morning activity function. Moreover, the ITP+ LNs express higher mRNA levels than other LNs of the type 1-like dopamine receptor Dop1R1. Consistent with the importance of Dop1R1, CRISPR/Cas9 mutagenesis of this receptor only in the two ITP+ LNs renders flies significantly less active in the morning, and ex vivo live imaging shows that dopamine increases cAMP levels in these two neurons; cell-specific mutagenesis of Dop1R1 eliminates this cAMP response to dopamine. Notably, the response is more robust in the morning, reflecting higher morning Dop1R1 mRNA levels in the two neurons. As morning levels are not elevated in constant darkness, this suggests light-dependent upregulation of morning Dop1R1 transcript levels. Taken together with enhanced morning cAMP response to dopamine, the data indicate how light stimulates morning wakefulness in flies, which mimics the important effect of light on morning wakefulness in humans.
Collapse
Affiliation(s)
- Jasmine Quynh Le
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| | - Dingbang Ma
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Xihuimin Dai
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| |
Collapse
|
15
|
Ryvkin J, Omesi L, Kim YK, Levi M, Pozeilov H, Barak-Buchris L, Agranovich B, Abramovich I, Gottlieb E, Jacob A, Nässel DR, Heberlein U, Shohat-Ophir G. Failure to mate enhances investment in behaviors that may promote mating reward and impairs the ability to cope with stressors via a subpopulation of Neuropeptide F receptor neurons. PLoS Genet 2024; 20:e1011054. [PMID: 38236837 PMCID: PMC10795991 DOI: 10.1371/journal.pgen.1011054] [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: 08/01/2023] [Accepted: 11/06/2023] [Indexed: 01/22/2024] Open
Abstract
Living in dynamic environments such as the social domain, where interaction with others determines the reproductive success of individuals, requires the ability to recognize opportunities to obtain natural rewards and cope with challenges that are associated with achieving them. As such, actions that promote survival and reproduction are reinforced by the brain reward system, whereas coping with the challenges associated with obtaining these rewards is mediated by stress-response pathways, the activation of which can impair health and shorten lifespan. While much research has been devoted to understanding mechanisms underlying the way by which natural rewards are processed by the reward system, less attention has been given to the consequences of failure to obtain a desirable reward. As a model system to study the impact of failure to obtain a natural reward, we used the well-established courtship suppression paradigm in Drosophila melanogaster as means to induce repeated failures to obtain sexual reward in male flies. We discovered that beyond the known reduction in courtship actions caused by interaction with non-receptive females, repeated failures to mate induce a stress response characterized by persistent motivation to obtain the sexual reward, reduced male-male social interaction, and enhanced aggression. This frustrative-like state caused by the conflict between high motivation to obtain sexual reward and the inability to fulfill their mating drive impairs the capacity of rejected males to tolerate stressors such as starvation and oxidative stress. We further show that sensitivity to starvation and enhanced social arousal is mediated by the disinhibition of a small population of neurons that express receptors for the fly homologue of neuropeptide Y. Our findings demonstrate for the first time the existence of social stress in flies and offers a framework to study mechanisms underlying the crosstalk between reward, stress, and reproduction in a simple nervous system that is highly amenable to genetic manipulation.
Collapse
Affiliation(s)
- Julia Ryvkin
- The Mina & Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center and the Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Liora Omesi
- The Mina & Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center and the Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Yong-Kyu Kim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | - Mali Levi
- The Mina & Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center and the Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Hadar Pozeilov
- The Mina & Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center and the Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Lital Barak-Buchris
- The Mina & Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center and the Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Bella Agranovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Ifat Abramovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Eyal Gottlieb
- Ruth and Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Avi Jacob
- The Kanbar scientific equipment center. The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Dick R. Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Ulrike Heberlein
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | - Galit Shohat-Ophir
- The Mina & Everard Goodman Faculty of Life Sciences, The Leslie and Susan Gonda Multidisciplinary Brain Research Center and the Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
16
|
Yadav RSP, Ansari F, Bera N, Kent C, Agrawal P. Lessons from lonely flies: Molecular and neuronal mechanisms underlying social isolation. Neurosci Biobehav Rev 2024; 156:105504. [PMID: 38061597 DOI: 10.1016/j.neubiorev.2023.105504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/26/2023]
Abstract
Animals respond to changes in the environment which affect their internal state by adapting their behaviors. Social isolation is a form of passive environmental stressor that alters behaviors across animal kingdom, including humans, rodents, and fruit flies. Social isolation is known to increase violence, disrupt sleep and increase depression leading to poor mental and physical health. Recent evidences from several model organisms suggest that social isolation leads to remodeling of the transcriptional and epigenetic landscape which alters behavioral outcomes. In this review, we explore how manipulating social experience of fruit fly Drosophila melanogaster can shed light on molecular and neuronal mechanisms underlying isolation driven behaviors. We discuss the recent advances made using the powerful genetic toolkit and behavioral assays in Drosophila to uncover role of neuromodulators, sensory modalities, pheromones, neuronal circuits and molecular mechanisms in mediating social isolation. The insights gained from these studies could be crucial for developing effective therapeutic interventions in future.
Collapse
Affiliation(s)
- R Sai Prathap Yadav
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India
| | - Faizah Ansari
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India
| | - Neha Bera
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India
| | - Clement Kent
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Pavan Agrawal
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India.
| |
Collapse
|
17
|
Sato K, Yamamoto D. Molecular and cellular origins of behavioral sex differences: a tiny little fly tells a lot. Front Mol Neurosci 2023; 16:1284367. [PMID: 37928065 PMCID: PMC10622783 DOI: 10.3389/fnmol.2023.1284367] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
Behavioral sex differences primarily derive from the sexually dimorphic organization of neural circuits that direct the behavior. In Drosophila melanogaster, the sex-determination genes fruitless (fru) and doublesex (dsx) play pivotal roles in producing the sexual dimorphism of neural circuits for behavior. Here we examine three neural groups expressing fru and/or dsx, i.e., the P1 cluster, aSP-f and aSP-g cluster pairs and aDN cluster, in which causal relationships between the dimorphic behavior and dimorphic neural characteristics are best illustrated. aSP-f, aSP-g and aDN clusters represent examples where fru or dsx switches cell-autonomously their neurite structures between the female-type and male-type. Processed sensory inputs impinging on these neurons may result in outputs that encode different valences, which culminate in the execution of distinct behavior according to the sex. In contrast, the P1 cluster is male-specific as its female counterpart undergoes dsx-driven cell death, which lowers the threshold for the induction of male-specific behaviors. We propose that the products of fru and dsx genes, as terminal selectors in sexually dimorphic neuronal wiring, induce and maintain the sex-typical chromatin state at postembryonic stages, orchestrating the transcription of effector genes that shape single neuron structures and govern cell survival and death.
Collapse
Affiliation(s)
- Kosei Sato
- Neuro-ICT Laboratory, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| | - Daisuke Yamamoto
- Neuro-ICT Laboratory, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| |
Collapse
|
18
|
Yu Z, Shi J, Jiang X, Song Y, Du J, Zhao Z. Neuropeptide F regulates feeding via the juvenile hormone pathway in Ostrinia furnacalis larvae. PEST MANAGEMENT SCIENCE 2023; 79:1193-1203. [PMID: 36396604 DOI: 10.1002/ps.7289] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Feeding by pests is one of the most important reasons for reductions in agricultural crop yield. This study aimed to reveal how juvenile hormone (JH) participates in larval feeding regulation of the Asian corn borer Ostrinia furnacalis. RESULTS Larvae of O. furnacalis exhibit a daily circadian feeding rhythm, with a peak at ZT18 and a trough at ZT6 under both photoperiod (LD) and constant dark (DD) conditions, which may be eliminated by application of fenoxycarb, a JH active analogue. JH negatively regulates larval feeding as a downstream factor of neuropeptide F (NPF), in which knocking down JH increases larval feeding amount along with body weight and length. The production of JH in the brain-corpora cardiaca-corpora allata (brain-CC-CA) is regulated by brain NPF rather than gut NPF, which was demonstrated in Drosophila larvae through GAL4/UAS genetic analysis. In addition, feeding regulation of JH is closely related to energy homeostasis in the fat body by inhibiting energy storage and promoting degradation. The JH analogue fenoxycarb is an effective pesticide against O. furnacalis, controlling feeding and metabolism. CONCLUSION The brain NPF system regulates JH, with functions in food consumption, feeding rhythms, energy homeostasis and body size. This study provides an important basis for understanding the feeding mechanism and potential pest control of O. furnacalis. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhuofan Yu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Jian Shi
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Xuemin Jiang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Yu Song
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Juan Du
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Zhangwu Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
19
|
Chen SL, Liu BT, Lee WP, Liao SB, Deng YB, Wu CL, Ho SM, Shen BX, Khoo GH, Shiu WC, Chang CH, Shih HW, Wen JK, Lan TH, Lin CC, Tsai YC, Tzeng HF, Fu TF. WAKE-mediated modulation of cVA perception via a hierarchical neuro-endocrine axis in Drosophila male-male courtship behaviour. Nat Commun 2022; 13:2518. [PMID: 35523813 PMCID: PMC9076693 DOI: 10.1038/s41467-022-30165-2] [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: 03/12/2021] [Accepted: 04/19/2022] [Indexed: 12/18/2022] Open
Abstract
The nervous and endocrine systems coordinate with each other to closely influence physiological and behavioural responses in animals. Here we show that WAKE (encoded by wide awake, also known as wake) modulates membrane levels of GABAA receptor Resistance to Dieldrin (Rdl), in insulin-producing cells of adult male Drosophila melanogaster. This results in changes to secretion of insulin-like peptides which is associated with changes in juvenile hormone biosynthesis in the corpus allatum, which in turn leads to a decrease in 20-hydroxyecdysone levels. A reduction in ecdysone signalling changes neural architecture and lowers the perception of the male-specific sex pheromone 11-cis-vaccenyl acetate by odorant receptor 67d olfactory neurons. These finding explain why WAKE-deficient in Drosophila elicits significant male-male courtship behaviour. The authors show that the Drosophila master regulator WAKE modulates the secretion of insulin-like peptides, triggering a decrease in 20-hydroxyecdysone levels. This lowers the perception of a male-specific sex pheromone and explains why WAKE-deficient Drosophila flies show male-male courtship behaviour.
Collapse
Affiliation(s)
- Shiu-Ling Chen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Bo-Ting Liu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Wang-Pao Lee
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Sin-Bo Liao
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.,Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yao-Bang Deng
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Shuk-Man Ho
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Bing-Xian Shen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Guan-Hock Khoo
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan
| | - Wei-Chiang Shiu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chih-Hsuan Chang
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan.,Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Hui-Wen Shih
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan
| | - Jung-Kun Wen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Tsuo-Hung Lan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Tsaotun Psychiatric Center, Ministry of Health and Welfare, Nantou, Taiwan.,Department of Psychiatry, Taichung Veterans General Hospital, Taichung, Taiwan.,Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chih-Chien Lin
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chen Tsai
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan.
| | - Huey-Fen Tzeng
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| | - Tsai-Feng Fu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| |
Collapse
|
20
|
Suzuki Y, Kurata Y, Sakai T. Dorsal‐lateral clock neurons modulate consolidation and maintenance of long‐term memory in
Drosophila. Genes Cells 2022; 27:266-279. [DOI: 10.1111/gtc.12923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Yuki Suzuki
- Department of Biological Sciences Tokyo Metropolitan University Tokyo 192‐0397 Japan
| | - Yuto Kurata
- Department of Biological Sciences Tokyo Metropolitan University Tokyo 192‐0397 Japan
| | - Takaomi Sakai
- Department of Biological Sciences Tokyo Metropolitan University Tokyo 192‐0397 Japan
| |
Collapse
|
21
|
Bhat US, Shahi N, Surendran S, Babu K. Neuropeptides and Behaviors: How Small Peptides Regulate Nervous System Function and Behavioral Outputs. Front Mol Neurosci 2021; 14:786471. [PMID: 34924955 PMCID: PMC8674661 DOI: 10.3389/fnmol.2021.786471] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
One of the reasons that most multicellular animals survive and thrive is because of the adaptable and plastic nature of their nervous systems. For an organism to survive, it is essential for the animal to respond and adapt to environmental changes. This is achieved by sensing external cues and translating them into behaviors through changes in synaptic activity. The nervous system plays a crucial role in constantly evaluating environmental cues and allowing for behavioral plasticity in the organism. Multiple neurotransmitters and neuropeptides have been implicated as key players for integrating sensory information to produce the desired output. Because of its simple nervous system and well-established neuronal connectome, C. elegans acts as an excellent model to understand the mechanisms underlying behavioral plasticity. Here, we critically review how neuropeptides modulate a wide range of behaviors by allowing for changes in neuronal and synaptic signaling. This review will have a specific focus on feeding, mating, sleep, addiction, learning and locomotory behaviors in C. elegans. With a view to understand evolutionary relationships, we explore the functions and associated pathophysiology of C. elegans neuropeptides that are conserved across different phyla. Further, we discuss the mechanisms of neuropeptidergic signaling and how these signals are regulated in different behaviors. Finally, we attempt to provide insight into developing potential therapeutics for neuropeptide-related disorders.
Collapse
Affiliation(s)
- Umer Saleem Bhat
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Navneet Shahi
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Siju Surendran
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Kavita Babu
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
22
|
Zhu D, Ge J, Guo S, Hou L, Shi R, Zhou X, Nie X, Wang X. Independent variations in genome-wide expression, alternative splicing, and DNA methylation in brain tissues among castes of the buff-tailed bumblebee, Bombus terrestris. J Genet Genomics 2021; 48:681-694. [PMID: 34315685 DOI: 10.1016/j.jgg.2021.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 10/21/2022]
Abstract
Caste differentiation in social hymenopterans is an intriguing example of phenotypic plasticity. However, the co-ordination among gene regulatory factors to mediate caste differentiation remains inconclusive. In this study, we determined the role of gene regulation and related epigenetic processes in pre-imaginal caste differentiation in the primitively eusocial bumblebee Bombus terrestris. By combining RNA-Seq data from Illumina and PacBio and accurately quantifying methylation at whole-genomic base pair resolution, we found that queens, workers, and drones mainly differentiate in gene expression but not in alternative splicing and DNA methylation. Gynes are the most distinct with the lowest global level of whole-genomic methylation and with the largest number of caste-specific transcripts and alternative splicing events. By contrast, workers exhibit few uniquely expressed or alternatively spliced genes. Moreover, several genes involved in hormone and neurotransmitter metabolism are related to caste differentiation, whereas several neuropeptides are linked with sex differentiation. Despite little genome-wide association among differential gene expression, splicing, and differential DNA methylation, the overlapped gene ontology (GO) terms point to nutrition-related activity. Therefore, variations in gene regulation correlate with the behavioral differences among castes and highlight the specialization of toolkit genes in bumblebee gynes at the beginning of the adult stage.
Collapse
Affiliation(s)
- Dan Zhu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China; CAS Centre for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Ge
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China; CAS Centre for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyuan Guo
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China; CAS Centre for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Hou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China; CAS Centre for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rangjun Shi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China; CAS Centre for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xian Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China; CAS Centre for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Nie
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China
| | - Xianhui Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, China; CAS Centre for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
23
|
|
24
|
Yoshinari Y, Kosakamoto H, Kamiyama T, Hoshino R, Matsuoka R, Kondo S, Tanimoto H, Nakamura A, Obata F, Niwa R. The sugar-responsive enteroendocrine neuropeptide F regulates lipid metabolism through glucagon-like and insulin-like hormones in Drosophila melanogaster. Nat Commun 2021; 12:4818. [PMID: 34376687 PMCID: PMC8355161 DOI: 10.1038/s41467-021-25146-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/24/2021] [Indexed: 02/08/2023] Open
Abstract
The enteroendocrine cell (EEC)-derived incretins play a pivotal role in regulating the secretion of glucagon and insulins in mammals. Although glucagon-like and insulin-like hormones have been found across animal phyla, incretin-like EEC-derived hormones have not yet been characterised in invertebrates. Here, we show that the midgut-derived hormone, neuropeptide F (NPF), acts as the sugar-responsive, incretin-like hormone in the fruit fly, Drosophila melanogaster. Secreted NPF is received by NPF receptor in the corpora cardiaca and in insulin-producing cells. NPF-NPFR signalling resulted in the suppression of the glucagon-like hormone production and the enhancement of the insulin-like peptide secretion, eventually promoting lipid anabolism. Similar to the loss of incretin function in mammals, loss of midgut NPF led to significant metabolic dysfunction, accompanied by lipodystrophy, hyperphagia, and hypoglycaemia. These results suggest that enteroendocrine hormones regulate sugar-dependent metabolism through glucagon-like and insulin-like hormones not only in mammals but also in insects.
Collapse
Affiliation(s)
- Yuto Yoshinari
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hina Kosakamoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Takumi Kamiyama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Ryo Hoshino
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Rena Matsuoka
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shu Kondo
- Genetic Strains Research Center, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Akira Nakamura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory of Germline Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Fumiaki Obata
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- AMED-PRIME, Japan Agency for Medical Research and Development Chiyoda-ku, Tokyo, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan.
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan.
| |
Collapse
|
25
|
Liu B, Fu D, Gao H, Ning H, Sun Y, Chen H, Tang M. Cloning and Expression of the Neuropeptide F and Neuropeptide F Receptor Genes and Their Regulation of Food Intake in the Chinese White Pine Beetle Dendroctonus armandi. Front Physiol 2021; 12:662651. [PMID: 34220532 PMCID: PMC8249871 DOI: 10.3389/fphys.2021.662651] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/12/2021] [Indexed: 01/31/2023] Open
Abstract
Neuropeptide F (NPF) is an important signaling molecule that acts as a neuromodulator to regulate a diversity of physiological and behavioral processes from vertebrates to invertebrates by interaction with NPF receptors, which are G protein-coupled receptors (GPCR). However, nothing is known about NPF in Chinese white pine beetle, Dendroctonus armandi, a destructive pest of natural and coniferous forests in the middle Qinling Mountains of China. We have cloned and characterized cDNAs encoding one NPF precursor and two NPF receptors in D. armandi and made bioinformatics predictions according to the deduced amino acid sequences. They were highly similar to that of Dendroctonus ponderosa. The transcription levels of these genes were different between larvae and adults of sexes, and there were significant differences among the different developmental stages and tissues and between beetles under starvation and following re-feeding states. Additionally, downregulation of NPF and NPFR by injecting dsRNA into beetles reduced their food intake, caused increases of mortality and decreases of body weight, and also resulted in a decrease of glycogen and free fatty acid and an increase of trehalose. These results indicate that the NPF signaling pathway plays a significant positive role in the regulation of food intake and provides a potential target for the sustainable management of this pest.
Collapse
Affiliation(s)
- Bin Liu
- College of Forestry, Northwest A&F University, Xianyang, China
| | - Danyang Fu
- College of Forestry, Northwest A&F University, Xianyang, China
| | - Haiming Gao
- College of Forestry, Northwest A&F University, Xianyang, China
| | - Hang Ning
- College of Forestry, Northwest A&F University, Xianyang, China
| | - Yaya Sun
- College of Forestry, Northwest A&F University, Xianyang, China
| | - Hui Chen
- College of Forestry, Northwest A&F University, Xianyang, China.,State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Key Laboratory for Innovative Development and Utilization of Forest Plant Germplasm, College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou, China
| | - Ming Tang
- College of Forestry, Northwest A&F University, Xianyang, China.,State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Key Laboratory for Innovative Development and Utilization of Forest Plant Germplasm, College of Forestry and Landscape Architecture, South China Agricultural University, Guangzhou, China
| |
Collapse
|
26
|
Zhang SX, Glantz EH, Miner LE, Rogulja D, Crickmore MA. Hormonal control of motivational circuitry orchestrates the transition to sexuality in Drosophila. SCIENCE ADVANCES 2021; 7:eabg6926. [PMID: 34134981 PMCID: PMC8208730 DOI: 10.1126/sciadv.abg6926] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/04/2021] [Indexed: 06/12/2023]
Abstract
Newborns and hatchlings can perform incredibly sophisticated behaviors, but many animals abstain from sexual activity at the beginning of life. Hormonal changes have long been known to drive both physical and behavioral changes during adolescence, leading to the largely untested assumption that sexuality emerges from organizational changes to neuronal circuitry. We show that the transition to sexuality in male Drosophila is controlled by hormonal changes, but this regulation is functional rather than structural. In very young males, a broadly acting hormone directly inhibits the activity of three courtship-motivating circuit elements, ensuring the complete suppression of sexual motivation and behavior. Blocking or overriding these inhibitory mechanisms evokes immediate and robust sexual behavior from very young and otherwise asexual males. Similarities to mammalian adolescence suggest a general principle in which hormonal changes gate the transition to sexuality not by constructing new circuitry but by permitting activity in otherwise latent motivational circuit elements.
Collapse
Affiliation(s)
- Stephen X Zhang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Ethan H Glantz
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lauren E Miner
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dragana Rogulja
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Michael A Crickmore
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Abstract
Circadian clocks are biochemical time-keeping machines that synchronize animal behavior and physiology with planetary rhythms. In Drosophila, the core components of the clock comprise a transcription/translation feedback loop and are expressed in seven neuronal clusters in the brain. Although it is increasingly evident that the clocks in each of the neuronal clusters are regulated differently, how these clocks communicate with each other across the circadian neuronal network is less clear. Here, we review the latest evidence that describes the physical connectivity of the circadian neuronal network . Using small ventral lateral neurons as a starting point, we summarize how one clock may communicate with another, highlighting the signaling pathways that are both upstream and downstream of these clocks. We propose that additional efforts are required to understand how temporal information generated in each circadian neuron is integrated across a neuronal circuit to regulate rhythmic behavior.
Collapse
Affiliation(s)
- Myra Ahmad
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Wanhe Li
- Laboratory of Genetics, The Rockefeller University, New York, NY, USA
| | - Deniz Top
- Department of Pediatrics, Division of Medical Genetics, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
28
|
Sadanandappa MK, Sathyanarayana SH, Kondo S, Bosco G. Neuropeptide F signaling regulates parasitoid-specific germline development and egg-laying in Drosophila. PLoS Genet 2021; 17:e1009456. [PMID: 33770070 PMCID: PMC8026082 DOI: 10.1371/journal.pgen.1009456] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 04/07/2021] [Accepted: 03/01/2021] [Indexed: 01/08/2023] Open
Abstract
Drosophila larvae and pupae are at high risk of parasitoid infection in nature. To circumvent parasitic stress, fruit flies have developed various survival strategies, including cellular and behavioral defenses. We show that adult Drosophila females exposed to the parasitic wasps, Leptopilina boulardi, decrease their total egg-lay by deploying at least two strategies: Retention of fully developed follicles reduces the number of eggs laid, while induction of caspase-mediated apoptosis eliminates the vitellogenic follicles. These reproductive defense strategies require both visual and olfactory cues, but not the MB247-positive mushroom body neuronal function, suggesting a novel mode of sensory integration mediates reduced egg-laying in the presence of a parasitoid. We further show that neuropeptide F (NPF) signaling is necessary for both retaining matured follicles and activating apoptosis in vitellogenic follicles. Whereas previous studies have found that gut-derived NPF controls germ stem cell proliferation, we show that sensory-induced changes in germ cell development specifically require brain-derived NPF signaling, which recruits a subset of NPFR-expressing cell-types that control follicle development and retention. Importantly, we found that reduced egg-lay behavior is specific to parasitic wasps that infect the developing Drosophila larvae, but not the pupae. Our findings demonstrate that female fruit flies use multimodal sensory integration and neuroendocrine signaling via NPF to engage in parasite-specific cellular and behavioral survival strategies.
Collapse
Affiliation(s)
- Madhumala K. Sadanandappa
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Shivaprasad H. Sathyanarayana
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Shu Kondo
- Invertebrate Genetics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Giovanni Bosco
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
29
|
Wan GJ, Jiang SL, Zhang M, Zhao JY, Zhang YC, Pan WD, Sword GA, Chen FJ. Geomagnetic field absence reduces adult body weight of a migratory insect by disrupting feeding behavior and appetite regulation. INSECT SCIENCE 2021; 28:251-260. [PMID: 32065478 DOI: 10.1111/1744-7917.12765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 06/10/2023]
Abstract
The geomagnetic field (GMF) is well documented for its essential role as a cue used in animal orientation or navigation. Recent evidence indicates that the absence of GMF (mimicked by the near-zero magnetic field, NZMF) can trigger stress-like responses such as reduced body weight, as we have previously shown in the brown planthopper, Nilaparvata lugens. In this study, we found that consistent with the significantly decreased body weight of newly emerged female (-14.67%) and male (-13.17%) adult N. lugens, the duration of the phloem ingestion feeding waveform was significantly reduced by 32.02% in 5th instar nymphs reared under the NZMF versus GMF. Interestingly, 5th instar nymphs that exhibited reduced feeding had significantly higher glucose levels (+16.98% and +20.05%; 24 h and 48 h after molting), which are associated with food aversion, and expression patterns of their appetite-related neuropeptide genes (neuropeptide F, down-regulated overall; short neuropeptide F, down-regulated overall; adipokinetic hormone, up-regulated overall; and adipokinetic hormone receptor, down-regulated overall) were also altered under the absence of GMF in a manner consistent with diminishing appetite. Moreover, the expressions of the potential magnetosensor cryptochromes (Crys) were found significantly altered under the absence of GMF, indicating the likely upstream signaling of the Cry-mediated magnetoreception mechanisms. These findings support the hypothesis that strong changes in GMF intensity can reduce adult body weight through affecting insect feeding behavior and underlying regulatory processes including appetite regulation. Our results highlight that GMF could be necessary for the maintenance of energy homeostasis in insects.
Collapse
Affiliation(s)
- Gui-Jun Wan
- Department of Entomology, Nanjing Agricultural University, Nanjing, China
| | - Shou-Lin Jiang
- Department of Entomology, Nanjing Agricultural University, Nanjing, China
| | - Ming Zhang
- Department of Entomology, Nanjing Agricultural University, Nanjing, China
| | - Jing-Yu Zhao
- Department of Entomology, Nanjing Agricultural University, Nanjing, China
| | - Ying-Chao Zhang
- Beijing Key Laboratory of Bioelectromagetics, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| | - Wei-Dong Pan
- Beijing Key Laboratory of Bioelectromagetics, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| | - Gregory A Sword
- Department of Entomology, Texas A&M University, College Station, TX, U.S.A
| | - Fa-Jun Chen
- Department of Entomology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
30
|
Ma D, Przybylski D, Abruzzi KC, Schlichting M, Li Q, Long X, Rosbash M. A transcriptomic taxonomy of Drosophila circadian neurons around the clock. eLife 2021; 10:63056. [PMID: 33438579 PMCID: PMC7837698 DOI: 10.7554/elife.63056] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Many different functions are regulated by circadian rhythms, including those orchestrated by discrete clock neurons within animal brains. To comprehensively characterize and assign cell identity to the 75 pairs of Drosophila circadian neurons, we optimized a single-cell RNA sequencing method and assayed clock neuron gene expression at different times of day. The data identify at least 17 clock neuron categories with striking spatial regulation of gene expression. Transcription factor regulation is prominent and likely contributes to the robust circadian oscillation of many transcripts, including those that encode cell-surface proteins previously shown to be important for cell recognition and synapse formation during development. The many other clock-regulated genes also constitute an important resource for future mechanistic and functional studies between clock neurons and/or for temporal signaling to circuits elsewhere in the fly brain.
Collapse
Affiliation(s)
- Dingbang Ma
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Dariusz Przybylski
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Katharine C Abruzzi
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | | | - Qunlong Li
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| | - Xi Long
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, United States
| | - Michael Rosbash
- Howard Hughes Medical Institute, Brandeis University, Waltham, United States
| |
Collapse
|
31
|
Qiao H, Jiang S, Xiong Y, Zhang W, Xu L, Jin S, Gong Y, Wu Y, Fu H. Molecular cloning, characterization and functional analysis of two neuropeptide F genes from the oriental river prawn (Macrobrachium nipponense). Comp Biochem Physiol A Mol Integr Physiol 2020; 253:110844. [PMID: 33186705 DOI: 10.1016/j.cbpa.2020.110844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 10/23/2022]
Abstract
In invertebrates, neuropeptide F (NPF) has many regulatory functions, similar to NPY, its homologous peptide. In this study, two neuropeptide F genes were identified in Macrobrachium nipponense: Mn-NPF1 and Mn-NPF2. Mn-NPF2 shared the same amino acid sequence with Mn-NPF1, except for a 37 amino acid insert in the middle of the NPF region. The quantitative-PCR (qPCR) results indicated that Mn-NPF1 expression was positively correlated with ovarian maturation, whereas Mn-NPF2 had opposing expression patterns. Both Mn-NPFs were poorly expressed at early embryonic stages, but enhanced expression levels were observed up to day 10 after hatching, when the gonads began to differentiate. Ovary in situ hybridization (ISH) analyses showed that both Mn-NPFs were present at all stages, but were differentially localized to distinct compartments. Temperature gradient studies showed that both Mn-NPFs were implicated in the seasonal regulation of reproduction. A double-stranded (ds) RNA-Mn-NPF2 injection led to a significant 38.5% increase in the vitellogenin (VG) transcript (P < 0.05). These results demonstrated that Mn-NPF2 plays an important role in inhibiting ovarian maturation.
Collapse
Affiliation(s)
- Hui Qiao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China
| | - Sufei Jiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China
| | - Yiwei Xiong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China
| | - Wenyi Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China
| | - Lei Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu, People's Republic of China
| | - Shubo Jin
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China
| | - Yongsheng Gong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China
| | - Yan Wu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China
| | - Hongtuo Fu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, Jiangsu, People's Republic of China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, Jiangsu, People's Republic of China.
| |
Collapse
|
32
|
Zeng H, Qin Y, Du E, Wei Q, Li Y, Huang D, Wang G, Veenstra JA, Li S, Li N. Genomics- and Peptidomics-Based Discovery of Conserved and Novel Neuropeptides in the American Cockroach. J Proteome Res 2020; 20:1217-1228. [PMID: 33166158 DOI: 10.1021/acs.jproteome.0c00596] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
As a model hemimetabolous insect species and an invasive urban pest that is globally distributed, the American cockroach, Periplaneta americana, is of great interest in both basic and applied research. Previous studies on P. americana neuropeptide identification have been based on biochemical isolation and molecular cloning. In the present study, an integrated approach of genomics- and peptidomics-based discovery was performed for neuropeptide identification in this insect species. First, 67 conserved neuropeptide or neurohormone precursor genes were predicted via an in silico analysis of the P. americana genome and transcriptome. Using a large-scale peptidomic analysis of peptide extracts from four different tissues (the central nervous system, corpora cardiac and corpora allata complex, midgut, and male accessory gland), 35 conserved (predicted) neuropeptides and a potential (novel) neuropeptide were then identified. Subsequent experiments revealed the tissue distribution, sex difference, and developmental patterns of two conserved neuropeptides (allatostatin B and short neuropeptide F) and a novel neuropeptide (PaOGS36577). Our study shows a comprehensive neuropeptidome and detailed spatiotemporal distribution patterns, providing a solid basis for future functional studies of neuropeptides in the American cockroach (data are available via ProteomeXchange with identifier PXD021660).
Collapse
Affiliation(s)
- Huanchao Zeng
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.,Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| | - Yiru Qin
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.,Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| | - Erxia Du
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.,Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| | - Qiulan Wei
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Danyan Huang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Guirong Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Jan A Veenstra
- INCIA, UMR 5287 CNRS, Université de Bordeaux, Pessac F33615, France
| | - Sheng Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.,Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| | - Na Li
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.,Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, South China Normal University, Meizhou 514779, China
| |
Collapse
|
33
|
Winters GC, Polese G, Di Cosmo A, Moroz LL. Mapping of neuropeptide Y expression in Octopus brains. J Morphol 2020; 281:790-801. [PMID: 32384206 DOI: 10.1002/jmor.21141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/31/2022]
Abstract
Neuropeptide Y (NPY) is an evolutionarily conserved neurosecretory molecule implicated in a diverse complement of functions across taxa and in regulating feeding behavior and reproductive maturation in Octopus. However, little is known about the precise molecular circuitry of NPY-mediated behaviors and physiological processes, which likely involve a complex interaction of multiple signal molecules in specific brain regions. Here, we examined the expression of NPY throughout the Octopus central nervous system. The sequence analysis of Octopus NPY precursor confirmed the presence of both, signal peptide and putative active peptides, which are highly conserved across bilaterians. In situ hybridization revealed distinct expression of NPY in specialized compartments, including potential "integration centers," where visual, tactile, and other behavioral circuitries converge. These centers integrating separate circuits may maintain and modulate learning and memory or other behaviors not yet attributed to NPY-dependent modulation in Octopus. Extrasomatic localization of NPY mRNA in the neurites of specific neuron populations in the brain suggests a potential demand for immediate translation at synapses and a crucial temporal role for NPY in these cell populations. We also documented the presence of NPY mRNA in a small cell population in the olfactory lobe, which is a component of the Octopus feeding and reproductive control centers. However, the molecular mapping of NPY expression only partially overlapped with that produced by immunohistochemistry in previous studies. Our study provides a precise molecular map of NPY mRNA expression that can be used to design and test future hypotheses about molecular signaling in various Octopus behaviors.
Collapse
Affiliation(s)
- Gabrielle C Winters
- Department of Neuroscience and McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Gianluca Polese
- Department of Biology, Di Cosmo Laboratory, University of Napoli Federico II, Naples, Italy
| | - Anna Di Cosmo
- Department of Biology, Di Cosmo Laboratory, University of Napoli Federico II, Naples, Italy
| | - Leonid L Moroz
- Department of Neuroscience and McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.,Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, Florida, USA
| |
Collapse
|
34
|
Toprak U. The Role of Peptide Hormones in Insect Lipid Metabolism. Front Physiol 2020; 11:434. [PMID: 32457651 PMCID: PMC7221030 DOI: 10.3389/fphys.2020.00434] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Lipids are the primary storage molecules and an essential source of energy in insects during reproduction, prolonged periods of flight, starvation, and diapause. The coordination center for insect lipid metabolism is the fat body, which is analogous to the vertebrate adipose tissue and liver. The fat body is primarily composed of adipocytes, which accumulate triacylglycerols in intracellular lipid droplets. Genomics and proteomics, together with functional analyses, such as RNA interference and CRISPR/Cas9-targeted genome editing, identified various genes involved in lipid metabolism and elucidated their functions. However, the endocrine control of insect lipid metabolism, in particular the roles of peptide hormones in lipogenesis and lipolysis are relatively less-known topics. In the current review, the neuropeptides that directly or indirectly affect insect lipid metabolism are introduced. The primary lipolytic and lipogenic peptide hormones are adipokinetic hormone and the brain insulin-like peptides (ILP2, ILP3, ILP5). Other neuropeptides, such as insulin-growth factor ILP6, neuropeptide F, allatostatin-A, corazonin, leucokinin, tachykinins and limostatin, might stimulate lipolysis, while diapause hormone-pheromone biosynthesis activating neuropeptide, short neuropeptide F, CCHamide-2, and the cytokines Unpaired 1 and Unpaired 2 might induce lipogenesis. Most of these peptides interact with one another, but mostly with insulin signaling, and therefore affect lipid metabolism indirectly. Peptide hormones are also involved in lipid metabolism during reproduction, flight, diapause, starvation, infections and immunity; these are also highlighted. The review concludes with a discussion of the potential of lipid metabolism-related peptide hormones in pest management.
Collapse
Affiliation(s)
- Umut Toprak
- Molecular Entomology Lab., Department of Plant Protection Ankara, Faculty of Agriculture, Ankara University, Ankara, Turkey
| |
Collapse
|
35
|
CCAP regulates feeding behavior via the NPF pathway in Drosophila adults. Proc Natl Acad Sci U S A 2020; 117:7401-7408. [PMID: 32179671 DOI: 10.1073/pnas.1914037117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The intake of macronutrients is crucial for the fitness of any animal and is mainly regulated by peripheral signals to the brain. How the brain receives and translates these peripheral signals or how these interactions lead to changes in feeding behavior is not well-understood. We discovered that 2 crustacean cardioactive peptide (CCAP)-expressing neurons in Drosophila adults regulate feeding behavior and metabolism. Notably, loss of CCAP, or knocking down the CCAP receptor (CCAP-R) in 2 dorsal median neurons, inhibits the release of neuropeptide F (NPF), which regulates feeding behavior. Furthermore, under starvation conditions, flies normally have an increased sensitivity to sugar; however, loss of CCAP, or CCAP-R in 2 dorsal median NPF neurons, inhibited sugar sensitivity in satiated and starved flies. Separate from its regulation of NPF signaling, the CCAP peptide also regulates triglyceride levels. Additionally, genetic and optogenetic studies demonstrate that CCAP signaling is necessary and sufficient to stimulate a reflexive feeding behavior, the proboscis extension reflex (PER), elicited when external food cues are interpreted as palatable. Dopaminergic signaling was also sufficient to induce a PER. On the other hand, although necessary, NPF neurons were not able to induce a PER. These data illustrate that the CCAP peptide is a central regulator of feeding behavior and metabolism in adult flies, and that NPF neurons have an important regulatory role within this system.
Collapse
|
36
|
Homodimerization of Drosophila Class A neuropeptide GPCRs: Evidence for conservation of GPCR dimerization throughout metazoan evolution. Biochem Biophys Res Commun 2020; 523:322-327. [PMID: 31864711 DOI: 10.1016/j.bbrc.2019.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022]
Abstract
While many instances of GPCR dimerization have been reported for vertebrate receptors, invertebrate GPCR dimerization remains poorly investigated, with few invertebrate GPCRs having been shown to assemble as dimers. To date, no Drosophila GPCRs have been shown to assemble as dimers. To explore the evolutionary conservation of GPCR dimerization, we employed an acceptor-photobleaching FRET methodology to evaluate whether multiple subclasses of Drosophila GPCRs assembled as homodimers when heterologously expressed in HEK-293 T cells. We C-terminally tagged multiple Drosophila neuropeptide GPCRs that exhibited structural homology with a vertebrate GPCR family member previously shown to assemble as a dimer with CFP and YFP fluorophores and visualized these receptors through confocal microscopy. FRET responses were determined based on the increase in CFP emission intensity following YFP photobleaching for each receptor pair tested. A significant FRET response was observed for each receptor expressed as a homodimer pair, while non-significant FRET responses were displayed by both cytosolic CFP and YFP expressed alone, and a heterodimeric pair of receptors from unrelated families. These findings suggest that receptors exhibiting positive FRET responses assemble as homodimers at the plasma membrane and are the first to suggest that Drosophila GPCRs assemble as homodimeric complexes. We propose that GPCR dimerization arose early in metazoan evolution and likely plays an important and underappreciated role in the cellular signaling of all animals.
Collapse
|
37
|
Liu W, Ganguly A, Huang J, Wang Y, Ni JD, Gurav AS, Aguilar MA, Montell C. Neuropeptide F regulates courtship in Drosophila through a male-specific neuronal circuit. eLife 2019; 8:e49574. [PMID: 31403399 PMCID: PMC6721794 DOI: 10.7554/elife.49574] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/07/2019] [Indexed: 12/02/2022] Open
Abstract
Male courtship is provoked by perception of a potential mate. In addition, the likelihood and intensity of courtship are influenced by recent mating experience, which affects sexual drive. Using Drosophila melanogaster, we found that the homolog of mammalian neuropeptide Y, neuropeptide F (NPF), and a cluster of male-specific NPF (NPFM) neurons, regulate courtship through affecting courtship drive. Disrupting NPF signaling produces sexually hyperactive males, which are resistant to sexual satiation, and whose courtship is triggered by sub-optimal stimuli. We found that NPFM neurons make synaptic connections with P1 neurons, which comprise the courtship decision center. Activation of P1 neurons elevates NPFM neuronal activity, which then act through NPF receptor neurons to suppress male courtship, and maintain the proper level of male courtship drive.
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Molecular, Cellular and Developmental Biology, and the Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraUnited States
| | - Anindya Ganguly
- Department of Molecular, Cellular and Developmental Biology, and the Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraUnited States
| | - Jia Huang
- Institute of Insect SciencesZhejiang UniversityHangzhouChina
| | - Yijin Wang
- Department of Molecular, Cellular and Developmental Biology, and the Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraUnited States
| | - Jinfei D Ni
- Department of Molecular, Cellular and Developmental Biology, and the Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraUnited States
| | - Adishthi S Gurav
- Department of Molecular, Cellular and Developmental Biology, and the Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraUnited States
| | - Morris A Aguilar
- Department of Molecular, Cellular and Developmental Biology, and the Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraUnited States
| | - Craig Montell
- Department of Molecular, Cellular and Developmental Biology, and the Neuroscience Research InstituteUniversity of California, Santa BarbaraSanta BarbaraUnited States
| |
Collapse
|
38
|
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
|
39
|
Hou L, Li B, Ding D, Kang L, Wang X. CREB-B acts as a key mediator of NPF/NO pathway involved in phase-related locomotor plasticity in locusts. PLoS Genet 2019; 15:e1008176. [PMID: 31150381 PMCID: PMC6561586 DOI: 10.1371/journal.pgen.1008176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/12/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022] Open
Abstract
Gene expression changes in neural systems are essential for environment-induced behavioral plasticity in animals; however, neuronal signaling pathways mediating the effect of external stimuli on transcriptional changes are largely unknown. Recently, we have demonstrated that the neuropeptide F (NPF)/nitric oxide (NO) signaling pathway plays a regulatory role in phase-related locomotor plasticity in the migratory locust, Locusta migratoria. Here, we report that a conserved transcription factor, cAMP response element-binding protein B (CREB-B), is a key mediator involved in the signaling pathway from NPF2 to NOS in the migratory locust, triggering locomotor activity shift between solitarious and gregarious phases. We find that CREB-B directly activates brain NOS expression by interacting with NOS promoter region. The phosphorylation at serine 110 site of CREB-B dynamically changes in response to population density variation and is negatively controlled by NPF2. The involvement of CREB-B in NPF2-regulated locomotor plasticity is further validated by RNAi experiment and behavioral assay. Furthermore, we reveal that protein kinase A mediates the regulatory effects of NPF2 on CREB-B phosphorylation and NOS transcription. These findings highlight a precise signal cascade underlying environment-induced behavioral plasticity.
Collapse
Affiliation(s)
- Li Hou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Beibei Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ding Ding
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Le Kang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Xianhui Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Nässel DR, Zandawala M. Recent advances in neuropeptide signaling in Drosophila, from genes to physiology and behavior. Prog Neurobiol 2019; 179:101607. [PMID: 30905728 DOI: 10.1016/j.pneurobio.2019.02.003] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
This review focuses on neuropeptides and peptide hormones, the largest and most diverse class of neuroactive substances, known in Drosophila and other animals to play roles in almost all aspects of daily life, as w;1;ell as in developmental processes. We provide an update on novel neuropeptides and receptors identified in the last decade, and highlight progress in analysis of neuropeptide signaling in Drosophila. Especially exciting is the huge amount of work published on novel functions of neuropeptides and peptide hormones in Drosophila, largely due to the rapid developments of powerful genetic methods, imaging techniques and innovative assays. We critically discuss the roles of peptides in olfaction, taste, foraging, feeding, clock function/sleep, aggression, mating/reproduction, learning and other behaviors, as well as in regulation of development, growth, metabolic and water homeostasis, stress responses, fecundity, and lifespan. We furthermore provide novel information on neuropeptide distribution and organization of peptidergic systems, as well as the phylogenetic relations between Drosophila neuropeptides and those of other phyla, including mammals. As will be shown, neuropeptide signaling is phylogenetically ancient, and not only are the structures of the peptides, precursors and receptors conserved over evolution, but also many functions of neuropeptide signaling in physiology and behavior.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden.
| | - Meet Zandawala
- Department of Zoology, Stockholm University, Stockholm, Sweden; Department of Neuroscience, Brown University, Providence, RI, USA.
| |
Collapse
|
41
|
Čižmár D, Roller L, Pillerová M, Sláma K, Žitňan D. Multiple neuropeptides produced by sex-specific neurons control activity of the male accessory glands and gonoducts in the silkworm Bombyx mori. Sci Rep 2019; 9:2253. [PMID: 30783175 PMCID: PMC6381147 DOI: 10.1038/s41598-019-38761-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 12/10/2018] [Indexed: 12/20/2022] Open
Abstract
The male accessory glands (AG) and gonoducts of moths develop during metamorphosis and are essential for successful fertilization of females. We found that these reproductive organs are innervated by a sex-specific cluster of peptidergic neurons in the posterior 9th neuromere of the terminal abdominal ganglion (TAG). This cluster of ~20 neurons differentiate during metamorphosis to innervate the accessory glands and sperm ducts. Using immunohistochemistry and in situ hybridization (ISH) we showed that these neurons express four neuropeptide precursors encoding calcitonin-like diuretic hormone (CT-DH), allatotropin (AT) and AT-like peptides (ATLI-III), allatostatin C (AST-C), and myoinhibitory peptides (MIPs). We used contraction bioassay in vitro to determine roles of these neuropeptides in the gonoduct and accessory gland activity. Spontaneous contractions of the seminal vesicle and AG were stimulated in a dose depended manner by CT-DH and AT, whereas AST-C and MIP elicited dose dependent inhibition. Using quantitative RT-PCR we confirmed expression of receptors for these neuropeptides in organs innervated by the male specific cluster of neurons. Our results suggest a role of these neuropeptides in regulation of seminal fluid movements during copulation.
Collapse
Affiliation(s)
- Daniel Čižmár
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 84506, Bratislava, Slovakia
| | - Ladislav Roller
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 84506, Bratislava, Slovakia
| | - Miriam Pillerová
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 84506, Bratislava, Slovakia
| | - Karel Sláma
- Biology Centre of Czech Academy of Sciences, Institute of Entomology, Drnovská 507, 16100, Praha 6, Czech Republic
| | - Dušan Žitňan
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 84506, Bratislava, Slovakia.
| |
Collapse
|
42
|
Scolari F, Attardo GM, Aksoy E, Weiss B, Savini G, Takac P, Abd-Alla A, Parker AG, Aksoy S, Malacrida AR. Symbiotic microbes affect the expression of male reproductive genes in Glossina m. morsitans. BMC Microbiol 2018; 18:169. [PMID: 30470198 PMCID: PMC6251095 DOI: 10.1186/s12866-018-1289-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Tsetse flies (Diptera, Glossinidae) display unique reproductive biology traits. Females reproduce through adenotrophic viviparity, nourishing the growing larva into their modified uterus until parturition. Males transfer their sperm and seminal fluid, produced by both testes and male accessory glands, in a spermatophore capsule transiently formed within the female reproductive tract upon mating. Both sexes are obligate blood feeders and have evolved tight relationships with endosymbionts, already shown to provide essential nutrients lacking in their diet. However, the partnership between tsetse and its symbionts has so far been investigated, at the molecular, genomic and metabolomics level, only in females, whereas the roles of microbiota in male reproduction are still unexplored. Results Here we begin unravelling the impact of microbiota on Glossina m. morsitans (G. morsitans) male reproductive biology by generating transcriptomes from the reproductive tissues of males deprived of their endosymbionts (aposymbiotic) via maternal antibiotic treatment and dietary supplementation. We then compared the transcriptional profiles of genes expressed in the male reproductive tract of normal and these aposymbiotic flies. We showed that microbiota removal impacts several male reproductive genes by depressing the activity of genes in the male accessory glands (MAGs), including sequences encoding seminal fluid proteins, and increasing expression of genes in the testes. In the MAGs, in particular, the expression of genes related to mating, immunity and seminal fluid components’ synthesis is reduced. In the testes, the absence of symbionts activates genes involved in the metabolic apparatus at the basis of male reproduction, including sperm production, motility and function. Conclusions Our findings mirrored the complementary roles male accessory glands and testes play in supporting male reproduction and open new avenues for disentangling the interplay between male insects and endosymbionts. From an applied perspective, unravelling the metabolic and functional relationships between tsetse symbionts and male reproductive physiology will provide fundamental information useful to understanding the biology underlying improved male reproductive success in tsetse. This information is of particular importance in the context of tsetse population control via Sterile Insect Technique (SIT) and its impact on trypanosomiasis transmission. Electronic supplementary material The online version of this article (10.1186/s12866-018-1289-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francesca Scolari
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Geoffrey Michael Attardo
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, CT, 06520, USA.,Present Address: Department of Entomology and Nematology, University of California Davis, Davis, CA, 95616, USA
| | - Emre Aksoy
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, CT, 06520, USA
| | - Brian Weiss
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, CT, 06520, USA
| | - Grazia Savini
- Department of Biology and Biotechnology, University of Pavia, 27100, Pavia, Italy
| | - Peter Takac
- Section of Molecular and Applied Zoology, Institute of Zoology, Slovak Academy of Sciences, 845 06, Bratislava, SR, Slovakia
| | - Adly Abd-Alla
- International Atomic Energy Agency, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, IPC Laboratory, A-1400, Vienna, Austria
| | - Andrew Gordon Parker
- International Atomic Energy Agency, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, IPC Laboratory, A-1400, Vienna, Austria
| | - Serap Aksoy
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, CT, 06520, USA
| | | |
Collapse
|
43
|
Ameku T, Yoshinari Y, Texada MJ, Kondo S, Amezawa K, Yoshizaki G, Shimada-Niwa Y, Niwa R. Midgut-derived neuropeptide F controls germline stem cell proliferation in a mating-dependent manner. PLoS Biol 2018; 16:e2005004. [PMID: 30248087 PMCID: PMC6152996 DOI: 10.1371/journal.pbio.2005004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 08/20/2018] [Indexed: 01/21/2023] Open
Abstract
Stem cell maintenance is established by neighboring niche cells that promote stem cell self-renewal. However, it is poorly understood how stem cell activity is regulated by systemic, tissue-extrinsic signals in response to environmental cues and changes in physiological status. Here, we show that neuropeptide F (NPF) signaling plays an important role in the pathway regulating mating-induced germline stem cell (GSC) proliferation in the fruit fly Drosophila melanogaster. NPF expressed in enteroendocrine cells (EECs) of the midgut is released in response to the seminal-fluid protein sex peptide (SP) upon mating. This midgut-derived NPF controls mating-induced GSC proliferation via ovarian NPF receptor (NPFR) activity, which modulates bone morphogenetic protein (BMP) signaling levels in GSCs. Our study provides a molecular mechanism that describes how a gut-derived systemic factor couples stem cell behavior to physiological status, such as mating, through interorgan communication. Communication between different organs is essential to respond quickly to environmental cues or changes in the physiological status of an organism. Recent studies have shown the existence of humoral factors or hormones, which are transported by the circulatory system to different organs and achieve coordination between them. Here, we have analyzed the communication mechanism between organs that regulates proliferation of germline stem cells (GSCs) in the ovary of the fruit fly Drosophila melanogaster. We show that a peptide hormone called neuropeptide F (NPF) is a key player in this process. This peptide is produced in both the brain and the midgut, and, remarkably, we find that only NPF released from the midgut is crucial for controlling post-mating GSC proliferation. Our data suggest that mating stimulates the release of NPF from the endocrine cells of the midgut stimulated by the presence of a seminal peptide. Midgut-derived NPF is then transduced through an NPF-specific G-protein–coupled receptor expressed in the ovary, and this triggers GSC proliferation. Our study identifies an essential interaction between the digestive system and the ovary that regulates the size of stem cell populations in flies depending on mating.
Collapse
Affiliation(s)
- Tomotsune Ameku
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yuto Yoshinari
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Michael J Texada
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | - Shu Kondo
- Genetic Strains Research Center, National Institute of Genetics, Mishima, Japan
| | - Kotaro Amezawa
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Goro Yoshizaki
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Yuko Shimada-Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Ryusuke Niwa
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
44
|
Uno T, Ozakiya Y, Furutani M, Sakamoto K, Uno Y, Kajiwara H, Kanamaru K, Mizoguchi A. Functional characterization of insect-specific RabX6 of Bombyx mori. Histochem Cell Biol 2018; 151:187-198. [DOI: 10.1007/s00418-018-1710-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2018] [Indexed: 10/28/2022]
|
45
|
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
|
46
|
Nässel DR. Substrates for Neuronal Cotransmission With Neuropeptides and Small Molecule Neurotransmitters in Drosophila. Front Cell Neurosci 2018; 12:83. [PMID: 29651236 PMCID: PMC5885757 DOI: 10.3389/fncel.2018.00083] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/08/2018] [Indexed: 01/11/2023] Open
Abstract
It has been known for more than 40 years that individual neurons can produce more than one neurotransmitter and that neuropeptides often are colocalized with small molecule neurotransmitters (SMNs). Over the years much progress has been made in understanding the functional consequences of cotransmission in the nervous system of mammals. There are also some excellent invertebrate models that have revealed roles of coexpressed neuropeptides and SMNs in increasing complexity, flexibility, and dynamics in neuronal signaling. However, for the fly Drosophila there are surprisingly few functional studies on cotransmission, although there is ample evidence for colocalization of neuroactive compounds in neurons of the CNS, based both on traditional techniques and novel single cell transcriptome analysis. With the hope to trigger interest in initiating cotransmission studies, this review summarizes what is known about Drosophila neurons and neuronal circuits where different neuropeptides and SMNs are colocalized. Coexistence of neuroactive substances has been recorded in different neuron types such as neuroendocrine cells, interneurons, sensory cells and motor neurons. Some of the circuits highlighted here are well established in the analysis of learning and memory, circadian clock networks regulating rhythmic activity and sleep, as well as neurons and neuroendocrine cells regulating olfaction, nociception, feeding, metabolic homeostasis, diuretic functions, reproduction, and developmental processes. One emerging trait is the broad role of short neuropeptide F in cotransmission and presynaptic facilitation in a number of different neuronal circuits. This review also discusses the functional relevance of coexisting peptides in the intestine. Based on recent single cell transcriptomics data, it is likely that the neuronal systems discussed in this review are just a fraction of the total set of circuits where cotransmission occurs in Drosophila. Thus, a systematic search for colocalized neuroactive compounds in further neurons in anatomically defined circuits is of interest for the near future.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| |
Collapse
|
47
|
Guevara A, Gates H, Urbina B, French R. Developmental Ethanol Exposure Causes Reduced Feeding and Reveals a Critical Role for Neuropeptide F in Survival. Front Physiol 2018; 9:237. [PMID: 29623043 PMCID: PMC5875382 DOI: 10.3389/fphys.2018.00237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 03/02/2018] [Indexed: 12/25/2022] Open
Abstract
Food intake is necessary for survival, and natural reward circuitry has evolved to help ensure that animals ingest sufficient food to maintain development, growth, and survival. Drugs of abuse, including alcohol, co-opt the natural reward circuitry in the brain, and this is a major factor in the reinforcement of drug behaviors leading to addiction. At the junction of these two aspects of reward are alterations in feeding behavior due to alcohol consumption. In particular, developmental alcohol exposure (DAE) results in a collection of physical and neurobehavioral disorders collectively referred to as Fetal Alcohol Spectrum Disorder (FASD). The deleterious effects of DAE include intellectual disabilities and other neurobehavioral changes, including altered feeding behaviors. Here we use Drosophila melanogaster as a genetic model organism to study the effects of DAE on feeding behavior and the expression and function of Neuropeptide F. We show that addition of a defined concentration of ethanol to food leads to reduced feeding at all stages of development. Further, genetic conditions that reduce or eliminate NPF signaling combine with ethanol exposure to further reduce feeding, and the distribution of NPF is altered in the brains of ethanol-supplemented larvae. Most strikingly, we find that the vast majority of flies with a null mutation in the NPF receptor die early in larval development when reared in ethanol, and provide evidence that this lethality is due to voluntary starvation. Collectively, we find a critical role for NPF signaling in protecting against altered feeding behavior induced by developmental ethanol exposure.
Collapse
Affiliation(s)
- Amanda Guevara
- Biological Sciences, San Jose State University, San Jose, CA, United States
| | - Hillary Gates
- Biological Sciences, San Jose State University, San Jose, CA, United States
| | - Brianna Urbina
- Biological Sciences, San Jose State University, San Jose, CA, United States
| | - Rachael French
- Biological Sciences, San Jose State University, San Jose, CA, United States
| |
Collapse
|
48
|
Li X, Qu MJ, Zhang Y, Li JW, Liu TX. Expression of Neuropeptide F Gene and Its Regulation of Feeding Behavior in the Pea Aphid, Acyrthosiphon pisum. Front Physiol 2018; 9:87. [PMID: 29487538 PMCID: PMC5816806 DOI: 10.3389/fphys.2018.00087] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/25/2018] [Indexed: 01/20/2023] Open
Abstract
Neuropeptide F (NPF) signaling systems are widespread and highly evolutionarily conserved from vertebrates to invertebrates. In fact, NPF has been identified in many insect species and plays regulatory roles in diverse physiological processes, such as feeding, learning, reproduction and stress responses. NPF operates by interacting with the NPF receptor (NPFR). Here, we characterized and determined the presumed role of NPF signaling in the wingless parthenogenetic pea aphid, Acyrthosiphon pisum. Quantitative real-time reverse transcription-PCR (qRT-PCR) revealed that the expression levels of both NPF and NPFR transcripts varied across developmental stages, which implies that the NPF signaling system might participate in the developmental regulation of aphid physiological processes or behaviors. The NPF transcript was mainly detected in the head but not in the gut, whereas the NPFR transcript was mainly detected in both the gut and head. In addition, the NPF transcript levels were markedly up-regulated in starved aphids compared with satiated aphids, and the transcript levels recovered after re-feeding. In contrast, the NPFR transcript levels remained stable in starved and re-fed aphids. Furthermore, RNAi knockdown by the injection of NPF dsRNA into wingless adult aphids significantly reduced their food intake. Further analysis of the modification of aphid feeding behavior on broad bean plants using electrical penetration graphs (EPGs) revealed that both the probing time and the total duration of phloem activity decreased significantly in the NPF treatment group. These results indicated a lower appetite for food after NPF knockdown, which could explain the reduction in aphid food intake. NPF silencing was also shown to reduce reproduction but not survival in aphids. Overall, the results of these experiments suggest that NPF plays an important role in regulation of feeding in A. pisum.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Crop Stress Biology for Arid Areas, and Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, Northwest A&F University, Yangling, China.,Shandong Peanut Research Institute, Qingdao, Shandong, China
| | - Ming-Jing Qu
- Shandong Peanut Research Institute, Qingdao, Shandong, China
| | - Yi Zhang
- State Key Laboratory of Crop Stress Biology for Arid Areas, and Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Jian-Wen Li
- College of Life Science, Yangtze University, Jingzhou, China
| | - Tong-Xian Liu
- State Key Laboratory of Crop Stress Biology for Arid Areas, and Key Laboratory of Integrated Pest Management on the Loess Plateau of Ministry of Agriculture, Northwest A&F University, Yangling, China
| |
Collapse
|
49
|
Gui SH, Jiang HB, Smagghe G, Wang JJ. The neuropeptides and protein hormones of the agricultural pest fruit fly Bactrocera dorsalis: What do we learn from the genome sequencing and tissue-specific transcriptomes? Peptides 2017; 98:29-34. [PMID: 29061318 DOI: 10.1016/j.peptides.2017.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 01/02/2023]
Abstract
Neuropeptides and protein hormones are very important signaling molecules, and are involved in the regulation and coordination of various physiological processes in invertebrates and vertebrates. Using a bioinformatics approach, we screened the recently sequenced genome and six tissue-specific transcriptome databases (central nervous system, fat body, ovary, testes, male accessory glands, antennae) of the oriental fruit fly (Bactrocera dorsalis) that is economically one of the most important pest insects of tropical and subtropical fruit. Thirty-nine candidate genes were found to encode neuropeptides or protein hormones. These include most of the known insect neuropeptides and protein hormones, with the exception of adipokinetic hormone-corazonin-related peptide, allatropin, diuretic hormone 34, diuretic hormone 45, IMFamide, inotocin, and sex peptide. Our results showed the neuropeptides and protein hormones of Diptera insects appear to have a reduced repertoire compared to some other insects. Moreover, there are also differences between B. dorsalis and the super-model of Drosophila melanogaster. Interesting features of the oriental fruit fly are the absence of genes coding for sex peptide and the presence of neuroparsin and two genes coding neuropeptide F. The majority of the identified neuropeptides and protein hormones is present in the central nervous system, with only a limited number of these in the other tissues. Moreover, we predicted their physiological functions via comparing with data of FlyBase and FlyAtlas. Taken together, owing to the large number of identified peptides, this study can be used as a reference about structure, tissue distribution and physiological functions for comparative studies in other model and important pest insects.
Collapse
Affiliation(s)
- Shun-Hua Gui
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400715, China; Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China; Department of Crop Protection, Ghent University, Ghent, Belgium
| | - Hong-Bo Jiang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400715, China; Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China
| | - Guy Smagghe
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400715, China; Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China; Department of Crop Protection, Ghent University, Ghent, Belgium.
| | - Jin-Jun Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400715, China; Academy of Agricultural Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
50
|
Dissection of the Drosophila neuropeptide F circuit using a high-throughput two-choice assay. Proc Natl Acad Sci U S A 2017; 114:E8091-E8099. [PMID: 28874527 DOI: 10.1073/pnas.1710552114] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In their classic experiments, Olds and Milner showed that rats learn to lever press to receive an electric stimulus in specific brain regions. This led to the identification of mammalian reward centers. Our interest in defining the neuronal substrates of reward perception in the fruit fly Drosophila melanogaster prompted us to develop a simpler experimental approach wherein flies could implement behavior that induces self-stimulation of specific neurons in their brains. The high-throughput assay employs optogenetic activation of neurons when the fly occupies a specific area of a behavioral chamber, and the flies' preferential occupation of this area reflects their choosing to experience optogenetic stimulation. Flies in which neuropeptide F (NPF) neurons are activated display preference for the illuminated side of the chamber. We show that optogenetic activation of NPF neuron is rewarding in olfactory conditioning experiments and that the preference for NPF neuron activation is dependent on NPF signaling. Finally, we identify a small subset of NPF-expressing neurons located in the dorsomedial posterior brain that are sufficient to elicit preference in our assay. This assay provides the means for carrying out unbiased screens to map reward neurons in flies.
Collapse
|