1
|
Xia X, Li Y. A high-performance GRAB sensor reveals differences in the dynamics and molecular regulation between neuropeptide and neurotransmitter release. Nat Commun 2025; 16:819. [PMID: 39827209 PMCID: PMC11743212 DOI: 10.1038/s41467-025-56129-w] [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: 07/10/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
The co-existence and co-transmission of neuropeptides and small molecule neurotransmitters within individual neuron represent a fundamental characteristic observed across various species. However, the differences regarding their in vivo spatiotemporal dynamics and underlying molecular regulation remain poorly understood. Here, we develop a GPCR-activation-based (GRAB) sensor for detecting short neuropeptide F (sNPF) with high sensitivity and spatiotemporal resolution. Furthermore, we investigate the in vivo dynamics and molecular regulation differences between sNPF and acetylcholine (ACh) from the same neurons. Interestingly, our findings reveal distinct spatiotemporal dynamics in the release of sNPF and ACh. Notably, our results indicate that distinct synaptotagmins (Syt) are involved in these two processes, as Syt7 and Sytα for sNPF release, while Syt1 for ACh release. Thus, this high-performance GRAB sensor provides a robust tool for studying neuropeptide release and shedding insights into the unique release dynamics and molecular regulation that distinguish neuropeptides from small molecule neurotransmitters.
Collapse
Affiliation(s)
- Xiju Xia
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
2
|
Yoshinari Y, Nishimura T, Yoshii T, Kondo S, Tanimoto H, Kobayashi T, Matsuyama M, Niwa R. A high-protein diet-responsive gut hormone regulates behavioral and metabolic optimization in Drosophila melanogaster. Nat Commun 2024; 15:10819. [PMID: 39737959 PMCID: PMC11685984 DOI: 10.1038/s41467-024-55050-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Protein is essential for all living organisms; however, excessive protein intake can have adverse effects, such as hyperammonemia. Although mechanisms responding to protein deficiency are well-studied, there is a significant gap in our understanding of how organisms adaptively suppress excessive protein intake. In the present study, utilizing the fruit fly, Drosophila melanogaster, we discover that the peptide hormone CCHamide1 (CCHa1), secreted by enteroendocrine cells in response to a high-protein diet (HPD), is vital for suppressing overconsumption of protein. Gut-derived CCHa1 is received by a small subset of enteric neurons that produce short neuropeptide F, thereby modulating protein-specific satiety. Importantly, impairment of the CCHa1-mediated gut-enteric neuronal axis results in ammonia accumulation and a shortened lifespan under HPD conditions. Collectively, our findings unravel the crosstalk of gut hormone and neuronal pathways that orchestrate physiological responses to prevent and adapt to dietary protein overload.
Collapse
Affiliation(s)
- Yuto Yoshinari
- Metabolic Regulation and Genetics, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Takashi Nishimura
- Metabolic Regulation and Genetics, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan.
| | - Taishi Yoshii
- Graduate School of Environmental, Life, Natural Science and Technology, Okayama University, Okayama, Japan
| | - Shu Kondo
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Tokyo, Japan
- Invertebrate Genetics Laboratory, National Institute of Genetics, Mishima, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Tomoe Kobayashi
- Division of Molecular Genetics, Shigei Medical Research Institute, Okayama, Japan
| | - Makoto Matsuyama
- Division of Molecular Genetics, Shigei Medical Research Institute, Okayama, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
3
|
Toprak U, İnak E, Nauen R. Lipid Metabolism as a Target Site in Pest Control. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39466572 DOI: 10.1007/5584_2024_822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Lipid metabolism is essential to insect life as insects use lipids for their development, reproduction, flight, diapause, and a wide range of other functions. The central organ for insect lipid metabolism is the fat body, which is analogous to mammalian adipose tissue and liver, albeit less structured. Various other systems including the midgut, brain, and neural organs also contribute functionally to insect lipid metabolism. Lipid metabolism is under the control of core lipogenic [e.g. acetyl-CoA-carboxylase (ACC), fatty acid synthase (FAS), perilipin 2 (LSD2)], and lipolytic (lipases, perilipin 1) enzymes that are primarily expressed in the fat body, as well as hormones [insulin-like peptides (ILP), adipokinetic hormone (AKH)], transcription factors (SREBPs, foxO, and CREB), secondary messengers (calcium) and post-translational modifications (phosphorylation). Essential roles of the fat body, together with the fact that proper coordination of lipid metabolism is critical for insects, render lipid metabolism an attractive target site in pest control. In the current chapter, we focus on pest control tactics that target insect lipid metabolism. Various classes of traditional chemical insecticides [e.g. organophosphates, pyrethroids, neonicotinoids, and chitin synthesis inhibitors (Sects. 2.1 and 2.2)] have been shown to interfere with lipid metabolism, albeit it is not their primary site of action. However, the discovery of "lipid biosynthesis inhibitors", tetronic and tetramic acid derivatives commonly known as ketoenols (Sect. 2.3), was a milestone in applied entomology as they directly target lipid biosynthesis, particularly in sucking pests. Spirodiclofen, spiromesifen, and spirotetramat targeting ACC act against various insect and mite pests, while spiropidion and spidoxamat have been introduced to the market only recently. Efforts have concentrated on the development of chemical alternatives, such as hormone agonists and antagonists (Sect. 2.4), dsRNA-based pesticides that depend on RNA interference, which have great potential in pest control (Sect. 2.5) and other eco-friendly alternatives (Sect. 2.6).
Collapse
Affiliation(s)
- Umut Toprak
- Faculty of Agriculture, Department of Plant Protection Ankara, Molecular Entomology Lab, Ankara University, Ankara, Turkey.
| | - Emre İnak
- Faculty of Agriculture, Department of Plant Protection Ankara, Molecular Entomology Lab, Ankara University, Ankara, Turkey
| | - Ralf Nauen
- Bayer AG, Crop Science Division, Monheim, Germany.
| |
Collapse
|
4
|
Ignatiou A, Pitsouli C. Host-diet-microbiota interplay in intestinal nutrition and health. FEBS Lett 2024; 598:2482-2517. [PMID: 38946050 DOI: 10.1002/1873-3468.14966] [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: 04/21/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
The intestine is populated by a complex and dynamic assortment of microbes, collectively called gut microbiota, that interact with the host and contribute to its metabolism and physiology. Diet is considered a key regulator of intestinal microbiota, as ingested nutrients interact with and shape the resident microbiota composition. Furthermore, recent studies underscore the interplay of dietary and microbiota-derived nutrients, which directly impinge on intestinal stem cells regulating their turnover to ensure a healthy gut barrier. Although advanced sequencing methodologies have allowed the characterization of the human gut microbiome, mechanistic studies assessing diet-microbiota-host interactions depend on the use of genetically tractable models, such as Drosophila melanogaster. In this review, we first discuss the similarities between the human and fly intestines and then we focus on the effects of diet and microbiota on nutrient-sensing signaling cascades controlling intestinal stem cell self-renewal and differentiation, as well as disease. Finally, we underline the use of the Drosophila model in assessing the role of microbiota in gut-related pathologies and in understanding the mechanisms that mediate different whole-body manifestations of gut dysfunction.
Collapse
Affiliation(s)
- Anastasia Ignatiou
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
5
|
Xia X, Li Y. A new GRAB sensor reveals differences in the dynamics and molecular regulation between neuropeptide and neurotransmitter release. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595424. [PMID: 38826473 PMCID: PMC11142204 DOI: 10.1101/2024.05.22.595424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The co-existence and co-transmission of neuropeptides and small molecule neurotransmitters in the same neuron is a fundamental aspect of almost all neurons across various species. However, the differences regarding their in vivo spatiotemporal dynamics and underlying molecular regulation remain poorly understood. Here, we developed a GPCR-activation-based (GRAB) sensor for detecting short neuropeptide F (sNPF) with high sensitivity and spatiotemporal resolution. Furthermore, we explore the differences of in vivo dynamics and molecular regulation between sNPF and acetylcholine (ACh) from the same neurons. Interestingly, the release of sNPF and ACh shows different spatiotemporal dynamics. Notably, we found that distinct synaptotagmins (Syt) are involved in these two processes, as Syt7 and Sytα for sNPF release, while Syt1 for ACh release. Thus, this new GRAB sensor provides a powerful tool for studying neuropeptide release and providing new insights into the distinct release dynamics and molecular regulation between neuropeptides and small molecule neurotransmitters.
Collapse
Affiliation(s)
- Xiju Xia
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University–Tsinghua University–National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies (AAIS), and Peking University–Tsinghua University–National Institute of Biological Sciences Joint Graduate Program (PTN), Peking University, Beijing, 100871, China
- Chinese Institute for Brain Research, Beijing 102206, China
| |
Collapse
|
6
|
Li MM, Yang Q, Chen LH, Li YY, Wu JX, Xu XL. Effect of short neuropeptide F signaling on larval feeding in Mythimna separata. INSECT SCIENCE 2024; 31:417-434. [PMID: 37464946 DOI: 10.1111/1744-7917.13246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/27/2023] [Accepted: 05/22/2023] [Indexed: 07/20/2023]
Abstract
Mythimna separata is a notorious phytophagous pest which poses serious threats to cereal crops owing to the gluttony of the larvae. Because short neuropeptide F (sNPF) and its receptor sNPFR are involved in a diversity of physiological functions, especially in functions related to feeding in insects, it is a molecular target for pest control. Herein, an sNPF and 2 sNPFRs were identified and cloned from M. separata. Bioinformatics analysis revealed that the sNPF and its receptors had a highly conserved RLRFamide C-terminus and 7 transmembrane domains, respectively. The sNPF and its receptor genes were distributed across larval periods and tissues, but 2 receptors had distinct expression patterns. The starvation-induced assay elucidated that sNPF and sNPFR expression levels were downregulated under food deprivation and recovered with subsequent re-feeding. RNA interference knockdown of sNPF, sNPFR1, and sNPFR2 by injection of double-stranded RNA into larvae not only suppressed food consumption and increased body size and weight, but also led to decrease of glycogen and total lipid contents, and increase of trehalose compared with double-stranded green fluorescent protein injection. Furthermore, molecular docking was performed on the interaction mode between sNPFR protein and its ligand sNPF based on the 3-dimensional models constructed by AlphaFold; the results indicated that both receptors were presumably activated by the mature peptide sNPF-2. These results revealed that sNPF signaling played a considerably vital role in the feeding regulation of M. separata and represents a potential control target for this pest.
Collapse
Affiliation(s)
- Mei-Mei Li
- State Key Laboratory of Crop Stress Biology in Arid Areas, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi Province, China
| | - Qi Yang
- State Key Laboratory of Crop Stress Biology in Arid Areas, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi Province, China
| | - Li-Hui Chen
- School of Agricultural Sciences, Jiangxi Agricultural University, Nanchang, China
| | - Yan-Ying Li
- State Key Laboratory of Crop Stress Biology in Arid Areas, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi Province, China
| | - Jun-Xiang Wu
- State Key Laboratory of Crop Stress Biology in Arid Areas, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi Province, China
| | - Xiang-Li Xu
- State Key Laboratory of Crop Stress Biology in Arid Areas, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi Province, China
| |
Collapse
|
7
|
Chiang MH, Lin YC, Chen SF, Lee PS, Fu TF, Wu T, Wu CL. Independent insulin signaling modulators govern hot avoidance under different feeding states. PLoS Biol 2023; 21:e3002332. [PMID: 37847673 PMCID: PMC10581474 DOI: 10.1371/journal.pbio.3002332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 09/11/2023] [Indexed: 10/19/2023] Open
Abstract
Thermosensation is critical for the survival of animals. However, mechanisms through which nutritional status modulates thermosensation remain unclear. Herein, we showed that hungry Drosophila exhibit a strong hot avoidance behavior (HAB) compared to food-sated flies. We identified that hot stimulus increases the activity of α'β' mushroom body neurons (MBns), with weak activity in the sated state and strong activity in the hungry state. Furthermore, we showed that α'β' MBn receives the same level of hot input from the mALT projection neurons via cholinergic transmission in sated and hungry states. Differences in α'β' MBn activity between food-sated and hungry flies following heat stimuli are regulated by distinct Drosophila insulin-like peptides (Dilps). Dilp2 is secreted by insulin-producing cells (IPCs) and regulates HAB during satiety, whereas Dilp6 is secreted by the fat body and regulates HAB during the hungry state. We observed that Dilp2 induces PI3K/AKT signaling, whereas Dilp6 induces Ras/ERK signaling in α'β' MBn to regulate HAB in different feeding conditions. Finally, we showed that the 2 α'β'-related MB output neurons (MBONs), MBON-α'3 and MBON-β'1, are necessary for the output of integrated hot avoidance information from α'β' MBn. Our results demonstrate the presence of dual insulin modulation pathways in α'β' MBn, which are important for suitable behavioral responses in Drosophila during thermoregulation under different feeding states.
Collapse
Affiliation(s)
- Meng-Hsuan Chiang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chun Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Sheng-Fu Chen
- NHRI Institute of Biomedical Engineering & Nanomedicine, Miaoli, Taiwan
| | - Peng-Shiuan Lee
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsai-Feng Fu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Tony Wu
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City, Taiwan
| | - Chia-Lin Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurology, New Taipei Municipal TuCheng Hospital, Chang Gung Memorial Hospital, New Taipei City, Taiwan
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
8
|
Gendron CM, Chakraborty TS, Duran C, Dono T, Pletcher SD. Ring neurons in the Drosophila central complex act as a rheostat for sensory modulation of aging. PLoS Biol 2023; 21:e3002149. [PMID: 37310911 PMCID: PMC10263353 DOI: 10.1371/journal.pbio.3002149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 05/03/2023] [Indexed: 06/15/2023] Open
Abstract
Sensory perception modulates aging, yet we know little about how. An understanding of the neuronal mechanisms through which animals orchestrate biological responses to relevant sensory inputs would provide insight into the control systems that may be important for modulating lifespan. Here, we provide new awareness into how the perception of dead conspecifics, or death perception, which elicits behavioral and physiological effects in many different species, affects lifespan in the fruit fly, Drosophila melanogaster. Previous work demonstrated that cohousing Drosophila with dead conspecifics decreases fat stores, reduces starvation resistance, and accelerates aging in a manner that requires both sight and the serotonin receptor 5-HT2A. In this manuscript, we demonstrate that a discrete, 5-HT2A-expressing neural population in the ellipsoid body (EB) of the Drosophila central complex, identified as R2/R4 neurons, acts as a rheostat and plays an important role in transducing sensory information about the presence of dead individuals to modulate lifespan. Expression of the insulin-responsive transcription factor foxo in R2/R4 neurons and insulin-like peptides dilp3 and dilp5, but not dilp2, are required, with the latter likely altered in median neurosecretory cells (MNCs) after R2/R4 neuronal activation. These data generate new insights into the neural underpinnings of how perceptive events may impact aging and physiology across taxa.
Collapse
Affiliation(s)
- Christi M. Gendron
- Department of Molecular and Integrative Physiology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Tuhin S. Chakraborty
- Department of Molecular and Integrative Physiology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Cathryn Duran
- Department of Molecular and Integrative Physiology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Thomas Dono
- Department of Molecular and Integrative Physiology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Scott D. Pletcher
- Department of Molecular and Integrative Physiology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
9
|
Medla M, Daubnerová I, Koči J, Roller L, Slovák M, Žitňan D. Identification and expression of short neuropeptide F and its receptors in the tick Ixodes ricinus. JOURNAL OF INSECT PHYSIOLOGY 2023; 147:104524. [PMID: 37201579 DOI: 10.1016/j.jinsphys.2023.104524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/19/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023]
Abstract
In Europe, the tick Ixodes ricinus is the most important vector of numerous pathogens that are transmitted during blood feeding on their vertebrate hosts. To elucidate mechanisms controlling blood intake and associated transmission of pathogens we identified and described expression of short neuropeptide F (sNPF) and its receptors which are known to regulate feeding in insects. Using in situ hybridization (ISH) and immunohistochemistry (IHC) we stained numerous neurons producing sNPF in the central nervous system (CNS; synganglion), while a few peripheral neurons were detected anteriorly to the synganglion, and on the surface of the hindgut and leg muscles. Apparent sNPF expression was also found in enteroendocrine cells individually scattered in anterior lobes of the midgut. In silico analyses and BLAST search for sNPF receptors revealed two putative G protein-coupled receptors (sNPFR1 and sNPFR2) in the I. ricinus genome. Aequorin-based functional assay in CHO cells showed that both receptors were specific and sensitive to sNPF in nanomolar concentrations. Increased expression levels of these receptors in the gut during blood intake suggest that sNPF signaling may be involved in regulation of feeding and digestion processes of I. ricinus.
Collapse
Affiliation(s)
- Matej Medla
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia; Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia
| | - Ivana Daubnerová
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Juraj Koči
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Virology, Biomedical Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ladislav Roller
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Molecular Physiology and Genetics, Centre of Biosciences SAS, Bratislava, Slovakia
| | - Mirko Slovák
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Dušan Žitňan
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia.
| |
Collapse
|
10
|
Liessem S, Held M, Bisen RS, Haberkern H, Lacin H, Bockemühl T, Ache JM. Behavioral state-dependent modulation of insulin-producing cells in Drosophila. Curr Biol 2023; 33:449-463.e5. [PMID: 36580915 DOI: 10.1016/j.cub.2022.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/01/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022]
Abstract
Insulin signaling plays a pivotal role in metabolic control and aging, and insulin accordingly is a key factor in several human diseases. Despite this importance, the in vivo activity dynamics of insulin-producing cells (IPCs) are poorly understood. Here, we characterized the effects of locomotion on the activity of IPCs in Drosophila. Using in vivo electrophysiology and calcium imaging, we found that IPCs were strongly inhibited during walking and flight and that their activity rebounded and overshot after cessation of locomotion. Moreover, IPC activity changed rapidly during behavioral transitions, revealing that IPCs are modulated on fast timescales in behaving animals. Optogenetic activation of locomotor networks ex vivo, in the absence of actual locomotion or changes in hemolymph sugar levels, was sufficient to inhibit IPCs. This demonstrates that the behavioral state-dependent inhibition of IPCs is actively controlled by neuronal pathways and is independent of changes in glucose concentration. By contrast, the overshoot in IPC activity after locomotion was absent ex vivo and after starvation, indicating that it was not purely driven by feedforward signals but additionally required feedback derived from changes in hemolymph sugar concentration. We hypothesize that IPC inhibition during locomotion supports mobilization of fuel stores during metabolically demanding behaviors, while the rebound in IPC activity after locomotion contributes to replenishing muscle glycogen stores. In addition, the rapid dynamics of IPC modulation support a potential role of insulin in the state-dependent modulation of sensorimotor processing.
Collapse
Affiliation(s)
- Sander Liessem
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Martina Held
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Rituja S Bisen
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Hannah Haberkern
- HHMI Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Haluk Lacin
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St Louis, MO 63110, USA
| | - Till Bockemühl
- Department of Biology, Institute of Zoology, University of Cologne, Zülpicher Str. 47b, 50674 Cologne, Germany
| | - Jan M Ache
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| |
Collapse
|
11
|
Buhr A, Schiemann R, Meyer H. Neprilysin 4: an essential peptidase with multifaceted physiological relevance. Biol Chem 2023; 404:513-520. [PMID: 36653344 DOI: 10.1515/hsz-2022-0286] [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: 09/22/2022] [Accepted: 12/27/2022] [Indexed: 01/20/2023]
Abstract
Neprilysins are highly conserved ectoenzymes that hydrolyze and thus inactivate signaling peptides in the extracellular space. Herein, we focus on Neprilysin 4 from Drosophila melanogaster and evaluate the existing knowledge on the physiological relevance of the peptidase. Particular attention is paid to the role of the neprilysin in regulating feeding behavior and the expression of insulin-like peptides in the central nervous system. In addition, we assess the function of the peptidase in controlling the activity of the sarcoplasmic and endoplasmic reticulum Ca2+ ATPase in myocytes, as well as the underlying molecular mechanism in detail.
Collapse
Affiliation(s)
- Annika Buhr
- Department of Biology/Chemistry, Zoology and Developmental Biology Section, Osnabruck University, Barbarastrasse 11, D-49076 Osnabruck, Germany
| | - Ronja Schiemann
- Department of Biology/Chemistry, Zoology and Developmental Biology Section, Osnabruck University, Barbarastrasse 11, D-49076 Osnabruck, Germany
| | - Heiko Meyer
- Department of Biology/Chemistry, Zoology and Developmental Biology Section, Osnabruck University, Barbarastrasse 11, D-49076 Osnabruck, Germany.,Center for Cellular Nanoanalytics (CellNanOs), Barbarastrasse 11, D-49076 Osnabruck, Germany
| |
Collapse
|
12
|
Yoon S, Shin M, Shim J. Inter-organ regulation by the brain in Drosophila development and physiology. J Neurogenet 2022:1-13. [DOI: 10.1080/01677063.2022.2137162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Sunggyu Yoon
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Mingyu Shin
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
| | - Jiwon Shim
- Department of Life Sciences, College of Natural Science, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Science, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
13
|
Suh GSB, Yu K, Kim YJ, Oh Y, Park JJ. History of Drosophila neurogenetic research in South Korea. J Neurogenet 2022:1-7. [PMID: 36165786 DOI: 10.1080/01677063.2022.2115040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Neurogenetic research using the Drosophila model has immensely expanded around the world. Likewise, scientists in South Korea have leveraged the advantages of Drosophila genetic tools to understand various neurobiological processes. In this special issue, we will overview the history of Drosophila neurogenetic research in South Korea that led to significant discoveries and notably implications. We will describe how Drosophila system was first introduced to elevate neural developmental studies in 1990s. Establishing Drosophila-related resources has been a key venture, which led to the generation of over 100,000 mutant lines and the launch of the K-Gut initiative with Korea Drosophila Research Center (KDRC). These resources have supported the pioneer studies in modeling human disease and understanding genes and neural circuits that regulate animal behavior and physiology.
Collapse
Affiliation(s)
- Greg S B Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kweon Yu
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Metabolism and Neurophysiology Research Group, Daejeon, Republic of Korea
| | - Young-Joon Kim
- Department of Biological Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yangkyun Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Joong-Jean Park
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
14
|
Yoon S, Kim MA, Lee JS, Sohn YC. Functional analysis of LFRFamide signaling in Pacific abalone, Haliotis discus hannai. PLoS One 2022; 17:e0267039. [PMID: 35511902 PMCID: PMC9071130 DOI: 10.1371/journal.pone.0267039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/31/2022] [Indexed: 12/29/2022] Open
Abstract
The invertebrate LFRFamide (LFRFa) and short neuropeptide F (sNPF), consisting of 6 to 10 amino acids, are orthologs for bilaterian NPF/Y, which consist of 36 to 40 amino acids. Recently, a molluscan G protein-coupled receptor (GPCR) for NPF was characterized in Pacific abalone (Haliotis discus hannai). To address the functional evolutionary route of the invertebrate LFRFa and NPF signaling system, in this study, we identified cDNAs encoding LFRFa precursors and the sNPF receptor (Hdh-sNPFR) in Pacific abalone. Four LFRFa mature peptides with 6 or 7 amino acids were predicted: GSLFRFa, GGLFRFa, GTLFRFa, and GSTLFRFa. Hdh-sNPFR was identified as a classical rhodopsin-like GPCR and classified into a molluscan sNPFR group. In HEK293 cells, Hdh-sNPFR was mainly localized in the cell membranes and internalized in the cytoplasm following treatment with LFRFa peptides. Reporter assays demonstrated that LFRFa peptides inhibit forskolin-stimulated cAMP accumulation in Hdh-sNPFR-expressing HEK293 cells. LFRFa precursor and Hdh-sNPFR transcripts were more strongly expressed in the cerebral and pleural-pedal ganglia of Pacific abalone than in the peripheral tissues such as the ovary, gills, intestine, and hepatopancreas. The levels of LFRFa transcripts in the ovary, intestine, and hepatopancreas were significantly higher in mature female abalone than in immature females. Injection of LFRFa induced the egg release and spawning behavior of mature abalone, but suppressed food intake. These results suggest that LFRFa peptides are endogenous ligands for Hdh-sNPFR involved in food intake and reproduction through a Gαi-protein dependent signaling pathway.
Collapse
Affiliation(s)
- Sungwoo Yoon
- Department of Marine Bioscience, Gangneung-Wonju National University, Gangneung, Gangwon-do, Republic of Korea
| | - Mi Ae Kim
- Department of Marine Bioscience, Gangneung-Wonju National University, Gangneung, Gangwon-do, Republic of Korea
- East Coast Life Sciences Institute, Gangneung-Wonju National University, Gangneung, Gangwon, Republic of Korea
| | - Jung Sick Lee
- Department of Aqualife Medicine, Chonnam National University, Gwangju, Jeonnam, Republic of Korea
| | - Young Chang Sohn
- Department of Marine Bioscience, Gangneung-Wonju National University, Gangneung, Gangwon-do, Republic of Korea
- * E-mail:
| |
Collapse
|
15
|
Short Neuropeptide F and Its Receptor Regulate Feeding Behavior in Pea Aphid (Acyrthosiphon pisum). INSECTS 2022; 13:insects13030282. [PMID: 35323580 PMCID: PMC8950018 DOI: 10.3390/insects13030282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 12/04/2022]
Abstract
Simple Summary We know that neuropeptides and G protein-coupled receptors regulate the physiology and behavior of animals and that the pea aphid (Acyrthosiphon pisum) is a serious agricultural pest and model insect. In this study, we investigated the short neuropeptide F and its receptor in pea aphid. Feeding analysis showed that the probing time and total phloem duration significantly decreased in response to sNPF and predicted sNPFR gene silencing in RNAi assays. The silencing of sNPF significantly reduced the aphid’s reproduction but not survival. Our findings will help in the design of control strategies by using the molecular biological approach. Abstract Insect short neuropeptide F (sNPF), an ortholog of prolactin-releasing peptide of invertebrates, regulates diverse biological processes, including feeding, olfaction, locomotion, and sleep homeostasis in insects. However, its function is still unclear in an important model insect and agricultural pest, the pea aphid (Acyrthosiphon pisum). Here, we investigated short neuropeptide F (ApsNPF) and its receptor (ApsNPFR) in A. pisum. The sNPF gene contains three exons and two long introns. In addition, the genome contains a single sNPF receptor with seven transmembrane domains. Stage- and tissue-specific transcript profiling by qRT-PCR revealed that ApsNPF and ApsNPFR were mainly expressed in the central nervous system. The receptor was also detected in antennae, midgut, and integument. The highest expression levels were found in first instar nymphs compared to other developmental stages. Besides, the starvation-induced pattern indicated that the sNPF network depends on the nutritional state of the insect. An electrical penetration graph showed that probing time and phloem duration of A. pisum on broad bean plants decreased in response to dssNPF and dssNPFR in RNAi assays. sNPF silencing reduced the number of nymphs per female but not aphid survival. We believe that our results advance in-depth knowledge of the sNPF/sNPFR signaling cascade and its place in regulating feeding behavior in insects. In turn, it may contribute to the potential design of new strategies to control aphids, with a focus on the sNPF system.
Collapse
|
16
|
Nutrient Sensing via Gut in Drosophila melanogaster. Int J Mol Sci 2022; 23:ijms23052694. [PMID: 35269834 PMCID: PMC8910450 DOI: 10.3390/ijms23052694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Nutrient-sensing mechanisms in animals' sense available nutrients to generate a physiological regulatory response involving absorption, digestion, and regulation of food intake and to maintain glucose and energy homeostasis. During nutrient sensing via the gastrointestinal tract, nutrients interact with receptors on the enteroendocrine cells in the gut, which in return respond by secreting various hormones. Sensing of nutrients by the gut plays a critical role in transmitting food-related signals to the brain and other tissues informing the composition of ingested food to digestive processes. These signals modulate feeding behaviors, food intake, metabolism, insulin secretion, and energy balance. The increasing significance of fly genetics with the availability of a vast toolbox for studying physiological function, expression of chemosensory receptors, and monitoring the gene expression in specific cells of the intestine makes the fly gut the most useful tissue for studying the nutrient-sensing mechanisms. In this review, we emphasize on the role of Drosophila gut in nutrient-sensing to maintain metabolic homeostasis and gut-brain cross talk using endocrine and neuronal signaling pathways stimulated by internal state or the consumption of various dietary nutrients. Overall, this review will be useful in understanding the post-ingestive nutrient-sensing mechanisms having a physiological and pathological impact on health and diseases.
Collapse
|
17
|
Xu Z, Wei Y, Huang H, Guo S, Ye H. Immunomodulatory role of short neuropeptide F in the mud crab Scylla paramamosain. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 126:104260. [PMID: 34536467 DOI: 10.1016/j.dci.2021.104260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 06/13/2023]
Abstract
Short neuropeptide F (sNPF) is bioactive peptide secreted by neurons of invertebrates. It is one of the important pleiotropic neural molecules that is associated with a variety of physiological processes in invertebrates. However, little is known about the role of sNPF in the immune response. This study aimed to determine the distribution, localization, functional characteristics and signaling mechanisms of the sNPF gene and sNPF receptor (sNPF-R) gene in the mud crab Scylla paramamosain. Results of this study showed that Sp-sNPF and Sp-sNPF-R were widely expressed in neural tissue and other tissues including hemocytes. Further, in situ hybridization analysis revealed that Sp-sNPF and Sp-sNPF-R have specific localization in cerebral ganglion and hemocytes. It was also found that immune stimuli significantly induced Sp-sNPF expression in cerebral ganglion. The hemocyte-derived Sp-sNPF and Sp-sNPF-R were also efficiently activated upon immune stimulation. In vitro sNPF peptide administration enhanced phagocytic ability of hemocytes. However, this activity could be blocked through knockdown of sNPF-R-dsRNA or using adenylate cyclase inhibitors SQ 22536. The results of this study also demonstrated that the contents of signaling molecule adenylyl cyclase (AC), cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA) in hemocytes can be up-regulated after incubation with sNPF peptide. In addition, the results of in vivo experiments showed that sNPF increased concentration of nitric oxide (NO) and enhanced phagocytic potential in S. paramamosain. The sNPF also significantly induced the expression of immune-related molecules at the gene level in S. paramamosain. In conclusion, the findings of this study indicate that sNPF mediates hemocyte phagocytosis via sNPF-R receptor-coupled AC-cAMP-PKA pathway and influences the innate immune processes in S. paramamosain.
Collapse
Affiliation(s)
- Zhanning Xu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Yujie Wei
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Huiyang Huang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Songlin Guo
- College of Fisheries, Jimei University, Xiamen 361021, China
| | - Haihui Ye
- College of Fisheries, Jimei University, Xiamen 361021, China.
| |
Collapse
|
18
|
Li H, Huang X, Yang Y, Chen X, Yang Y, Wang J, Jiang H. The short neuropeptide F receptor regulates olfaction-mediated foraging behavior in the oriental fruit fly Bactrocera dorsalis (Hendel). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 140:103697. [PMID: 34843938 DOI: 10.1016/j.ibmb.2021.103697] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/14/2021] [Accepted: 11/14/2021] [Indexed: 06/13/2023]
Abstract
The short neuropeptide F (sNPF) signaling system, consisting of sNPF and its receptor (sNPFR), influences many physiological processes in insects, including feeding, growth and olfactory memory. We previously showed that sNPF regulates olfactory sensitivity in the oriental fruit fly Bactrocera dorsalis (Hendel) during starvation. However, the functional analysis of sNPFR is constrained by the failure of RNA interference in this species. Here, we generated a null sNPFR mutant using the CRISPR/Cas9 system to investigate the physiological roles of this receptor in more detail. G0 adults were produced at a frequency of 60.8%, and sNPFR-/- mutants were obtained after several generations of backcrossing followed by self-crossing among heterozygous flies. We found that the mutants were significantly less successful at foraging for certain foods and showed increased foraging latency. Electroantennogram (EAG) assays indicated that the mutants had significantly lower electrophysiological responses to three tested odorants. Furthermore, qPCR data revealed the inhibition of several olfactory receptor genes, including Orco. Immunohistochemistry showed that BdsNPFR was localized in cells under the sensillum on the antennae. Based on their shape and size, the BdsNPFR+ cells differ from odorant receptor neurons (ORNs), which were labeled using a Drosophila melanogaster Orco antibody. Our data suggest that sNPFR regulates olfaction-mediated foraging behavior by mediating interactions between BdsNPFR+ cells and selected ORNs.
Collapse
Affiliation(s)
- Hongfei Li
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400716, China; Academy of Agricultural Sciences, Southwest University, Chongqing, 400716, China.
| | - Xingying Huang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400716, China; Academy of Agricultural Sciences, Southwest University, Chongqing, 400716, China.
| | - Yahui Yang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400716, China; Academy of Agricultural Sciences, Southwest University, Chongqing, 400716, China.
| | - Xiaofeng Chen
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400716, China; Academy of Agricultural Sciences, Southwest University, Chongqing, 400716, China.
| | - Yang Yang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400716, China; Academy of Agricultural Sciences, Southwest University, Chongqing, 400716, China.
| | - Jinjun Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400716, China; Academy of Agricultural Sciences, Southwest University, Chongqing, 400716, China.
| | - Hongbo Jiang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400716, China; Academy of Agricultural Sciences, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
19
|
Internal state effects on behavioral shifts in freely behaving praying mantises (Tenodera sinensis). PLoS Comput Biol 2021; 17:e1009618. [PMID: 34928939 PMCID: PMC8751982 DOI: 10.1371/journal.pcbi.1009618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/11/2022] [Accepted: 11/05/2021] [Indexed: 11/19/2022] Open
Abstract
How we interact with our environment largely depends on both the external cues presented by our surroundings and the internal state from within. Internal states are the ever-changing physiological conditions that communicate the immediate survival needs and motivate the animal to behaviorally fulfill them. Satiety level constitutes such a state, and therefore has a dynamic influence on the output behaviors of an animal. In predatory insects like the praying mantis, hunting tactics, grooming, and mating have been shown to change hierarchical organization of behaviors depending on satiety. Here, we analyze behavior sequences of freely hunting praying mantises (Tenodera sinensis) to explore potential differences in sequential patterning of behavior as a correlate of satiety. First, our data supports previous work that showed starved praying mantises were not just more often attentive to prey, but also more often attentive to further prey. This was indicated by the increased time fraction spent in attentive bouts such as prey monitoring, head turns (to track prey), translations (closing the distance to the prey), and more strike attempts. With increasing satiety, praying mantises showed reduced time in these behaviors and exhibited them primarily towards close-proximity prey. Furthermore, our data demonstrates that during states of starvation, the praying mantis exhibits a stereotyped pattern of behavior that is highly motivated by prey capture. As satiety increased, the sequenced behaviors became more variable, indicating a shift away from the necessity of prey capture to more fluid presentations of behavior assembly.
Collapse
|
20
|
Carr AL, Rinker DC, Dong Y, Dimopoulos G, Zwiebel LJ. Transcriptome profiles of Anopheles gambiae harboring natural low-level Plasmodium infection reveal adaptive advantages for the mosquito. Sci Rep 2021; 11:22578. [PMID: 34799605 PMCID: PMC8604914 DOI: 10.1038/s41598-021-01842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/03/2021] [Indexed: 11/09/2022] Open
Abstract
Anopheline mosquitoes are the sole vectors for the Plasmodium pathogens responsible for malaria, which is among the oldest and most devastating of human diseases. The continuing global impact of malaria reflects the evolutionary success of a complex vector-pathogen relationship that accordingly has been the long-term focus of both debate and study. An open question in the biology of malaria transmission is the impact of naturally occurring low-level Plasmodium infections of the vector on the mosquito's health and longevity as well as critical behaviors such as host-preference/seeking. To begin to answer this, we have completed a comparative RNAseq-based transcriptome profile study examining the effect of biologically salient, salivary gland transmission-stage Plasmodium infection on the molecular physiology of Anopheles gambiae s.s. head, sensory appendages, and salivary glands. When compared with their uninfected counterparts, Plasmodium infected mosquitoes exhibit increased transcript abundance of genes associated with olfactory acuity as well as a range of synergistic processes that align with increased fitness based on both anti-aging and reproductive advantages. Taken together, these data argue against the long-held paradigm that malaria infection is pathogenic for anophelines and, instead suggests there are biological and evolutionary advantages for the mosquito that drive the preservation of its high vectorial capacity.
Collapse
Affiliation(s)
- Ann L Carr
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - David C Rinker
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Laurence J Zwiebel
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
21
|
Biglou SG, Bendena WG, Chin-Sang I. An overview of the insulin signaling pathway in model organisms Drosophila melanogaster and Caenorhabditis elegans. Peptides 2021; 145:170640. [PMID: 34450203 DOI: 10.1016/j.peptides.2021.170640] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/01/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022]
Abstract
The insulin/insulin-like growth factor signaling pathway is an evolutionary conserved pathway across metazoans and is required for development, metabolism and behavior. This pathway is associated with various human metabolic disorders and cancers. Thus, model organisms including Drosophila melanogaster and Caenorhabditis elegans provide excellent opportunities to examine the structure and function of this pathway and its influence on cellular metabolism and proliferation. In this review, we will provide an overview of human insulin and the human insulin signaling pathway and explore the recent discoveries in model organisms Drosophila melanogaster and Caenorhabditis elegans. Our review will provide information regarding the various insulin-like peptides in model organisms as well as the conserved functions of insulin signaling pathways. Further investigation of the insulin signaling pathway in model organisms could provide a promising opportunity to develop novel therapies for various metabolic disorders and insulin-mediated cancers.
Collapse
Affiliation(s)
- Sanaz G Biglou
- Department of Biology, Queen's University Kingston, ON, K7L3N6, Canada
| | - William G Bendena
- Department of Biology, Queen's University Kingston, ON, K7L3N6, Canada; Centre for Neuroscience, Queen's University, Kingston, ON, K7L3N6, Canada.
| | - Ian Chin-Sang
- Department of Biology, Queen's University Kingston, ON, K7L3N6, Canada
| |
Collapse
|
22
|
Choi M, Kim AK, Ham Y, Lee JY, Kim D, Yang A, Jo MJ, Yoon E, Heo JN, Han SB, Ki MH, Lee KS, Cho S. Aristolactam BIII, a naturally derived DYRK1A inhibitor, rescues Down syndrome-related phenotypes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 92:153695. [PMID: 34500300 DOI: 10.1016/j.phymed.2021.153695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is a significant pathogenic factor in Down syndrome (DS), wherein DYRK1A is overexpressed by 1.5-fold because of trisomy of human chromosome 21. Thus, DYRK1A inhibition is considered a therapeutic strategy to modify the disease. PURPOSE This study aims to identify a novel DYRK1A inhibitor and validate its therapeutic potential in DS-related pathological conditions. STUDY DESIGN In order to identify a novel DYRK1A inhibitor, we carried out two-step screening: a structure-based virtual screening of > 300,000 chemical library (first step) and cell-based nuclear factor of activated T-cells (NFAT)-response element (RE) promoter assay (second step). Primary hits were evaluated for their DYRK1A inhibitory activity using in vitro kinase assay and Tau phosphorylation in mammalian cells. Confirmed hit was further evaluated in pathological conditions including DYRK1A-overexpressing fibroblasts, flies, and mice. RESULTS We identified aristolactam BIII, a natural product derived from herbal plants, as a novel DYRK1A inhibitor. It potently inhibited the kinase activity of DYRK1A in vitro (IC50 = 9.67 nM) and effectively suppressed DYRK1A-mediated hyperphosphorylation of Tau in mammalian cells. Aristolactam BIII rescued the proliferative defects of DYRK1A transgenic (TG) mouse-derived fibroblasts and neurological and phenotypic defects of DS-like Drosophila models. Oral administration of aristolactam BIII acutely suppressed Tau hyperphosphorylation in the brain of DYRK1A TG mice. In the open field test, aristolactam BIII significantly ameliorated the exploratory behavioral deficit of DYRK1A TG mice. CONCLUSION Our work revealed that aristolactam BIII as a novel DYRK1A inhibitor rescues DS phenotypes in cells and in vivo and suggested its therapeutic potential for the treatment of DYRK1A-related diseases.
Collapse
Affiliation(s)
- Miri Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk 28116, Republic of Korea; College of Pharmacy, Chungbuk National University, 30-1 Yeonje-ri, Osong-eup, Heungduk-gu, Cheongju-si, Chungbuk 28644, Republic of Korea
| | - Ae-Kyeong Kim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Youngwook Ham
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk 28116, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Joo-Youn Lee
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Jang-dong, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Daeyong Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk 28116, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ansook Yang
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk 28116, Republic of Korea; College of Pharmacy, Chungbuk National University, 30-1 Yeonje-ri, Osong-eup, Heungduk-gu, Cheongju-si, Chungbuk 28644, Republic of Korea
| | - Min Ju Jo
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk 28116, Republic of Korea; College of Pharmacy, Chungbuk National University, 30-1 Yeonje-ri, Osong-eup, Heungduk-gu, Cheongju-si, Chungbuk 28644, Republic of Korea
| | - Eunyoung Yoon
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Jang-dong, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Jung-Nyoung Heo
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Jang-dong, Yuseong-gu, Daejeon 34114, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 30-1 Yeonje-ri, Osong-eup, Heungduk-gu, Cheongju-si, Chungbuk 28644, Republic of Korea
| | - Min-Hyo Ki
- Center Research Institute, Samjin Pharm. Co., Ltd., 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi 13488, Republic of Korea
| | - Kyu-Sun Lee
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sungchan Cho
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk 28116, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea.
| |
Collapse
|
23
|
Insects as a New Complex Model in Hormonal Basis of Obesity. Int J Mol Sci 2021; 22:ijms222011066. [PMID: 34681728 PMCID: PMC8540125 DOI: 10.3390/ijms222011066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 11/30/2022] Open
Abstract
Nowadays, one of the biggest problems in healthcare is an obesity epidemic. Consumption of cheap and low-quality energy-rich diets, low physical activity, and sedentary work favor an increase in the number of obesity cases within many populations/nations. This is a burden on society, public health, and the economy with many deleterious consequences. Thus, studies concerning this disorder are extremely needed, including searching for new, effective, and fitting models. Obesity may be related, among other factors, to disrupting adipocytes activity, disturbance of metabolic homeostasis, dysregulation of hormonal balance, cardiovascular problems, or disorders in nutrition which may lead to death. Because of the high complexity of obesity, it is not easy to find an ideal model for its studies which will be suitable for genetic and physiological analysis including specification of different compounds’ (hormones, neuropeptides) functions, as well as for signaling pathways analysis. In recent times, in search of new models for human diseases there has been more and more attention paid to insects, especially in neuro-endocrine regulation. It seems that this group of animals might also be a new model for human obesity. There are many arguments that insects are a good, multidirectional, and complex model for this disease. For example, insect models can have similar conservative signaling pathways (e.g., JAK-STAT signaling pathway), the presence of similar hormonal axis (e.g., brain–gut axis), or occurrence of structural and functional homologues between neuropeptides (e.g., neuropeptide F and human neuropeptide Y, insulin-like peptides, and human insulin) compared to humans. Here we give a hint to use insects as a model for obesity that can be used in multiple ways: as a source of genetic and peptidomic data about etiology and development correlated with obesity occurrence as well as a model for novel hormonal-based drug activity and their impact on mechanism of disease occurrence.
Collapse
|
24
|
Semaniuk U, Strilbytska O, Malinovska K, Storey KB, Vaiserman A, Lushchak V, Lushchak O. Factors that regulate expression patterns of insulin-like peptides and their association with physiological and metabolic traits in Drosophila. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 135:103609. [PMID: 34146686 DOI: 10.1016/j.ibmb.2021.103609] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/05/2021] [Accepted: 06/13/2021] [Indexed: 06/12/2023]
Abstract
Insulin-like peptides (ILPs) and components of the insulin signaling pathway are conserved across different animal phyla. Eight ILPs (called DILPs) and two receptors, dInR and Lgr3, have been described in Drosophila. DILPs regulate varied physiological traits including lifespan, reproduction, development, feeding behavior, stress resistance and metabolism. At the same time, different conditions such as nutrition, dietary supplements and environmental factors affect the expression of DILPs. This review focuses primarily on DILP2, DILP3, and DILP5 which are produced by insulin-producing cells in the brain of Drosophila. Although they are produced by the same cells and can potentially compensate for each other, DILP2, DILP3, and DILP5 expression may be differentially regulated at the mRNA level. Thus, we summarized available data on the conditions affecting the expression profiles of these DILPs in adult Drosophila. The accumulated data indicate that transcript levels of DILPs are determined by (a) nutritional conditions such as the protein-to-carbohydrate ratio, (b) carbohydrate type within the diet, (c) malnutrition or complete starvation; (d) environmental factors such as stress or temperature; (e) mutations of single peptides that induce changes in the expression of the other peptides; and (f) dietary supplements of drugs or natural substances. Furthermore, manipulation of specific genes in a cell- and tissue-specific manner affects mRNA levels for DILPs and, thereby, modulates various physiological traits and metabolism in Drosophila.
Collapse
Affiliation(s)
- Uliana Semaniuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Olha Strilbytska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | - Karina Malinovska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | | | | | - Volodymyr Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine; Research and Development University, Ivano-Frankivsk, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine; Research and Development University, Ivano-Frankivsk, Ukraine.
| |
Collapse
|
25
|
Fouks B, Brand P, Nguyen HN, Herman J, Camara F, Ence D, Hagen DE, Hoff KJ, Nachweide S, Romoth L, Walden KKO, Guigo R, Stanke M, Narzisi G, Yandell M, Robertson HM, Koeniger N, Chantawannakul P, Schatz MC, Worley KC, Robinson GE, Elsik CG, Rueppell O. The genomic basis of evolutionary differentiation among honey bees. Genome Res 2021; 31:1203-1215. [PMID: 33947700 PMCID: PMC8256857 DOI: 10.1101/gr.272310.120] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
In contrast to the western honey bee, Apis mellifera, other honey bee species have been largely neglected despite their importance and diversity. The genetic basis of the evolutionary diversification of honey bees remains largely unknown. Here, we provide a genome-wide comparison of three honey bee species, each representing one of the three subgenera of honey bees, namely the dwarf (Apis florea), giant (A. dorsata), and cavity-nesting (A. mellifera) honey bees with bumblebees as an outgroup. Our analyses resolve the phylogeny of honey bees with the dwarf honey bees diverging first. We find that evolution of increased eusocial complexity in Apis proceeds via increases in the complexity of gene regulation, which is in agreement with previous studies. However, this process seems to be related to pathways other than transcriptional control. Positive selection patterns across Apis reveal a trade-off between maintaining genome stability and generating genetic diversity, with a rapidly evolving piRNA pathway leading to genomes depleted of transposable elements, and a rapidly evolving DNA repair pathway associated with high recombination rates in all Apis species. Diversification within Apis is accompanied by positive selection in several genes whose putative functions present candidate mechanisms for lineage-specific adaptations, such as migration, immunity, and nesting behavior.
Collapse
Affiliation(s)
- Bertrand Fouks
- Department of Biology, University of North Carolina at Greensboro, Greensboro, North Carolina 27403, USA
- Institute for Evolution and Biodiversity, Molecular Evolution and Bioinformatics, Westfälische Wilhelms-Universität, 48149 Münster, Germany
| | - Philipp Brand
- Department of Evolution and Ecology, Center for Population Biology, University of California, Davis, Davis, California 95161, USA
- Laboratory of Neurophysiology and Behavior, The Rockefeller University, New York, New York 10065, USA
| | - Hung N Nguyen
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri 65211, USA
| | - Jacob Herman
- Department of Biology, University of North Carolina at Greensboro, Greensboro, North Carolina 27403, USA
| | - Francisco Camara
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08036 Barcelona, Spain
| | - Daniel Ence
- School of Forest Resources and Conservation, University of Florida, Gainesville, Florida 32611, USA
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
| | - Darren E Hagen
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, Oklahoma 74078, USA
| | - Katharina J Hoff
- University of Greifswald, Institute for Mathematics and Computer Science, Bioinformatics Group, 17489 Greifswald, Germany
- University of Greifswald, Center for Functional Genomics of Microbes, 17489 Greifswald, Germany
| | - Stefanie Nachweide
- University of Greifswald, Institute for Mathematics and Computer Science, Bioinformatics Group, 17489 Greifswald, Germany
| | - Lars Romoth
- University of Greifswald, Institute for Mathematics and Computer Science, Bioinformatics Group, 17489 Greifswald, Germany
| | - Kimberly K O Walden
- Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Roderic Guigo
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08036 Barcelona, Spain
- Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Mario Stanke
- University of Greifswald, Institute for Mathematics and Computer Science, Bioinformatics Group, 17489 Greifswald, Germany
- University of Greifswald, Center for Functional Genomics of Microbes, 17489 Greifswald, Germany
| | | | - Mark Yandell
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
- Utah Center for Genetic Discovery, University of Utah, Salt Lake City, Utah 84112, USA
| | - Hugh M Robertson
- Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Nikolaus Koeniger
- Department of Behavioral Physiology and Sociobiology (Zoology II), University of Würzburg, 97074 Würzburg, Germany
| | - Panuwan Chantawannakul
- Environmental Science Research Center (ESRC) and Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Michael C Schatz
- Departments of Computer Science and Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Kim C Worley
- Department of Molecular and Human Genetics, Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Gene E Robinson
- Department of Entomology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Christine G Elsik
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri 65211, USA
- Division of Animal Sciences, University of Missouri, Columbia, Missouri 65211, USA
- Division of Plant Sciences, University of Missouri, Columbia, Missouri 65211, USA
| | - Olav Rueppell
- Department of Biology, University of North Carolina at Greensboro, Greensboro, North Carolina 27403, USA
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| |
Collapse
|
26
|
Chowański S, Walkowiak-Nowicka K, Winkiel M, Marciniak P, Urbański A, Pacholska-Bogalska J. Insulin-Like Peptides and Cross-Talk With Other Factors in the Regulation of Insect Metabolism. Front Physiol 2021; 12:701203. [PMID: 34267679 PMCID: PMC8276055 DOI: 10.3389/fphys.2021.701203] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
The insulin-like peptide (ILP) and insulin-like growth factor (IGF) signalling pathways play a crucial role in the regulation of metabolism, growth and development, fecundity, stress resistance, and lifespan. ILPs are encoded by multigene families that are expressed in nervous and non-nervous organs, including the midgut, salivary glands, and fat body, in a tissue- and stage-specific manner. Thus, more multidirectional and more complex control of insect metabolism can occur. ILPs are not the only factors that regulate metabolism. ILPs interact in many cross-talk interactions of different factors, for example, hormones (peptide and nonpeptide), neurotransmitters and growth factors. These interactions are observed at different levels, and three interactions appear to be the most prominent/significant: (1) coinfluence of ILPs and other factors on the same target cells, (2) influence of ILPs on synthesis/secretion of other factors regulating metabolism, and (3) regulation of activity of cells producing/secreting ILPs by various factors. For example, brain insulin-producing cells co-express sulfakinins (SKs), which are cholecystokinin-like peptides, another key regulator of metabolism, and express receptors for tachykinin-related peptides, the next peptide hormones involved in the control of metabolism. It was also shown that ILPs in Drosophila melanogaster can directly and indirectly regulate AKH. This review presents an overview of the regulatory role of insulin-like peptides in insect metabolism and how these factors interact with other players involved in its regulation.
Collapse
Affiliation(s)
- Szymon Chowański
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Karolina Walkowiak-Nowicka
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Magdalena Winkiel
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Pawel Marciniak
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Arkadiusz Urbański
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland.,HiProMine S.A., Robakowo, Poland
| | - Joanna Pacholska-Bogalska
- Department of Animal Physiology and Development, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
27
|
Han B, Wei Q, Wu F, Hu H, Ma C, Meng L, Zhang X, Feng M, Fang Y, Rueppell O, Li J. Tachykinin signaling inhibits task-specific behavioral responsiveness in honeybee workers. eLife 2021; 10:64830. [PMID: 33760729 PMCID: PMC8016481 DOI: 10.7554/elife.64830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/23/2021] [Indexed: 12/11/2022] Open
Abstract
Behavioral specialization is key to the success of social insects and leads to division of labor among colony members. Response thresholds to task-specific stimuli are thought to proximally regulate behavioral specialization, but their neurobiological regulation is complex and not well understood. Here, we show that response thresholds to task-relevant stimuli correspond to the specialization of three behavioral phenotypes of honeybee workers in the well-studied and important Apis mellifera and Apis cerana. Quantitative neuropeptidome comparisons suggest two tachykinin-related peptides (TRP2 and TRP3) as candidates for the modification of these response thresholds. Based on our characterization of their receptor binding and downstream signaling, we confirm a functional role of tachykinin signaling in regulating specific responsiveness of honeybee workers: TRP2 injection and RNAi-mediated downregulation cause consistent, opposite effects on responsiveness to task-specific stimuli of each behaviorally specialized phenotype but not to stimuli that are unrelated to their tasks. Thus, our study demonstrates that TRP signaling regulates the degree of task-specific responsiveness of specialized honeybee workers and may control the context specificity of behavior in animals more generally.
Collapse
Affiliation(s)
- Bin Han
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China.,Department of Biology, University of North Carolina Greensboro, Greensboro, United States
| | - Qiaohong Wei
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China
| | - Fan Wu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China.,Biometrology and Inspection & Quarantine, College of Life Science, China Jiliang University, Hangzhou, China
| | - Han Hu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China
| | - Chuan Ma
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China
| | - Lifeng Meng
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China
| | - Xufeng Zhang
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China.,Institute of Horticultural Research, Shanxi Academy of Agricultural Sciences, Shanxi Agricultural University, Taiyuan, China
| | - Mao Feng
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China
| | - Yu Fang
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China
| | - Olav Rueppell
- Department of Biology, University of North Carolina Greensboro, Greensboro, United States.,Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Jianke Li
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Science, Beijing, China
| |
Collapse
|
28
|
Millington JW, Brownrigg GP, Basner-Collins PJ, Sun Z, Rideout EJ. Genetic manipulation of insulin/insulin-like growth factor signaling pathway activity has sex-biased effects on Drosophila body size. G3 (BETHESDA, MD.) 2021; 11:jkaa067. [PMID: 33793746 PMCID: PMC8063079 DOI: 10.1093/g3journal/jkaa067] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022]
Abstract
In Drosophila raised in nutrient-rich conditions, female body size is approximately 30% larger than male body size due to an increased rate of growth and differential weight loss during the larval period. While the mechanisms that control this sex difference in body size remain incompletely understood, recent studies suggest that the insulin/insulin-like growth factor signaling pathway (IIS) plays a role in the sex-specific regulation of processes that influence body size during development. In larvae, IIS activity differs between the sexes, and there is evidence of sex-specific regulation of IIS ligands. Yet, we lack knowledge of how changes to IIS activity impact body size in each sex, as the majority of studies on IIS and body size use single- or mixed-sex groups of larvae and/or adult flies. The goal of our current study was to clarify the body size requirement for IIS activity in each sex. To achieve this goal, we used established genetic approaches to enhance, or inhibit, IIS activity, and quantified pupal size in males and females. Overall, genotypes that inhibited IIS activity caused a female-biased decrease in body size, whereas genotypes that augmented IIS activity caused a male-specific increase in body size. These data extend our current understanding of body size regulation by showing that most changes to IIS pathway activity have sex-biased effects, and highlights the importance of analyzing body size data according to sex.
Collapse
Affiliation(s)
- Jason W Millington
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - George P Brownrigg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Paige J Basner-Collins
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ziwei Sun
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
29
|
Clements J, Buhler K, Winant M, Vulsteke V, Callaerts P. Glial and Neuronal Neuroglian, Semaphorin-1a and Plexin A Regulate Morphological and Functional Differentiation of Drosophila Insulin-Producing Cells. Front Endocrinol (Lausanne) 2021; 12:600251. [PMID: 34276554 PMCID: PMC8281472 DOI: 10.3389/fendo.2021.600251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 06/11/2021] [Indexed: 11/21/2022] Open
Abstract
The insulin-producing cells (IPCs), a group of 14 neurons in the Drosophila brain, regulate numerous processes, including energy homeostasis, lifespan, stress response, fecundity, and various behaviors, such as foraging and sleep. Despite their importance, little is known about the development and the factors that regulate morphological and functional differentiation of IPCs. In this study, we describe the use of a new transgenic reporter to characterize the role of the Drosophila L1-CAM homolog Neuroglian (Nrg), and the transmembrane Semaphorin-1a (Sema-1a) and its receptor Plexin A (PlexA) in the differentiation of the insulin-producing neurons. Loss of Nrg results in defasciculation and abnormal neurite branching, including ectopic neurites in the IPC neurons. Cell-type specific RNAi knockdown experiments reveal that Nrg, Sema-1a and PlexA are required in IPCs and glia to control normal morphological differentiation of IPCs albeit with a stronger contribution of Nrg and Sema-1a in glia and of PlexA in the IPCs. These observations provide new insights into the development of the IPC neurons and identify a novel role for Sema-1a in glia. In addition, we show that Nrg, Sema-1a and PlexA in glia and IPCs not only regulate morphological but also functional differentiation of the IPCs and that the functional deficits are likely independent of the morphological phenotypes. The requirements of nrg, Sema-1a, and PlexA in IPC development and the expression of their vertebrate counterparts in the hypothalamic-pituitary axis, suggest that these functions may be evolutionarily conserved in the establishment of vertebrate endocrine systems.
Collapse
|
30
|
Koyama T, Texada MJ, Halberg KA, Rewitz K. Metabolism and growth adaptation to environmental conditions in Drosophila. Cell Mol Life Sci 2020; 77:4523-4551. [PMID: 32448994 PMCID: PMC7599194 DOI: 10.1007/s00018-020-03547-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/19/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Organisms adapt to changing environments by adjusting their development, metabolism, and behavior to improve their chances of survival and reproduction. To achieve such flexibility, organisms must be able to sense and respond to changes in external environmental conditions and their internal state. Metabolic adaptation in response to altered nutrient availability is key to maintaining energy homeostasis and sustaining developmental growth. Furthermore, environmental variables exert major influences on growth and final adult body size in animals. This developmental plasticity depends on adaptive responses to internal state and external cues that are essential for developmental processes. Genetic studies have shown that the fruit fly Drosophila, similarly to mammals, regulates its metabolism, growth, and behavior in response to the environment through several key hormones including insulin, peptides with glucagon-like function, and steroid hormones. Here we review emerging evidence showing that various environmental cues and internal conditions are sensed in different organs that, via inter-organ communication, relay information to neuroendocrine centers that control insulin and steroid signaling. This review focuses on endocrine regulation of development, metabolism, and behavior in Drosophila, highlighting recent advances in the role of the neuroendocrine system as a signaling hub that integrates environmental inputs and drives adaptive responses.
Collapse
Affiliation(s)
- Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth A Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
31
|
Nunes C, Sucena É, Koyama T. Endocrine regulation of immunity in insects. FEBS J 2020; 288:3928-3947. [PMID: 33021015 DOI: 10.1111/febs.15581] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Organisms have constant contact with potentially harmful agents that can compromise their fitness. However, most of the times these agents fail to cause serious disease by virtue of the rapid and efficient immune responses elicited in the host that can range from behavioural adaptations to immune system triggering. The immune system of insects does not comprise the adaptive arm, making it less complex than that of vertebrates, but key aspects of the activation and regulation of innate immunity are conserved across different phyla. This is the case for the hormonal regulation of immunity as a part of the broad organismal responses to external conditions under different internal states. In insects, depending on the physiological circumstances, distinct hormones either enhance or suppress the immune response integrating individual (and often collective) responses physiologically and behaviourally. In this review, we provide an overview of our current knowledge on the endocrine regulation of immunity in insects, its mechanisms and implications on metabolic adaptation and behaviour. We highlight the importance of this multilayered regulation of immunity in survival and reproduction (fitness) and its dependence on the hormonal integration with other mechanisms and life-history traits.
Collapse
Affiliation(s)
| | - Élio Sucena
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Departamento de Biologia Animal, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Texada MJ, Koyama T, Rewitz K. Regulation of Body Size and Growth Control. Genetics 2020; 216:269-313. [PMID: 33023929 PMCID: PMC7536854 DOI: 10.1534/genetics.120.303095] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
The control of body and organ growth is essential for the development of adults with proper size and proportions, which is important for survival and reproduction. In animals, adult body size is determined by the rate and duration of juvenile growth, which are influenced by the environment. In nutrient-scarce environments in which more time is needed for growth, the juvenile growth period can be extended by delaying maturation, whereas juvenile development is rapidly completed in nutrient-rich conditions. This flexibility requires the integration of environmental cues with developmental signals that govern internal checkpoints to ensure that maturation does not begin until sufficient tissue growth has occurred to reach a proper adult size. The Target of Rapamycin (TOR) pathway is the primary cell-autonomous nutrient sensor, while circulating hormones such as steroids and insulin-like growth factors are the main systemic regulators of growth and maturation in animals. We discuss recent findings in Drosophila melanogaster showing that cell-autonomous environment and growth-sensing mechanisms, involving TOR and other growth-regulatory pathways, that converge on insulin and steroid relay centers are responsible for adjusting systemic growth, and development, in response to external and internal conditions. In addition to this, proper organ growth is also monitored and coordinated with whole-body growth and the timing of maturation through modulation of steroid signaling. This coordination involves interorgan communication mediated by Drosophila insulin-like peptide 8 in response to tissue growth status. Together, these multiple nutritional and developmental cues feed into neuroendocrine hubs controlling insulin and steroid signaling, serving as checkpoints at which developmental progression toward maturation can be delayed. This review focuses on these mechanisms by which external and internal conditions can modulate developmental growth and ensure proper adult body size, and highlights the conserved architecture of this system, which has made Drosophila a prime model for understanding the coordination of growth and maturation in animals.
Collapse
Affiliation(s)
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, 2100, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100, Denmark
| |
Collapse
|
33
|
Sheng L, Shields EJ, Gospocic J, Glastad KM, Ratchasanmuang P, Berger SL, Raj A, Little S, Bonasio R. Social reprogramming in ants induces longevity-associated glia remodeling. SCIENCE ADVANCES 2020; 6:eaba9869. [PMID: 32875108 PMCID: PMC7438095 DOI: 10.1126/sciadv.aba9869] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 07/09/2020] [Indexed: 05/16/2023]
Abstract
In social insects, workers and queens arise from the same genome but display profound differences in behavior and longevity. In Harpegnathos saltator ants, adult workers can transition to a queen-like state called gamergate, which results in reprogramming of social behavior and life-span extension. Using single-cell RNA sequencing, we compared the distribution of neuronal and glial populations before and after the social transition. We found that the conversion of workers into gamergates resulted in the expansion of neuroprotective ensheathing glia. Brain injury assays revealed that activation of the damage response gene Mmp1 was weaker in old workers, where the relative frequency of ensheathing glia also declined. On the other hand, long-lived gamergates retained a larger fraction of ensheathing glia and the ability to mount a strong Mmp1 response to brain injury into old age. We also observed molecular and cellular changes suggestive of age-associated decline in ensheathing glia in Drosophila.
Collapse
Affiliation(s)
- Lihong Sheng
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Emily J. Shields
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Janko Gospocic
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Karl M. Glastad
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Puttachai Ratchasanmuang
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shelley L. Berger
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arjun Raj
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Shawn Little
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Roberto Bonasio
- Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
34
|
Lee KS, Choi M, Kwon DW, Kim D, Choi JM, Kim AK, Ham Y, Han SB, Cho S, Cheon CK. A novel de novo heterozygous DYRK1A mutation causes complete loss of DYRK1A function and developmental delay. Sci Rep 2020; 10:9849. [PMID: 32555303 PMCID: PMC7299959 DOI: 10.1038/s41598-020-66750-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/14/2020] [Indexed: 01/01/2023] Open
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 1 A (DYRK1A) is essential for human development, and DYRK1A haploinsufficiency is associated with a recognizable developmental syndrome and variable clinical features. Here, we present a patient with DYRK1A haploinsufficiency syndrome, including facial dysmorphism, delayed motor development, cardiovascular system defects, and brain atrophy. Exome sequencing identified a novel de novo heterozygous mutation of the human DYRK1A gene (c.1185dup), which generated a translational termination codon and resulted in a C-terminally truncated protein (DYRK1A-E396ter). To study the molecular effect of this truncation, we generated mammalian cell and Drosophila models that recapitulated the DYRK1A protein truncation. Analysis of the structure and deformation energy of the mutant protein predicted a reduction in protein stability. Experimentally, the mutant protein was efficiently degraded by the ubiquitin-dependent proteasome pathway and was barely detectable in mammalian cells. More importantly, the mutant kinase was intrinsically inactive and had little negative impact on the wild-type protein. Similarly, the mutant protein had a minimal effect on Drosophila phenotypes, confirming its loss-of-function in vivo. Together, our results suggest that the novel heterozygous mutation of DYRK1A resulted in loss-of-function of the kinase activity of DYRK1A and may contribute to the developmental delay observed in the patient.
Collapse
Affiliation(s)
- Kyu-Sun Lee
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Gajeong-dong, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Miri Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk, 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, 30-1 Yeonje-ri, Osong-eup, Heungduk-gu, Cheongju-si, Chungbuk, 28644, Republic of Korea
| | - Dae-Woo Kwon
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Gajeong-dong, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Doyoun Kim
- Innovative Target Research Center, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Jang-dong, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Jong-Moon Choi
- Green Cross Genome, Green Cross Laboratories, 107 Ihyeon-ro 30 beon-gil, Giheung-gu, Yongin-si, Gyeonggi, 16924, Republic of Korea
| | - Ae-Kyeong Kim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Youngwook Ham
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk, 28116, Republic of Korea
- College of Pharmacy, Chungbuk National University, 30-1 Yeonje-ri, Osong-eup, Heungduk-gu, Cheongju-si, Chungbuk, 28644, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, 30-1 Yeonje-ri, Osong-eup, Heungduk-gu, Cheongju-si, Chungbuk, 28644, Republic of Korea
| | - Sungchan Cho
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungbuk, 28116, Republic of Korea.
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Gajeong-dong, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| | - Chong Kun Cheon
- Division of Medical Genetics and Metabolism, Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, 20 Geumo-ro, Mulgeum-eup, Yangsan-si, Gyeongnam, 50612, Republic of Korea.
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, 20 Geumo-ro, Mulgeum-eup, Yangsan-si, Gyeongnam, 50612, Republic of Korea.
| |
Collapse
|
35
|
Wilson KA, Beck JN, Nelson CS, Hilsabeck TA, Promislow D, Brem RB, Kapahi P. GWAS for Lifespan and Decline in Climbing Ability in Flies upon Dietary Restriction Reveal decima as a Mediator of Insulin-like Peptide Production. Curr Biol 2020; 30:2749-2760.e3. [PMID: 32502405 DOI: 10.1016/j.cub.2020.05.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 03/17/2020] [Accepted: 05/06/2020] [Indexed: 12/16/2022]
Abstract
Dietary restriction (DR) is the most robust means to extend lifespan and delay age-related diseases across species. An underlying assumption in the aging field is that DR enhances both lifespan and physical activity through similar mechanisms, but this has not been rigorously tested in different genetic backgrounds. Furthermore, nutrient response genes responsible for lifespan extension or age-related decline in functionality remain underexplored in natural populations. To address this, we measured nutrient-dependent changes in lifespan and age-related decline in climbing ability in the Drosophila Genetic Reference Panel fly strains. On average, DR extended lifespan and delayed decline in climbing ability, but there was a lack of correlation between these traits across individual strains, suggesting that distinct genetic factors modulate these traits independently and that genotype determines response to diet. Only 50% of strains showed positive response to DR for both lifespan and climbing ability, 14% showed a negative response for one trait but not both, and 35% showed no change in one or both traits. Through GWAS, we uncovered a number of genes previously not known to be diet responsive nor to influence lifespan or climbing ability. We validated decima as a gene that alters lifespan and daedalus as one that influences age-related decline in climbing ability. We found that decima influences insulin-like peptide transcription in the GABA receptor neurons downstream of short neuropeptide F precursor (sNPF) signaling. Modulating these genes produced independent effects on lifespan and physical activity decline, which suggests that these age-related traits can be regulated through distinct mechanisms.
Collapse
Affiliation(s)
- Kenneth A Wilson
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA; Davis School of Gerontology, University of Southern California, University Park, Los Angeles, CA 90007, USA
| | - Jennifer N Beck
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA; Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, Room A-632, San Francisco, CA 94143, USA
| | | | - Tyler A Hilsabeck
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA; Davis School of Gerontology, University of Southern California, University Park, Los Angeles, CA 90007, USA
| | - Daniel Promislow
- Department of Pathology, University of Washington, Seattle, WA 98195, USA; Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Rachel B Brem
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA; Davis School of Gerontology, University of Southern California, University Park, Los Angeles, CA 90007, USA; Department of Plant and Microbial Biology, University of California, Berkeley, 111 Koshland Hall, Berkeley, CA 94720, USA.
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, 8001 Redwood Blvd., Novato, CA 94945, USA; Davis School of Gerontology, University of Southern California, University Park, Los Angeles, CA 90007, USA; Department of Urology, University of California, San Francisco, 400 Parnassus Avenue, Room A-632, San Francisco, CA 94143, USA.
| |
Collapse
|
36
|
Toprak U, Hegedus D, Doğan C, Güney G. A journey into the world of insect lipid metabolism. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2020; 104:e21682. [PMID: 32335968 DOI: 10.1002/arch.21682] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 06/11/2023]
Abstract
Lipid metabolism is fundamental to life. In insects, it is critical, during reproduction, 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. Fat body contains various different cell types; however, adipocytes and oenocytes are the primary cells related to lipid metabolism. Lipid metabolism starts with the hydrolysis of dietary lipids, absorption of lipid monomers, followed by lipid transport from midgut to the fat body, lipogenesis or lipolysis in the fat body, and lipid transport from fat body to other sites demanding energy. Lipid metabolism is under the control of hormones, transcription factors, secondary messengers and posttranscriptional modifications. Primarily, lipogenesis is under the control of insulin-like peptides that activate lipogenic transcription factors, such as sterol regulatory element-binding proteins, whereas lipolysis is coordinated by the adipokinetic hormone that activates lipolytic transcription factors, such as forkhead box class O and cAMP-response element-binding protein. Calcium is the primary-secondary messenger affecting lipid metabolism and has different outcomes depending on the site of lipogenesis or lipolysis. Phosphorylation is central to lipid metabolism and multiple phosphorylases are involved in lipid accumulation or hydrolysis. Although most of the knowledge of insect lipid metabolism comes from the studies on the model Drosophila; other insects, in particular those with obligatory or facultative diapause, also have great potential to study lipid metabolism. The use of these models would significantly improve our knowledge of insect lipid metabolism.
Collapse
Affiliation(s)
- Umut Toprak
- Molecular Entomology Laboratory, Department of Plant Protection, Faculty of Agriculture, Ankara University, Ankara, Turkey
| | - Dwayne Hegedus
- Agriculture and Agri-Food Canada, Saskatoon Research Centre, Saskatoon, Saskatchewan, Canada
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cansu Doğan
- Molecular Entomology Laboratory, Department of Plant Protection, Faculty of Agriculture, Ankara University, Ankara, Turkey
| | - Gözde Güney
- Molecular Entomology Laboratory, Department of Plant Protection, Faculty of Agriculture, Ankara University, Ankara, Turkey
| |
Collapse
|
37
|
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
|
38
|
Abstract
Hunger is a motivational state that drives eating and food-seeking behaviour. In a psychological sense, hunger sets the goal that guides an animal in the pursuit of food. The biological basis underlying this purposive, goal-directed nature of hunger has been under intense investigation. With its rich behavioural repertoire and genetically tractable nervous system, the fruit fly Drosophila melanogaster has emerged as an excellent model system for studying the neural basis of hunger and hunger-driven behaviour. Here, we review our current understanding of how hunger is sensed, encoded and translated into foraging and feeding behaviours in the fruit fly.
Collapse
Affiliation(s)
- Suewei Lin
- 1 Institute of Molecular Biology, Academia Sinica , Taipei , Taiwan, Republic of China.,2 Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center , Taipei , Taiwan, Republic of China
| | - Bhagyashree Senapati
- 1 Institute of Molecular Biology, Academia Sinica , Taipei , Taiwan, Republic of China.,2 Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Sciences, National Defense Medical Center , Taipei , Taiwan, Republic of China
| | - Chang-Hui Tsao
- 1 Institute of Molecular Biology, Academia Sinica , Taipei , Taiwan, Republic of China
| |
Collapse
|
39
|
Sudhakar SR, Pathak H, Rehman N, Fernandes J, Vishnu S, Varghese J. Insulin signalling elicits hunger-induced feeding in Drosophila. Dev Biol 2020; 459:87-99. [DOI: 10.1016/j.ydbio.2019.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/21/2022]
|
40
|
Identification of neuropeptides from eyestalk transcriptome profiling analysis of female oriental river prawn (Macrobrachium nipponense) under hypoxia and reoxygenation conditions. Comp Biochem Physiol B Biochem Mol Biol 2020; 241:110392. [DOI: 10.1016/j.cbpb.2019.110392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/05/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023]
|
41
|
Lim S, Yunusbaev U, Ilyasov R, Lee HS, Kwon HW. Abdominal contact of fluvalinate induces olfactory deficit in Apis mellifera. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2020; 164:221-227. [PMID: 32284130 DOI: 10.1016/j.pestbp.2020.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/22/2020] [Accepted: 02/01/2020] [Indexed: 06/11/2023]
Abstract
τ-Fluvalinate (fluvalinate) is a highly selective pyrethroid insecticide compound used for controlling ectoparasitic mites that cause major damages in honey bee colonies. Although honey bees have resistance and low toxicity to this xenobiotic chemical, little is known about the effects of this chemical on sensory modulation and behaviors in honey bees. Here we addressed the effect on olfactory cognition at the behavioral, molecular, and neurophysiological levels. First, we found that topical application of fluvalinate to honeybee abdomen elicited somewhat severe toxicity to honey bees. Furthermore, honeybees treated with sublethal doses of fluvalinate showed a significant decrease in olfactory responses. At the molecular level, there was no change in gene expression levels of odorant receptor co-receptor (Orco), which is important for electrical conductivity induced by odorant binding in insects. Rather, small neuropeptide F (sNPF) signaling pathway was involved in olfactory fluctuation after treatment of fluvalinate. This indicates that olfactory deficits by abdominal contact of fluvalinate may stem from various internal molecular pathways in honey bees.
Collapse
Affiliation(s)
- Sooho Lim
- Department of Life Sciences & Convergence Research Center for Insect Vectors, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea.
| | - Ural Yunusbaev
- Department of Life Sciences & Convergence Research Center for Insect Vectors, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| | - Rustem Ilyasov
- Department of Life Sciences & Convergence Research Center for Insect Vectors, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea; Institute of Biochemistry and Genetics, Ufa Federal Research Centre, Russian Academy of Sciences, Ufa, Russia
| | - Hyun Sook Lee
- Department of Life Sciences & Convergence Research Center for Insect Vectors, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea
| | - Hyung Wook Kwon
- Department of Life Sciences & Convergence Research Center for Insect Vectors, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon 22012, Republic of Korea.
| |
Collapse
|
42
|
Liao S, Post S, Lehmann P, Veenstra JA, Tatar M, Nässel DR. Regulatory Roles of Drosophila Insulin-Like Peptide 1 (DILP1) in Metabolism Differ in Pupal and Adult Stages. Front Endocrinol (Lausanne) 2020; 11:180. [PMID: 32373064 PMCID: PMC7186318 DOI: 10.3389/fendo.2020.00180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/13/2020] [Indexed: 01/12/2023] Open
Abstract
The insulin/IGF-signaling pathway is central in control of nutrient-dependent growth during development, and in adult physiology and longevity. Eight insulin-like peptides (DILP1-8) have been identified in Drosophila, and several of these are known to regulate growth, metabolism, reproduction, stress responses, and lifespan. However, the functional role of DILP1 is far from understood. Previous work has shown that dilp1/DILP1 is transiently expressed mainly during the pupal stage and the first days of adult life. Here, we study the role of dilp1 in the pupa, as well as in the first week of adult life, and make some comparisons to dilp6 that displays a similar pupal expression profile, but is expressed in fat body rather than brain neurosecretory cells. We show that mutation of dilp1 diminishes organismal weight during pupal development, whereas overexpression increases it, similar to dilp6 manipulations. No growth effects of dilp1 or dilp6 manipulations were detected during larval development. We next show that dilp1 and dilp6 increase metabolic rate in the late pupa and promote lipids as the primary source of catabolic energy. Effects of dilp1 manipulations can also be seen in the adult fly. In newly eclosed female flies, survival during starvation is strongly diminished in dilp1 mutants, but not in dilp2 and dilp1/dilp2 mutants, whereas in older flies, only the double mutants display reduced starvation resistance. Starvation resistance is not affected in male dilp1 mutant flies, suggesting a sex dimorphism in dilp1 function. Overexpression of dilp1 also decreases survival during starvation in female flies and increases egg laying and decreases egg to pupal viability. In conclusion, dilp1 and dilp6 overexpression promotes metabolism and growth of adult tissues during the pupal stage, likely by utilization of stored lipids. Some of the effects of the dilp1 manipulations may carry over from the pupa to affect physiology in young adults, but our data also suggest that dilp1 signaling is important in metabolism and stress resistance in the adult stage.
Collapse
Affiliation(s)
- Sifang Liao
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Stephanie Post
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, United States
| | - Philipp Lehmann
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Jan A. Veenstra
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine (CNRS UMR5287), University of Bordeaux, Pessac, France
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, United States
| | - Dick R. Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
- *Correspondence: Dick R. Nässel
| |
Collapse
|
43
|
Oh Y, Lai JSY, Mills HJ, Erdjument-Bromage H, Giammarinaro B, Saadipour K, Wang JG, Abu F, Neubert TA, Suh GSB. A glucose-sensing neuron pair regulates insulin and glucagon in Drosophila. Nature 2019; 574:559-564. [PMID: 31645735 PMCID: PMC6857815 DOI: 10.1038/s41586-019-1675-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 09/16/2019] [Indexed: 01/08/2023]
Abstract
Although glucose-sensing neurons were identified more than 50 years ago, the physiological role of glucose sensing in metazoans remains unclear. Here we identify a pair of glucose-sensing neurons with bifurcated axons in the brain of Drosophila. One axon branch projects to insulin-producing cells to trigger the release of Drosophila insulin-like peptide 2 (dilp2) and the other extends to adipokinetic hormone (AKH)-producing cells to inhibit secretion of AKH, the fly analogue of glucagon. These axonal branches undergo synaptic remodelling in response to changes in their internal energy status. Silencing of these glucose-sensing neurons largely disabled the response of insulin-producing cells to glucose and dilp2 secretion, disinhibited AKH secretion in corpora cardiaca and caused hyperglycaemia, a hallmark feature of diabetes mellitus. We propose that these glucose-sensing neurons maintain glucose homeostasis by promoting the secretion of dilp2 and suppressing the release of AKH when haemolymph glucose levels are high.
Collapse
Affiliation(s)
- Yangkyun Oh
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Jason Sih-Yu Lai
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- QPS-Qualitix Taiwan, Ren-Ai Road, Taipei, Taiwan
| | - Holly J Mills
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Ascend Public Charter Schools, New York, NY, USA
| | - Hediye Erdjument-Bromage
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Benno Giammarinaro
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Vision Sciences Graduate Program, School of Optometry, UC Berkeley, Berkeley, CA, USA
| | - Khalil Saadipour
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Justin G Wang
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Farhan Abu
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Thomas A Neubert
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Greg S B Suh
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
44
|
Ahmad M, He L, Perrimon N. Regulation of insulin and adipokinetic hormone/glucagon production in flies. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e360. [PMID: 31379062 DOI: 10.1002/wdev.360] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/28/2019] [Accepted: 07/09/2019] [Indexed: 12/25/2022]
Abstract
Metabolic homeostasis is under strict regulation of humoral factors across various taxa. In particular, insulin and glucagon, referred to in Drosophila as Drosophila insulin-like peptides (DILPs) and adipokinetic hormone (AKH), respectively, are key hormones that regulate metabolism in most metazoa. While much is known about the regulation of DILPs, the mechanisms regulating AKH/glucagon production is still poorly understood. In this review, we describe the various factors that regulate the production of DILPs and AKH and emphasize the need for future studies to decipher how energy homeostasis is governed in Drosophila. This article is categorized under: Invertebrate Organogenesis > Flies Signaling Pathways > Global Signaling Mechanisms.
Collapse
Affiliation(s)
- Muhammad Ahmad
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Li He
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, Massachusetts.,Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
45
|
Dhiman N, Shweta K, Tendulkar S, Deshpande G, Ratnaparkhi GS, Ratnaparkhi A. Drosophila Mon1 constitutes a novel node in the brain-gonad axis that is essential for female germline maturation. Development 2019; 146:146/13/dev166504. [PMID: 31292144 DOI: 10.1242/dev.166504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/23/2019] [Indexed: 01/16/2023]
Abstract
Monensin-sensitive 1 (Mon1) is an endocytic regulator that participates in the conversion of Rab5-positive early endosomes to Rab7-positive late endosomes. In Drosophila, loss of mon1 leads to sterility as the mon1 mutant females have extremely small ovaries with complete absence of late stage egg chambers - a phenotype reminiscent of mutations in the insulin pathway genes. Here, we show that expression of many Drosophila insulin-like peptides (ILPs) is reduced in mon1 mutants and feeding mon1 adults an insulin-rich diet can rescue the ovarian defects. Surprisingly, however, mon1 functions in the tyramine/octopaminergic neurons (OPNs) and not in the ovaries or the insulin-producing cells (IPCs). Consistently, knockdown of mon1 in only the OPNs is sufficient to mimic the ovarian phenotype, while expression of the gene in the OPNs alone can 'rescue' the mutant defect. Last, we have identified ilp3 and ilp5 as critical targets of mon1. This study thus identifies mon1 as a novel molecular player in the brain-gonad axis and underscores the significance of inter-organ systemic communication during development.
Collapse
Affiliation(s)
- Neena Dhiman
- Agarkar Research Institute (ARI), Pune, India.,Indian Institute of Science Education & Research (IISER), Pune, India
| | | | - Shweta Tendulkar
- Indian Institute of Science Education & Research (IISER), Pune, India
| | - Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | | | | |
Collapse
|
46
|
Nagy D, Cusumano P, Andreatta G, Anduaga AM, Hermann-Luibl C, Reinhard N, Gesto J, Wegener C, Mazzotta G, Rosato E, Kyriacou CP, Helfrich-Förster C, Costa R. Peptidergic signaling from clock neurons regulates reproductive dormancy in Drosophila melanogaster. PLoS Genet 2019; 15:e1008158. [PMID: 31194738 PMCID: PMC6592559 DOI: 10.1371/journal.pgen.1008158] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/25/2019] [Accepted: 04/25/2019] [Indexed: 11/18/2022] Open
Abstract
With the approach of winter, many insects switch to an alternative protective developmental program called diapause. Drosophila melanogaster females overwinter as adults by inducing a reproductive arrest that is characterized by inhibition of ovarian development at previtellogenic stages. The insulin producing cells (IPCs) are key regulators of this process, since they produce and release insulin-like peptides that act as diapause-antagonizing hormones. Here we show that in D. melanogaster two neuropeptides, Pigment Dispersing Factor (PDF) and short Neuropeptide F (sNPF) inhibit reproductive arrest, likely through modulation of the IPCs. In particular, genetic manipulations of the PDF-expressing neurons, which include the sNPF-producing small ventral Lateral Neurons (s-LNvs), modulated the levels of reproductive dormancy, suggesting the involvement of both neuropeptides. We expressed a genetically encoded cAMP sensor in the IPCs and challenged brain explants with synthetic PDF and sNPF. Bath applications of both neuropeptides increased cAMP levels in the IPCs, even more so when they were applied together, suggesting a synergistic effect. Bath application of sNPF additionally increased Ca2+ levels in the IPCs. Our results indicate that PDF and sNPF inhibit reproductive dormancy by maintaining the IPCs in an active state.
Collapse
Affiliation(s)
- Dóra Nagy
- Department of Biology, University of Padova, Padova, Italy
| | - Paola Cusumano
- Department of Biology, University of Padova, Padova, Italy
| | | | - Ane Martin Anduaga
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christiane Hermann-Luibl
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Nils Reinhard
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - João Gesto
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christian Wegener
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | | | - Ezio Rosato
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Charalambos P. Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Rodolfo Costa
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
47
|
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: 211] [Impact Index Per Article: 35.2] [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
|
48
|
Fadda M, Hasakiogullari I, Temmerman L, Beets I, Zels S, Schoofs L. Regulation of Feeding and Metabolism by Neuropeptide F and Short Neuropeptide F in Invertebrates. Front Endocrinol (Lausanne) 2019; 10:64. [PMID: 30837946 PMCID: PMC6389622 DOI: 10.3389/fendo.2019.00064] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/23/2019] [Indexed: 12/19/2022] Open
Abstract
Numerous neuropeptide systems have been implicated to coordinately control energy homeostasis, both centrally and peripherally. However, the vertebrate neuropeptide Y (NPY) system has emerged as the best described one regarding this biological process. The protostomian ortholog of NPY is neuropeptide F, characterized by an RXRF(Y)amide carboxyterminal motif. A second neuropeptide system is short NPF, characterized by an M/T/L/FRF(W)amide carboxyterminal motif. Although both short and long NPF neuropeptide systems display carboxyterminal sequence similarities, they are evolutionary distant and likely already arose as separate signaling systems in the common ancestor of deuterostomes and protostomes, indicating the functional importance of both. Both NPF and short-NPF systems seem to have roles in the coordination of feeding across bilaterian species, but during chordate evolution, the short NPF system appears to have been lost or evolved into the prolactin releasing peptide signaling system, which regulates feeding and has been suggested to be orthologous to sNPF. Here we review the roles of both NPF and sNPF systems in the regulation of feeding and metabolism in invertebrates.
Collapse
Affiliation(s)
| | | | | | | | | | - Liliane Schoofs
- Department of Biology, Functional Genomics and Proteomics, KU Leuven, Leuven, Belgium
| |
Collapse
|
49
|
Post S, Liao S, Yamamoto R, Veenstra JA, Nässel DR, Tatar M. Drosophila insulin-like peptide dilp1 increases lifespan and glucagon-like Akh expression epistatic to dilp2. Aging Cell 2019; 18:e12863. [PMID: 30511458 PMCID: PMC6351851 DOI: 10.1111/acel.12863] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/24/2018] [Accepted: 09/27/2018] [Indexed: 12/17/2022] Open
Abstract
Insulin/IGF signaling (IIS) regulates essential processes including development, metabolism, and aging. The Drosophila genome encodes eight insulin/IGF‐like peptide (dilp) paralogs, including tandem‐encoded dilp1 and dilp2. Many reports show that longevity is increased by manipulations that decrease DILP2 levels. It has been shown that dilp1 is expressed primarily in pupal stages, but also during adult reproductive diapause. Here, we find that dilp1 is also highly expressed in adult dilp2 mutants under nondiapause conditions. The inverse expression of dilp1 and dilp2 suggests these genes interact to regulate aging. Here, we study dilp1 and dilp2 single and double mutants to describe epistatic and synergistic interactions affecting longevity, metabolism, and adipokinetic hormone (AKH), the functional homolog of glucagon. Mutants of dilp2 extend lifespan and increase Akh mRNA and protein in a dilp1‐dependent manner. Loss of dilp1 alone has no impact on these traits, whereas transgene expression of dilp1 increases lifespan in dilp1 − dilp2 double mutants. On the other hand, dilp1 and dilp2 redundantly or synergistically interact to control circulating sugar, starvation resistance, and compensatory dilp5 expression. These interactions do not correlate with patterns for how dilp1 and dilp2 affect longevity and AKH. Thus, repression or loss of dilp2 slows aging because its depletion induces dilp1, which acts as a pro‐longevity factor. Likewise, dilp2 regulates Akh through epistatic interaction with dilp1. Akh and glycogen affect aging in Caenorhabditis elegans and Drosophila. Our data suggest that dilp2 modulates lifespan in part by regulating Akh, and by repressing dilp1, which acts as a pro‐longevity insulin‐like peptide.
Collapse
Affiliation(s)
- Stephanie Post
- Department of Molecular Biology, Cell Biology and Biochemistry; Brown University; Providence Rhode Island
- Department of Ecology and Evolutionary Biology; Brown University; Providence Rhode Island
| | - Sifang Liao
- Department of Zoology; Stockholm University; Stockholm Sweden
| | - Rochele Yamamoto
- Department of Ecology and Evolutionary Biology; Brown University; Providence Rhode Island
| | - Jan A. Veenstra
- Institut de Neurosciences Cognitives et Intégratives d’Aquitaine (CNRS UMR5287); University of Bordeaux; Pessac France
| | - Dick R. Nässel
- Department of Zoology; Stockholm University; Stockholm Sweden
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology; Brown University; Providence Rhode Island
| |
Collapse
|
50
|
Matsumoto S, Ohara A, Nagai-Okatani C, Zhou YJ, Fujinaga D, Seike H, Nagata S. Antagonistic Effect of Short Neuropeptide F on Allatotropin-Inhibited Feeding Motivation of the Silkworm Larva, Bombyx mori. Zoolog Sci 2019; 36:58-67. [PMID: 31116539 DOI: 10.2108/zs180119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/30/2018] [Indexed: 11/17/2022]
Abstract
Here, we demonstrated an antagonistic effect of short neuropeptide F (sNPF) in modulating feeding motivation in the silkworm Bombyx mori; sNPF reduced the feeding-delaying effects caused by administration of an inhibitory peptide, allatotropin (AT). In situ hybridization and MALDI-TOF MS analysis revealed the presence of three subtypes of sNPFs (sNPF-1, -2, and -3) in the midgut enteroendocrine cells. Ca2+-imaging analyses revealed that three subtypes of sNPF receptors (sNPFRs) (BNGR-A7, -A10, and -A11) showed different affinities with the three subtypes of sNPFs. In addition, sNPF activated its signaling via ERK phosphorylation in the midgut, while mixture of sNPF and AT reduced the phosphorylation level, agreeing with the results of behavioral assay. Together, our current findings suggest that intestinal sNPF positively modulates the feeding motivation by reducing the inhibitory effects by AT within the midgut.
Collapse
Affiliation(s)
- Sumihiro Matsumoto
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa City, Chiba 277-8562, Japan
| | - Ayako Ohara
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Chiaki Nagai-Okatani
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan.,Current affiliation: Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-8568, Japan
| | - Yi-Jun Zhou
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa City, Chiba 277-8562, Japan.,Research Fellow of Japan Society for the Promotion of Science (JSPS), Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Daiki Fujinaga
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa City, Chiba 277-8562, Japan
| | - Hitomi Seike
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa City, Chiba 277-8562, Japan
| | - Shinji Nagata
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa City, Chiba 277-8562, Japan,
| |
Collapse
|