1
|
Wegener C, Amini E, Cavieres-Lepe J, Ewer J. Neuronal and endocrine mechanisms underlying the circadian gating of eclosion: insights from Drosophila. CURRENT OPINION IN INSECT SCIENCE 2024; 66:101286. [PMID: 39461671 DOI: 10.1016/j.cois.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
The circadian rhythm of adult emergence (aka eclosion) of the fruit fly Drosophila is a classic behavioural read-out that served in the first characterisation of the key features of circadian clocks and was also used for the identification of the first clock genes. Rhythmic eclosion requires the central clock in the brain, as well as a peripheral clock in the steroidogenic prothoracic gland. Here, we review recent findings on the timing and neuroendocrine coupling mechanisms of the two clocks. These findings identify rhythmic prothoracicotropic hormone and downstream ERK signalling as the main coupling pathway and show that the two clocks impose daily rhythmicity to the temporal pattern of eclosion by regulating the timing of the very last steps in metamorphosis.
Collapse
Affiliation(s)
- Christian Wegener
- Julius-Maximilians-Universität Würzburg, Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Am Hubland, 97074 Würzburg, Germany.
| | - Emad Amini
- Julius-Maximilians-Universität Würzburg, Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Am Hubland, 97074 Würzburg, Germany
| | - Javier Cavieres-Lepe
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - John Ewer
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile; Instituto de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
2
|
Zhao J, Yin J, Wang Z, Shen J, Dong M, Yan S. Complicated gene network for regulating feeding behavior: novel efficient target for pest management. PEST MANAGEMENT SCIENCE 2024. [PMID: 39390706 DOI: 10.1002/ps.8459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/10/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024]
Abstract
Feeding behavior is a fundamental activity for insects, which is essential for their growth, development and reproduction. The regulation of their feeding behavior is a complicated process influenced by a variety of factors, including external stimuli and internal physiological signals. The current review introduces the signaling pathways in brain, gut and fat body involved in insect feeding behavior, and provides a series of target genes for developing RNA pesticides. Additionally, this review summaries the current challenges for the identification and application of functional genes involved in feeding behavior, and finally proposes the future research direction. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiajia Zhao
- Sanya Institute of China Agricultural University, Sanya, China
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jiaming Yin
- Sanya Institute of China Agricultural University, Sanya, China
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Zeng Wang
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Shen
- Sanya Institute of China Agricultural University, Sanya, China
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Min Dong
- Sanya Institute of China Agricultural University, Sanya, China
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| | - Shuo Yan
- Sanya Institute of China Agricultural University, Sanya, China
- Department of Plant Biosecurity, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
3
|
Li J, Ning C, Liu Y, Deng B, Wang B, Shi K, Wang R, Fang R, Zhou C. The function of juvenile-adult transition axis in female sexual receptivity of Drosophila melanogaster. eLife 2024; 12:RP92545. [PMID: 39240259 PMCID: PMC11379460 DOI: 10.7554/elife.92545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Female sexual receptivity is essential for reproduction of a species. Neuropeptides play the main role in regulating female receptivity. However, whether neuropeptides regulate female sexual receptivity during the neurodevelopment is unknown. Here, we found the peptide hormone prothoracicotropic hormone (PTTH), which belongs to the insect PG (prothoracic gland) axis, negatively regulated virgin female receptivity through ecdysone during neurodevelopment in Drosophila melanogaster. We identified PTTH neurons as doublesex-positive neurons, they regulated virgin female receptivity before the metamorphosis during the third-instar larval stage. PTTH deletion resulted in the increased EcR-A expression in the whole newly formed prepupae. Furthermore, the ecdysone receptor EcR-A in pC1 neurons positively regulated virgin female receptivity during metamorphosis. The decreased EcR-A in pC1 neurons induced abnormal morphological development of pC1 neurons without changing neural activity. Among all subtypes of pC1 neurons, the function of EcR-A in pC1b neurons was necessary for virgin female copulation rate. These suggested that the changes of synaptic connections between pC1b and other neurons decreased female copulation rate. Moreover, female receptivity significantly decreased when the expression of PTTH receptor Torso was reduced in pC1 neurons. This suggested that PTTH not only regulates female receptivity through ecdysone but also through affecting female receptivity associated neurons directly. The PG axis has similar functional strategy as the hypothalamic-pituitary-gonadal axis in mammals to trigger the juvenile-adult transition. Our work suggests a general mechanism underlying which the neurodevelopment during maturation regulates female sexual receptivity.
Collapse
Affiliation(s)
- Jing Li
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chao Ning
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yaohua Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Department of Plant Protection, Shanxi Agricultural University, Jinzhong, China
| | - Bowen Deng
- Chinese Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Zhongguancun Life Sciences Park, Beijing, China
| | - Bingcai Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kai Shi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rencong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ruixin Fang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Chuan Zhou
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
4
|
Chen S, Fan H, Ran C, Hong Y, Feng H, Yue Z, Zhang H, Pontarotti P, Xu A, Huang S. The IL-17 pathway intertwines with neurotrophin and TLR/IL-1R pathways since its domain shuffling origin. Proc Natl Acad Sci U S A 2024; 121:e2400903121. [PMID: 38683992 PMCID: PMC11087794 DOI: 10.1073/pnas.2400903121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
The IL-17 pathway displays remarkably diverse functional modes between different subphyla, classes, and even orders, yet its driving factors remains elusive. Here, we demonstrate that the IL-17 pathway originated through domain shuffling between a Toll-like receptor (TLR)/IL-1R pathway and a neurotrophin-RTK (receptor-tyrosine-kinase) pathway (a Trunk-Torso pathway). Unlike other new pathways that evolve independently, the IL-17 pathway remains intertwined with its donor pathways throughout later evolution. This intertwining not only influenced the gains and losses of domains and components in the pathway but also drove the diversification of the pathway's functional modes among animal lineages. For instance, we reveal that the crustacean female sex hormone, a neurotrophin inducing sex differentiation, could interact with IL-17Rs and thus be classified as true IL-17s. Additionally, the insect prothoracicotropic hormone, a neurotrophin initiating ecdysis in Drosophila by binding to Torso, could bind to IL-17Rs in other insects. Furthermore, IL-17R and TLR/IL-1R pathways maintain crosstalk in amphioxus and zebrafish. Moreover, the loss of the Death domain in the pathway adaptor connection to IκB kinase and stress-activated protein kinase (CIKSs) dramatically reduced their abilities to activate nuclear factor-kappaB (NF-κB) and activator protein 1 (AP-1) in amphioxus and zebrafish. Reinstating this Death domain not only enhanced NF-κB/AP-1 activation but also strengthened anti-bacterial immunity in zebrafish larvae. This could explain why the mammalian IL-17 pathway, whose CIKS also lacks Death, is considered a weak signaling activator, relying on synergies with other pathways. Our findings provide insights into the functional diversity of the IL-17 pathway and unveil evolutionary principles that could govern the pathway and be used to redesign and manipulate it.
Collapse
Affiliation(s)
- Shenghui Chen
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao266237, China
| | - Huiping Fan
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Chenrui Ran
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Yun Hong
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Huixiong Feng
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Zirui Yue
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Hao Zhang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
| | - Pierre Pontarotti
- MEPHI (Microbes, Evolution, Phylogénie et Infection), Aix Marseille Université, Marseille, France
| | - Anlong Xu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing100029, China
| | - Shengfeng Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangzhou510275, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao266237, China
| |
Collapse
|
5
|
Cavieres-Lepe J, Amini E, Zabel M, Nässel DR, Stanewsky R, Wegener C, Ewer J. Timed receptor tyrosine kinase signaling couples the central and a peripheral circadian clock in Drosophila. Proc Natl Acad Sci U S A 2024; 121:e2308067121. [PMID: 38442160 PMCID: PMC10945756 DOI: 10.1073/pnas.2308067121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 02/01/2024] [Indexed: 03/07/2024] Open
Abstract
Circadian clocks impose daily periodicities to behavior, physiology, and metabolism. This control is mediated by a central clock and by peripheral clocks, which are synchronized to provide the organism with a unified time through mechanisms that are not fully understood. Here, we characterized in Drosophila the cellular and molecular mechanisms involved in coupling the central clock and the peripheral clock located in the prothoracic gland (PG), which together control the circadian rhythm of emergence of adult flies. The time signal from central clock neurons is transmitted via small neuropeptide F (sNPF) to neurons that produce the neuropeptide Prothoracicotropic Hormone (PTTH), which is then translated into daily oscillations of Ca2+ concentration and PTTH levels. PTTH signaling is required at the end of metamorphosis and transmits time information to the PG through changes in the expression of the PTTH receptor tyrosine kinase (RTK), TORSO, and of ERK phosphorylation, a key component of PTTH transduction. In addition to PTTH, we demonstrate that signaling mediated by other RTKs contributes to the rhythmicity of emergence. Interestingly, the ligand to one of these receptors (Pvf2) plays an autocrine role in the PG, which may explain why both central brain and PG clocks are required for the circadian gating of emergence. Our findings show that the coupling between the central and the PG clock is unexpectedly complex and involves several RTKs that act in concert and could serve as a paradigm to understand how circadian clocks are coordinated.
Collapse
Affiliation(s)
- Javier Cavieres-Lepe
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso2360102, Chile
| | - Emad Amini
- Julius-Maximilians-Universität Würzburg, Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Am Hubland, Würzburg97074, Germany
| | - Maia Zabel
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso2360102, Chile
| | - Dick R. Nässel
- Department of Zoology, Stockholm University, 10691Stockholm, Sweden
| | - Ralf Stanewsky
- Institute of Neuro- and Behavioral Biology, Multiscale Imaging Centre, University of Münster, 48149Münster, Germany
| | - Christian Wegener
- Julius-Maximilians-Universität Würzburg, Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Am Hubland, Würzburg97074, Germany
| | - John Ewer
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso2360102, Chile
- Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso2360102, Chile
| |
Collapse
|
6
|
Kanaoka Y, Onodera K, Watanabe K, Hayashi Y, Usui T, Uemura T, Hattori Y. Inter-organ Wingless/Ror/Akt signaling regulates nutrient-dependent hyperarborization of somatosensory neurons. eLife 2023; 12:79461. [PMID: 36647607 PMCID: PMC9844989 DOI: 10.7554/elife.79461] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 12/11/2022] [Indexed: 01/18/2023] Open
Abstract
Nutrition in early life has profound effects on an organism, altering processes such as organogenesis. However, little is known about how specific nutrients affect neuronal development. Dendrites of class IV dendritic arborization neurons in Drosophila larvae become more complex when the larvae are reared on a low-yeast diet compared to a high-yeast diet. Our systematic search for key nutrients revealed that the neurons increase their dendritic terminal densities in response to a combined deficiency in vitamins, metal ions, and cholesterol. The deficiency of these nutrients upregulates Wingless in a closely located tissue, body wall muscle. Muscle-derived Wingless activates Akt in the neurons through the receptor tyrosine kinase Ror, which promotes the dendrite branching. In larval muscles, the expression of wingless is regulated not only in this key nutrient-dependent manner, but also by the JAK/STAT signaling pathway. Additionally, the low-yeast diet blunts neuronal light responsiveness and light avoidance behavior, which may help larvae optimize their survival strategies under low-nutritional conditions. Together, our studies illustrate how the availability of specific nutrients affects neuronal development through inter-organ signaling.
Collapse
Affiliation(s)
| | - Koun Onodera
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Kaori Watanabe
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Yusaku Hayashi
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Tadao Usui
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Tadashi Uemura
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
- Research Center for Dynamic Living Systems, Kyoto UniversityKyotoJapan
- AMED-CRESTTokyoJapan
| | - Yukako Hattori
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
- JST FORESTTokyoJapan
| |
Collapse
|
7
|
Ramakrishnan P, Joshi A, Tulasi M, Yadav P. Monochromatic visible lights modulate the timing of pre-adult developmental traits in Drosophila melanogaster. PHOTOCHEMICAL & PHOTOBIOLOGICAL SCIENCES : OFFICIAL JOURNAL OF THE EUROPEAN PHOTOCHEMISTRY ASSOCIATION AND THE EUROPEAN SOCIETY FOR PHOTOBIOLOGY 2022; 22:867-881. [PMID: 36583814 DOI: 10.1007/s43630-022-00358-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Light exposure impacts several aspects of Drosophila development including the establishment of circadian rhythms, neuroendocrine regulation, life-history traits, etc. Introduction of artificial lights in the environment has caused almost all animals to develop ecological and physiological adaptations. White light which comprises different lights of differing wavelengths shortens the lifespan in fruit flies Drosophila melanogaster. The wavelength-specific effects of white light on Drosophila development remains poorly understood. In this study, we show that different wavelengths of white light differentially modulate Drosophila development in all its concomitant stages when maintained in a 12-h light: 12-h dark photoperiod. We observed that exposure to different monochromatic lights significantly alters larval behaviours such as feeding rate and phototaxis that influence pre-adult development. Larvae grown under shorter wavelengths of light experienced an altered feeding rate. Similarly, larvae were found to avoid shorter wavelengths of light but were highly attracted to the longer wavelengths of light. Most of the developmental processes were greatly accelerated under the green light regime while in other light regimes, the effects were highly varied. Interestingly, pre-adult survivorship remained unaltered across all light regimes but light exposure was found to show its impact on sex determination. Our study for the first time reveals how different wavelengths of white light modulate Drosophila development which in the future might help in developing non-invasive therapies and effective pest measures.
Collapse
Affiliation(s)
- Pooja Ramakrishnan
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, 613401, India
| | - Aradhana Joshi
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, 613401, India
| | - Muntimadugu Tulasi
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, 613401, India
| | - Pankaj Yadav
- Fly Laboratory # 210, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, Tamil Nadu, 613401, India.
| |
Collapse
|
8
|
A two-choice assay for noxious light avoidance with temporal distribution analysis in Drosophila melanogaster larvae. STAR Protoc 2022; 3:101787. [PMID: 36317171 PMCID: PMC9617195 DOI: 10.1016/j.xpro.2022.101787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Two-choice assays allow assessing of different behaviors including light avoidance in Drosophila larvae. Typically, the readout is limited to a preference index at a specific end point. We provide a detailed protocol to set up light avoidance assays and map the temporal distribution of larvae based on analysis of larval intensities. We describe the assay setup and implementation of scripts for analysis, which can be easily adapted to other two-choice assays and different model organisms. For complete details on the use and execution of this protocol, please refer to Imambocus et al. (2022). Preparation and staging of Drosophila larvae for behavioral analysis Video-based recording of larval behavior in two-choice light avoidance assays Script-based video processing and extraction of larval distribution Quantitative analysis of temporal distribution of animals during light avoidance
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
|
9
|
Liu J, Jin T, Ran L, Zhao Z, Zhu R, Xie G, Bi X. Profiling ATM regulated genes in Drosophila at physiological condition and after ionizing radiation. Hereditas 2022; 159:41. [PMID: 36271387 PMCID: PMC9587650 DOI: 10.1186/s41065-022-00254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
Background ATM (ataxia-telangiectasia mutated) protein kinase is highly conserved in metazoan, and plays a critical role at DNA damage response, oxidative stress, metabolic stress, immunity, RNA biogenesis etc. Systemic profiling of ATM regulated genes, including protein-coding genes, miRNAs, and long non-coding RNAs, will greatly improve our understanding of ATM functions and its regulation. Results 1) differentially expressed protein-coding genes, miRNAs, and long non-coding RNAs in atm mutated flies were identified at physiological condition and after X-ray irradiation. 2) functions of differentially expressed genes in atm mutated flies, regardless of protein-coding genes or non-coding RNAs, are closely related with metabolic process, immune response, DNA damage response or oxidative stress. 3) these phenomena are persistent after irradiation. 4) there is a cross-talk regulation towards miRNAs by ATM, E2f1, and p53 during development and after irradiation. 5) knock-out flies or knock-down flies of most irradiation-induced miRNAs were sensitive to ionizing radiation. Conclusions We provide a valuable resource of protein-coding genes, miRNAs, and long non-coding RNAs, for understanding ATM functions and regulations. Our work provides the new evidence of inter-dependence among ATM-E2F1-p53 for the regulation of miRNAs. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-022-00254-9.
Collapse
Affiliation(s)
- Jun Liu
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Tianyu Jin
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Lanxi Ran
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Ze Zhao
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Rui Zhu
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Gangcai Xie
- School of Medicine, Nantong University, Nantong, 226001, China.
| | - Xiaolin Bi
- School of Medicine, Nantong University, Nantong, 226001, China. .,College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
10
|
Circuit analysis reveals a neural pathway for light avoidance in Drosophila larvae. Nat Commun 2022; 13:5274. [PMID: 36071059 PMCID: PMC9452580 DOI: 10.1038/s41467-022-33059-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 08/31/2022] [Indexed: 11/30/2022] Open
Abstract
Understanding how neural circuits underlie behaviour is challenging even in the connectome era because it requires a combination of anatomical and functional analyses. This is exemplified in the circuit underlying the light avoidance behaviour displayed by Drosophila melanogaster larvae. While this behaviour is robust and the nervous system relatively simple, the circuit is only partially delineated with some contradictions among studies. Here, we devise trans-Tango MkII, an offshoot of the transsynaptic circuit tracing tool trans-Tango, and implement it in anatomical tracing together with functional analysis. We use neuronal inhibition to test necessity of particular neuronal types in light avoidance and selective neuronal activation to examine sufficiency in rescuing light avoidance deficiencies exhibited by photoreceptor mutants. Our studies reveal a four-order circuit for light avoidance connecting the light-detecting photoreceptors with a pair of neuroendocrine cells via two types of clock neurons. This approach can be readily expanded to studying other circuits. Studying neural circuits requires a multipronged approach. Here, the authors present a transsynaptic tracing tool in fruit fly larvae and combine it with neuronal inhibition and activation to study the circuit underlying light avoidance behaviour.
Collapse
|
11
|
Wagle M, Zarei M, Lovett-Barron M, Poston KT, Xu J, Ramey V, Pollard KS, Prober DA, Schulkin J, Deisseroth K, Guo S. Brain-wide perception of the emotional valence of light is regulated by distinct hypothalamic neurons. Mol Psychiatry 2022; 27:3777-3793. [PMID: 35484242 PMCID: PMC9613822 DOI: 10.1038/s41380-022-01567-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/25/2022] [Accepted: 04/06/2022] [Indexed: 02/08/2023]
Abstract
Salient sensory stimuli are perceived by the brain, which guides both the timing and outcome of behaviors in a context-dependent manner. Light is such a stimulus, which is used in treating mood disorders often associated with a dysregulated hypothalamic-pituitary-adrenal stress axis. Relationships between the emotional valence of light and the hypothalamus, and how they interact to exert brain-wide impacts remain unclear. Employing larval zebrafish with analogous hypothalamic systems to mammals, we show in free-swimming animals that hypothalamic corticotropin releasing factor (CRFHy) neurons promote dark avoidance, and such role is not shared by other hypothalamic peptidergic neurons. Single-neuron projection analyses uncover processes extended by individual CRFHy neurons to multiple targets including sensorimotor and decision-making areas. In vivo calcium imaging uncovers a complex and heterogeneous response of individual CRFHy neurons to the light or dark stimulus, with a reduced overall sum of CRF neuronal activity in the presence of light. Brain-wide calcium imaging under alternating light/dark stimuli further identifies distinct and distributed photic response neuronal types. CRFHy neuronal ablation increases an overall representation of light in the brain and broadly enhances the functional connectivity associated with an exploratory brain state. These findings delineate brain-wide photic perception, uncover a previously unknown role of CRFHy neurons in regulating the perception and emotional valence of light, and suggest that light therapy may alleviate mood disorders through reducing an overall sum of CRF neuronal activity.
Collapse
Affiliation(s)
- Mahendra Wagle
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA
| | - Mahdi Zarei
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA
| | - Matthew Lovett-Barron
- Department of Bioengineering, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Kristina Tyler Poston
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA
| | - Jin Xu
- Tianqiao and Chrissy Chen Institute for Neuroscience, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Vince Ramey
- Biophysics Graduate Group, University of California, Berkeley, CA, USA
- Invitae Inc., San Francisco, CA, USA
| | - Katherine S Pollard
- Gladstone Institute of Data Science & Biotechnology, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - David A Prober
- Tianqiao and Chrissy Chen Institute for Neuroscience, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Jay Schulkin
- Department of Obstetrics & Gynecology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Karl Deisseroth
- Department of Bioengineering, Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94143-2811, USA.
- Programs in Human Genetics and Biological Sciences, Kavli Institute of Fundamental Neuroscience, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Bakar Aging Research Institute, University of California, San Francisco, CA, 94143-2811, USA.
| |
Collapse
|
12
|
Liu X, Yang S, Yao Y, Wu S, Wu P, Zhai Z. Opsin1 regulates light-evoked avoidance behavior in Aedes albopictus. BMC Biol 2022; 20:110. [PMID: 35549721 PMCID: PMC9103082 DOI: 10.1186/s12915-022-01308-0] [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: 10/28/2021] [Accepted: 04/25/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Mosquitoes locate a human host by integrating various sensory cues including odor, thermo, and vision. However, their innate light preference and its genetic basis that may predict the spatial distribution of mosquitoes, a prerequisite to encounter a potential host and initiate host-seeking behaviors, remains elusive. RESULTS Here, we first studied mosquito visual features and surprisingly uncovered that both diurnal (Aedes aegypti and Aedes albopictus) and nocturnal (Culex quinquefasciatus) mosquitoes significantly avoided stronger light when given choices. With consistent results from multiple assays, we found that such negative phototaxis maintained throughout development to adult stages. Notably, female mosquitoes significantly preferred to bite hosts in a shaded versus illuminated area. Furthermore, silencing Opsin1, a G protein-coupled receptor that is most enriched in compound eyes, abolished light-evoked avoidance behavior of Aedes albopictus and attenuated photonegative behavior in Aedes aegypti. Finally, we found that field-collected Aedes albopictus also prefers darker area in an Opsin1-dependent manner. CONCLUSIONS This study reveals that mosquitoes consistently prefer darker environment and identifies the first example of a visual molecule that modulates mosquito photobehavior.
Collapse
Affiliation(s)
- Xinyi Liu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Shuzhen Yang
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Yuan Yao
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Si Wu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Pa Wu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Zongzhao Zhai
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
13
|
Jaszczak JS, DeVault L, Jan LY, Jan YN. Steroid hormone signaling activates thermal nociception during Drosophila peripheral nervous system development. eLife 2022; 11:e76464. [PMID: 35353036 PMCID: PMC8967384 DOI: 10.7554/elife.76464] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 12/27/2022] Open
Abstract
Sensory neurons enable animals to detect environmental changes and avoid harm. An intriguing open question concerns how the various attributes of sensory neurons arise in development. Drosophila melanogaster larvae undergo a behavioral transition by robustly activating a thermal nociceptive escape behavior during the second half of larval development (third instar). The Class IV dendritic arborization (C4da) neurons are multimodal sensors which tile the body wall of Drosophila larvae and detect nociceptive temperature, light, and mechanical force. In contrast to the increase in nociceptive behavior in the third instar, we find that ultraviolet light-induced Ca2+ activity in C4da neurons decreases during the same period of larval development. Loss of ecdysone receptor has previously been shown to reduce nociception in third instar larvae. We find that ligand-dependent activation of ecdysone signaling is sufficient to promote nociceptive responses in second instar larvae and suppress expression of subdued (encoding a TMEM16 channel). Reduction of subdued expression in second instar C4da neurons not only increases thermal nociception but also decreases the response to ultraviolet light. Thus, steroid hormone signaling suppresses subdued expression to facilitate the sensory switch of C4da neurons. This regulation of a developmental sensory switch through steroid hormone regulation of channel expression raises the possibility that ion channel homeostasis is a key target for tuning the development of sensory modalities.
Collapse
Affiliation(s)
- Jacob S Jaszczak
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Laura DeVault
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Department of Developmental Biology, Washington University Medical SchoolSaint LouisUnited States
| | - Lily Yeh Jan
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Yuh Nung Jan
- Department of Physiology, Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| |
Collapse
|
14
|
Costa CP, Okamoto N, Orr M, Yamanaka N, Woodard SH. Convergent Loss of Prothoracicotropic Hormone, A Canonical Regulator of Development, in Social Bee Evolution. Front Physiol 2022; 13:831928. [PMID: 35242055 PMCID: PMC8887954 DOI: 10.3389/fphys.2022.831928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 11/21/2022] Open
Abstract
The evolution of insect sociality has repeatedly involved changes in developmental events and their timing. Here, we propose the hypothesis that loss of a canonical regulator of moulting and metamorphosis, prothoracicotropic hormone (PTTH), and its receptor, Torso, is associated with the evolution of sociality in bees. Specifically, we posit that the increasing importance of social influences on early developmental timing in social bees has led to their decreased reliance on PTTH, which connects developmental timing with abiotic cues in solitary insects. At present, the evidence to support this hypothesis includes the absence of genes encoding PTTH and Torso from all fully-sequenced social bee genomes and its presence in all available genomes of solitary bees. Based on the bee phylogeny, the most parsimonious reconstruction of evolutionary events is that this hormone and its receptor have been lost multiple times, across independently social bee lineages. These gene losses shed light on possible molecular and cellular mechanisms that are associated with the evolution of social behavior in bees. We outline the available evidence for our hypothesis, and then contextualize it in light of what is known about developmental cues in social and solitary bees, and the multiple precedences of major developmental changes in social insects.
Collapse
Affiliation(s)
- Claudinéia P Costa
- Department of Entomology, University of California, Riverside, Riverside, CA, United States
| | - Naoki Okamoto
- Department of Entomology, University of California, Riverside, Riverside, CA, United States.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Michael Orr
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Naoki Yamanaka
- Department of Entomology, University of California, Riverside, Riverside, CA, United States
| | - S Hollis Woodard
- Department of Entomology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
15
|
Texada MJ, Lassen M, Pedersen LH, Koyama T, Malita A, Rewitz K. Insulin signaling couples growth and early maturation to cholesterol intake in Drosophila. Curr Biol 2022; 32:1548-1562.e6. [PMID: 35245460 DOI: 10.1016/j.cub.2022.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 12/10/2021] [Accepted: 02/04/2022] [Indexed: 11/28/2022]
Abstract
Nutrition is one of the most important influences on growth and the timing of maturational transitions including mammalian puberty and insect metamorphosis. Childhood obesity is associated with precocious puberty, but the assessment mechanism that links body fat to early maturation is unknown. During development, the intake of nutrients promotes signaling through insulin-like systems that govern the growth of cells and tissues and also regulates the timely production of the steroid hormones that initiate the juvenile-adult transition. We show here that the dietary lipid cholesterol, which is required as a component of cell membranes and as a substrate for steroid biosynthesis, also governs body growth and maturation in Drosophila via promoting the expression and release of insulin-like peptides. This nutritional input acts via the nutrient sensor TOR, which is regulated by the Niemann-Pick-type-C 1 (Npc1) cholesterol transporter, in the glia of the blood-brain barrier and cells of the adipose tissue to remotely drive systemic insulin signaling and body growth. Furthermore, increasing intracellular cholesterol levels in the steroid-producing prothoracic gland strongly promotes endoreduplication, leading to an accelerated attainment of a nutritional checkpoint that normally ensures that animals do not initiate maturation prematurely. These findings, therefore, show that a Npc1-TOR signaling system couples the sensing of the lipid cholesterol with cellular and systemic growth control and maturational timing, which may help explain both the link between cholesterol and cancer as well as the connection between body fat (obesity) and early puberty.
Collapse
Affiliation(s)
- Michael J Texada
- Department of Biology, Section for Cell and Neurobiology, University of Copenhagen, Universitetsparken 15, Building 3, 2100 Copenhagen, Denmark.
| | - Mette Lassen
- Department of Biology, Section for Cell and Neurobiology, University of Copenhagen, Universitetsparken 15, Building 3, 2100 Copenhagen, Denmark
| | - Lisa H Pedersen
- Department of Biology, Section for Cell and Neurobiology, University of Copenhagen, Universitetsparken 15, Building 3, 2100 Copenhagen, Denmark
| | - Takashi Koyama
- Department of Biology, Section for Cell and Neurobiology, University of Copenhagen, Universitetsparken 15, Building 3, 2100 Copenhagen, Denmark
| | - Alina Malita
- Department of Biology, Section for Cell and Neurobiology, University of Copenhagen, Universitetsparken 15, Building 3, 2100 Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, Section for Cell and Neurobiology, University of Copenhagen, Universitetsparken 15, Building 3, 2100 Copenhagen, Denmark.
| |
Collapse
|
16
|
Hao S, Gestrich JY, Zhang X, Xu M, Wang X, Liu L, Wei H. Neurotransmitters Affect Larval Development by Regulating the Activity of Prothoracicotropic Hormone-Releasing Neurons in Drosophila melanogaster. Front Neurosci 2021; 15:653858. [PMID: 34975366 PMCID: PMC8718639 DOI: 10.3389/fnins.2021.653858] [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: 01/15/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Ecdysone, an essential insect steroid hormone, promotes larval metamorphosis by coordinating growth and maturation. In Drosophila melanogaster, prothoracicotropic hormone (PTTH)-releasing neurons are considered to be the primary promoting factor in ecdysone biosynthesis. Recently, studies have reported that the regulatory mechanisms of PTTH release in Drosophila larvae are controlled by different neuropeptides, including allatostatin A and corazonin. However, it remains unclear whether neurotransmitters provide input to PTTH neurons and control the metamorphosis in Drosophila larvae. Here, we report that the neurotransmitters acetylcholine (ACh) affect larval development by modulating the activity of PTTH neurons. By downregulating the expression of different subunits of nicotinic ACh receptors in PTTH neurons, pupal volume was significantly increased, whereas pupariation timing was relatively unchanged. We also identified that PTTH neurons were excited by ACh application ex vivo in a dose-dependent manner via ionotropic nicotinic ACh receptors. Moreover, in our Ca2+ imaging experiments, relatively low doses of OA caused increased Ca2+ levels in PTTH neurons, whereas higher doses led to decreased Ca2+ levels. We also demonstrated that a low dose of OA was conveyed through OA β-type receptors. Additionally, our electrophysiological experiments revealed that PTTH neurons produced spontaneous activity in vivo, which provides the possibility of the bidirectional regulation, coming from neurons upstream of PTTH cells in Drosophila larvae. In summary, our findings indicate that several different neurotransmitters are involved in the regulation of larval metamorphosis by altering the activity of PTTH neurons in Drosophila.
Collapse
Affiliation(s)
- Shun Hao
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Julia Yvonne Gestrich
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xin Zhang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Mengbo Xu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Xinwei Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| | - Li Liu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Hongying Wei
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Imambocus BN, Zhou F, Formozov A, Wittich A, Tenedini FM, Hu C, Sauter K, Macarenhas Varela E, Herédia F, Casimiro AP, Macedo A, Schlegel P, Yang CH, Miguel-Aliaga I, Wiegert JS, Pankratz MJ, Gontijo AM, Cardona A, Soba P. A neuropeptidergic circuit gates selective escape behavior of Drosophila larvae. Curr Biol 2021; 32:149-163.e8. [PMID: 34798050 DOI: 10.1016/j.cub.2021.10.069] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 10/05/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022]
Abstract
Animals display selective escape behaviors when faced with environmental threats. Selection of the appropriate response by the underlying neuronal network is key to maximizing chances of survival, yet the underlying network mechanisms are so far not fully understood. Using synapse-level reconstruction of the Drosophila larval network paired with physiological and behavioral readouts, we uncovered a circuit that gates selective escape behavior for noxious light through acute and input-specific neuropeptide action. Sensory neurons required for avoidance of noxious light and escape in response to harsh touch, each converge on discrete domains of neuromodulatory hub neurons. We show that acute release of hub neuron-derived insulin-like peptide 7 (Ilp7) and cognate relaxin family receptor (Lgr4) signaling in downstream neurons are required for noxious light avoidance, but not harsh touch responses. Our work highlights a role for compartmentalized circuit organization and neuropeptide release from regulatory hubs, acting as central circuit elements gating escape responses.
Collapse
Affiliation(s)
- Bibi Nusreen Imambocus
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Fangmin Zhou
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Andrey Formozov
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Annika Wittich
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Federico M Tenedini
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Chun Hu
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Kathrin Sauter
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ednilson Macarenhas Varela
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Fabiana Herédia
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Andreia P Casimiro
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - André Macedo
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal
| | - Philipp Schlegel
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Chung-Hui Yang
- Department of Neurobiology, Duke University Medical School, 427E Bryan Research, Durham, NC 27710, USA
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - J Simon Wiegert
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Michael J Pankratz
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Alisson M Gontijo
- Integrative Biomedicine Laboratory, CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Rua do Instituto Bacteriológico 5, 1150-082 Lisbon, Portugal; The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Albert Cardona
- HHMI Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK; Department of Physiology, Development, and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany; Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany.
| |
Collapse
|
18
|
Brain adiponectin signaling controls peripheral insulin response in Drosophila. Nat Commun 2021; 12:5633. [PMID: 34561451 PMCID: PMC8463608 DOI: 10.1038/s41467-021-25940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 07/16/2021] [Indexed: 11/27/2022] Open
Abstract
The brain plays a key role in energy homeostasis, detecting nutrients, metabolites and circulating hormones from peripheral organs and integrating this information to control food intake and energy expenditure. Here, we show that a group of neurons in the Drosophila larval brain expresses the adiponectin receptor (AdipoR) and controls systemic growth and metabolism through insulin signaling. We identify glucose-regulated protein 78 (Grp78) as a circulating antagonist of AdipoR function produced by fat cells in response to dietary sugar. We further show that central AdipoR signaling inhibits peripheral Juvenile Hormone (JH) response, promoting insulin signaling. In conclusion, we identify a neuroendocrine axis whereby AdipoR-positive neurons control systemic insulin response. Circulating adiponectin controls sensitivity to insulin in tissues. Here, Arquier et al. show that adiponectin receptor activity in neurons of the Drosophila brain controls insulin response in peripheral tissues via juvenile hormone signaling.
Collapse
|
19
|
Coordination among multiple receptor tyrosine kinase signals controls Drosophila developmental timing and body size. Cell Rep 2021; 36:109644. [PMID: 34469735 PMCID: PMC8428980 DOI: 10.1016/j.celrep.2021.109644] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 05/10/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
In holometabolous insects, metamorphic timing and body size are controlled by a neuroendocrine axis composed of the ecdysone-producing prothoracic gland (PG) and its presynaptic neurons (PGNs) producing PTTH. Although PTTH/Torso signaling is considered the primary mediator of metamorphic timing, recent studies indicate that other unidentified PGN-derived factors also affect timing. Here, we demonstrate that the receptor tyrosine kinases anaplastic lymphoma kinase (Alk) and PDGF and VEGF receptor-related (Pvr), function in coordination with PTTH/Torso signaling to regulate pupariation timing and body size. Both Alk and Pvr trigger Ras/Erk signaling in the PG to upregulate expression of ecdysone biosynthetic enzymes, while Alk also suppresses autophagy by activating phosphatidylinositol 3-kinase (PI3K)/Akt. The Alk ligand Jelly belly (Jeb) is produced by the PGNs and serves as a second PGN-derived tropic factor, while Pvr activation mainly relies on autocrine signaling by PG-derived Pvf2 and Pvf3. These findings illustrate that a combination of juxtacrine and autocrine signaling regulates metamorphic timing, the defining event of holometabolous development.
Collapse
|
20
|
Poe AR, Mace KD, Kayser MS. Getting into rhythm: developmental emergence of circadian clocks and behaviors. FEBS J 2021; 289:6576-6588. [PMID: 34375504 DOI: 10.1111/febs.16157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/30/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022]
Abstract
Circadian clocks keep time to coordinate diverse behaviors and physiological functions. While molecular circadian rhythms are evident during early development, most behavioral rhythms, such as sleep-wake, do not emerge until far later. Here, we examine the development of circadian clocks, outputs, and behaviors across phylogeny, with a particular focus on Drosophila. We explore potential mechanisms for how central clocks and circadian output loci establish communication, and discuss why from an evolutionary perspective sleep-wake and other behavioral rhythms emerge long after central clocks begin keeping time.
Collapse
Affiliation(s)
- Amy R Poe
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kyla D Mace
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Pharmacology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew S Kayser
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Chronobiology and Sleep Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Focal laser stimulation of fly nociceptors activates distinct axonal and dendritic Ca 2+ signals. Biophys J 2021; 120:3222-3233. [PMID: 34175294 PMCID: PMC8390926 DOI: 10.1016/j.bpj.2021.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/11/2021] [Accepted: 06/02/2021] [Indexed: 11/22/2022] Open
Abstract
Drosophila class IV neurons are polymodal nociceptors that detect noxious mechanical, thermal, optical, and chemical stimuli. Escape behaviors in response to attacks by parasitoid wasps are dependent on class IV cells, whose highly branched dendritic arbors form a fine meshwork that is thought to enable detection of the wasp’s needle-like ovipositor barb. To understand how mechanical stimuli trigger cellular responses, we used a focused 405-nm laser to create highly localized lesions to probe the precise position needed to evoke responses. By imaging calcium signals in dendrites, axons, and soma in response to stimuli of varying positions, intensities, and spatial profiles, we discovered that there are two distinct nociceptive pathways. Direct stimulation to dendrites (the contact pathway) produces calcium responses in axons, dendrites, and the cell body, whereas stimulation adjacent to the dendrite (the noncontact pathway) produces calcium responses in the axons only. We interpret the noncontact pathway as damage to adjacent cells releasing diffusible molecules that act on the dendrites. Axonal responses have higher sensitivities and shorter latencies. In contrast, dendritic responses have lower sensitivities and longer latencies. Stimulation of finer, distal dendrites leads to smaller responses than stimulation of coarser, proximal dendrites, as expected if the contact response depends on the geometric overlap of the laser profile and the dendrite diameter. Because the axon signals to the central nervous system to trigger escape behaviors, we propose that the density of the dendritic meshwork is high not only to enable direct contact with the ovipositor but also to enable neuronal activation via diffusing signals from damaged surrounding cells. Dendritic contact evokes responses throughout the dendritic arbor, even to regions distant and distal from the stimulus. These dendrite-wide calcium signals may facilitate hyperalgesia or cellular morphological changes after dendritic damage.
Collapse
|
22
|
Neural substrates involved in the cognitive information processing in teleost fish. Anim Cogn 2021; 24:923-946. [PMID: 33907938 PMCID: PMC8360893 DOI: 10.1007/s10071-021-01514-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/25/2021] [Accepted: 03/06/2021] [Indexed: 02/04/2023]
Abstract
Over the last few decades, it has been shown that fish, comprising the largest group of vertebrates and in many respects one of the least well studied, possess many cognitive abilities comparable to those of birds and mammals. Despite a plethora of behavioural studies assessing cognition abilities and an abundance of neuroanatomical studies, only few studies have aimed to or in fact identified the neural substrates involved in the processing of cognitive information. In this review, an overview of the currently available studies addressing the joint research topics of cognitive behaviour and neuroscience in teleosts (and elasmobranchs wherever possible) is provided, primarily focusing on two fundamentally different but complementary approaches, i.e. ablation studies and Immediate Early Gene (IEG) analyses. More recently, the latter technique has become one of the most promising methods to visualize neuronal populations activated in specific brain areas, both during a variety of cognitive as well as non-cognition-related tasks. While IEG studies may be more elegant and potentially easier to conduct, only lesion studies can help researchers find out what information animals can learn or recall prior to and following ablation of a particular brain area.
Collapse
|
23
|
Ahuja A, Tyagi PK, Tyagi S, Kumar A, Kumar M, Sharifi-Rad J. Potential of Pueraria tuberosa (Willd.) DC. to rescue cognitive decline associated with BACE1 protein of Alzheimer's disease on Drosophila model: An integrated molecular modeling and in vivo approach. Int J Biol Macromol 2021; 179:586-600. [PMID: 33705837 DOI: 10.1016/j.ijbiomac.2021.03.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 01/04/2023]
Abstract
The indispensable role of Beta-site amyloid precursor protein cleaving enzyme-1 (BACE1) in Amyloid beta (Aβ) plaques generation and Aβ-mediated synaptic dysfunctions makes it a crucial target for therapeutic intervention in Alzheimer's disease (AD). In order to find out the potential inhibitors of BACE1, the present study focused on five phytochemicals from Pueraria tuberosa, namely, daidzin, genistin, mangiferin, puerarin, and tuberosin. A molecular docking study showed that all five phytochemicals presented the strongest BACE1 inhibition. Integrated molecular dynamics simulations and free energy calculations demonstrated that all five natural compounds have stable and favorable energies causing strong binding with the pocket site of BACE1 on 50 ns. All these molecules also passed Lipinski's rule of five. To validate the molecular modeling based findings, we primarily targeted the cognitive decline associated with BACE1 expression in AD flies with P. tuberosa. Significant improvement in cognitive decline was observed in AD flies in different behavioral assays such as Larval crawling assay (16.38%), Larval light preference assay (26.39%), Climbing assay (32.97%), Cold sensitivity assay (43.6%), and Thermal sensitivity assay (44.42%). The present findings suggest that P. tuberosa may be considered as a promising dietary supplement that can significantly ameliorate cognitive decline caused by BACE1 in Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Anami Ahuja
- Research Scholar, Department of Biotechnology, Dr. A.P.J. Abdul Kalam Technical University, Lucknow 226031, Uttar Pradesh, India; Department of Biotechnology, Meerut Institute of Engineering and Technology, Meerut 250005, Uttar Pradesh, India.
| | - Pankaj Kumar Tyagi
- Department of Biotechnology Engineering, Noida Institute of Engineering & Technology, Greater Noida 201306, Uttar Pradesh, India.
| | - Shruti Tyagi
- Department of Biotechnology Engineering, Noida Institute of Engineering & Technology, Greater Noida 201306, Uttar Pradesh, India
| | - Anuj Kumar
- Advanced Centre for Computational and Applied Biotechnology, Uttarakhand Council for Biotechnology (UCB), Dehradun 248007, Uttarakhand, India
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR - Central Institute for Research on Cotton Technology, Mumbai 400019, India
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador.
| |
Collapse
|
24
|
Romão D, Muzzopappa M, Barrio L, Milán M. The Upd3 cytokine couples inflammation to maturation defects in Drosophila. Curr Biol 2021; 31:1780-1787.e6. [PMID: 33609452 DOI: 10.1016/j.cub.2021.01.080] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/05/2020] [Accepted: 01/22/2021] [Indexed: 12/20/2022]
Abstract
Developmental transitions, such as puberty or metamorphosis, are tightly controlled by steroid hormones and can be delayed by the appearance of growth abnormalities, developmental tumors, or inflammatory disorders such as inflammatory bowel disease or cystic fibrosis.1-4 Here, we used a highly inflammatory epithelial model of malignant transformation in Drosophila5,6 to unravel the role of Upd3-a cytokine with homology to interleukin-6-and the JAK/STAT signaling pathway in coupling inflammation to a delay in metamorphosis. We present evidence that Upd3 produced by malignant and nearby cell populations signals to the prothoracic gland-an endocrine tissue primarily dedicated to the production of the steroid hormone ecdysone-to activate JAK/STAT and bantam microRNA (miRNA) and to delay metamorphosis. Upd cytokines produced by the tumor site contribute to increasing the systemic levels of Upd3 by amplifying its expression levels in a cell-autonomous manner and by inducing Upd3 expression in neighboring tissues in a non-autonomous manner, culminating in a major systemic response to prevent larvae from initiating pupa transition. Our results identify a new regulatory network impacting on ecdysone biosynthesis and provide new insights into the potential role of inflammatory cytokines and the JAK/STAT signaling pathway in coupling inflammation to delays in puberty.
Collapse
Affiliation(s)
- Daniela Romão
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Mariana Muzzopappa
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Lara Barrio
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Marco Milán
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
25
|
de la Riva-Carrasco R, Perez-Pandolfo S, Suárez Freire S, Romero NM, Bhujabal Z, Johansen T, Wappner P, Melani M. The immunophilin Zonda controls regulated exocytosis in endocrine and exocrine tissues. Traffic 2021; 22:111-122. [PMID: 33336828 DOI: 10.1111/tra.12777] [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: 06/28/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 11/30/2022]
Abstract
Exocytosis is a fundamental process in physiology, that ensures communication between cells, organs and even organisms. Hormones, neuropeptides and antibodies, among other cargoes are packed in exocytic vesicles that need to reach and fuse with the plasma membrane to release their content to the extracellular milieu. Hundreds of proteins participate in this process and several others in its regulation. We report here a novel component of the exocytic machinery, the Drosophila transmembrane immunophilin Zonda (Zda), previously found to participate in autophagy. Zda is highly expressed in secretory tissues, and regulates exocytosis in at least three of them: the ring gland, insulin-producing cells and the salivary gland. Using the salivary gland as a model system, we found that Zda is required at final steps of the exocytic process for fusion of secretory granules to the plasma membrane. In a genetic screen we identified the small GTPase RalA as a crucial regulator of secretory granule exocytosis that is required, similarly to Zda, for fusion between the secretory granule and the plasma membrane.
Collapse
Affiliation(s)
| | - Sebastián Perez-Pandolfo
- Laboratorio de Genética y Fisiología Molecular, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Sofía Suárez Freire
- Laboratorio de Genética y Fisiología Molecular, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Nuria M Romero
- Université Côte d'Azur, INRA, CNRS, Institut Sophia Agrobiotech, Sophia Antipolis, France
| | - Zambarlal Bhujabal
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Pablo Wappner
- Laboratorio de Genética y Fisiología Molecular, Fundación Instituto Leloir, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Melani
- Laboratorio de Genética y Fisiología Molecular, Fundación Instituto Leloir, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
26
|
Nässel DR, Zandawala M. Hormonal axes in Drosophila: regulation of hormone release and multiplicity of actions. Cell Tissue Res 2020; 382:233-266. [PMID: 32827072 PMCID: PMC7584566 DOI: 10.1007/s00441-020-03264-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Hormones regulate development, as well as many vital processes in the daily life of an animal. Many of these hormones are peptides that act at a higher hierarchical level in the animal with roles as organizers that globally orchestrate metabolism, physiology and behavior. Peptide hormones can act on multiple peripheral targets and simultaneously convey basal states, such as metabolic status and sleep-awake or arousal across many central neuronal circuits. Thereby, they coordinate responses to changing internal and external environments. The activity of neurosecretory cells is controlled either by (1) cell autonomous sensors, or (2) by other neurons that relay signals from sensors in peripheral tissues and (3) by feedback from target cells. Thus, a hormonal signaling axis commonly comprises several components. In mammals and other vertebrates, several hormonal axes are known, such as the hypothalamic-pituitary-gonad axis or the hypothalamic-pituitary-thyroid axis that regulate reproduction and metabolism, respectively. It has been proposed that the basic organization of such hormonal axes is evolutionarily old and that cellular homologs of the hypothalamic-pituitary system can be found for instance in insects. To obtain an appreciation of the similarities between insect and vertebrate neurosecretory axes, we review the organization of neurosecretory cell systems in Drosophila. Our review outlines the major peptidergic hormonal pathways known in Drosophila and presents a set of schemes of hormonal axes and orchestrating peptidergic systems. The detailed organization of the larval and adult Drosophila neurosecretory systems displays only very basic similarities to those in other arthropods and vertebrates.
Collapse
Affiliation(s)
- Dick R. Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Meet Zandawala
- Department of Neuroscience, Brown University, Providence, RI USA
| |
Collapse
|
27
|
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: 64] [Impact Index Per Article: 16.0] [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
|
28
|
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: 62] [Impact Index Per Article: 15.5] [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
|
29
|
Christensen CF, Koyama T, Nagy S, Danielsen ET, Texada MJ, Halberg KA, Rewitz K. Ecdysone-dependent feedback regulation of prothoracicotropic hormone controls the timing of developmental maturation. Development 2020; 147:dev188110. [PMID: 32631830 PMCID: PMC7390634 DOI: 10.1242/dev.188110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022]
Abstract
The activation of a neuroendocrine system that induces a surge in steroid production is a conserved initiator of the juvenile-to-adult transition in many animals. The trigger for maturation is the secretion of brain-derived neuropeptides, yet the mechanisms controlling the timely onset of this event remain ill-defined. Here, we show that a regulatory feedback circuit controlling the Drosophila neuropeptide Prothoracicotropic hormone (PTTH) triggers maturation onset. We identify the Ecdysone Receptor (EcR) in the PTTH-expressing neurons (PTTHn) as a regulator of developmental maturation onset. Loss of EcR in these PTTHn impairs PTTH signaling, which delays maturation. We find that the steroid ecdysone dose-dependently affects Ptth transcription, promoting its expression at lower concentrations and inhibiting it at higher concentrations. Our findings indicate the existence of a feedback circuit in which rising ecdysone levels trigger, via EcR activity in the PTTHn, the PTTH surge that generates the maturation-inducing ecdysone peak toward the end of larval development. Because steroid feedback is also known to control the vertebrate maturation-inducing hypothalamic-pituitary-gonadal axis, our findings suggest an overall conservation of the feedback-regulatory neuroendocrine circuitry that controls the timing of maturation initiation.
Collapse
Affiliation(s)
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - Stanislav Nagy
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - E Thomas Danielsen
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - Kenneth A Halberg
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark
| |
Collapse
|
30
|
Kamiyama T, Sun W, Tani N, Nakamura A, Niwa R. Poly(A) Binding Protein Is Required for Nuclear Localization of the Ecdysteroidogenic Transcription Factor Molting Defective in the Prothoracic Gland of Drosophila melanogaster. Front Genet 2020; 11:636. [PMID: 32676099 PMCID: PMC7333772 DOI: 10.3389/fgene.2020.00636] [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: 02/19/2020] [Accepted: 05/26/2020] [Indexed: 11/28/2022] Open
Abstract
Steroid hormone signaling contributes to the development of multicellular organisms. In insects, ecdysteroids, like ecdysone and the more biologically-active derivative 20-hydroxyecdysone (20E), promote molting and metamorphosis. Ecdysone is biosynthesized in the prothoracic gland (PG), via several steps catalyzed by ecdysteroidogenic enzymes that are encoded by Halloween genes. The spatio-temporal expression pattern of ecdysteroidogenic genes is strictly controlled, resulting in a proper fluctuation of the 20E titer during insect development. However, their transcriptional regulatory mechanism is still elusive. A previous study has found that the polyadenylated tail [poly(A)] deadenylation complex, called Carbon catabolite repressor 4-Negative on TATA (CCR4-NOT) regulates the expression of spookier (spok), which encodes one of the ecdysteroidogenic enzymes in the fruit fly Drosophila melanogaster. Based on this finding, we speculated whether any other poly(A)-related protein also regulates spok expression. In this study, we reported that poly(A) binding protein (Pabp) is involved in spok expression by regulating nuclear localization of the transcription factor molting defective (Mld). When pabp was knocked down specifically in the PG by transgenic RNAi, both spok mRNA and Spok protein levels were significantly reduced. In addition, the spok promoter-driven green fluorescence protein (GFP) signal was also reduced in the pabp-RNAi PG, suggesting that Pabp is involved in the transcriptional regulation of spok. We next examined which transcription factors are responsible for Pabp-dependent transcriptional regulation. Among the transcription factors acting in the PG, we primarily focused on the zinc-finger transcription factor Mld, as Mld is essential for spok transcription. Mld was localized in the nucleus of the control PG cells, while Mld abnormally accumulated in the cytoplasm of pabp-RNAi PG cells. In contrast, pabp-RNAi did not affect the nuclear localization of other transcription factors, including ventral vein lacking (Vvl) and POU domain motif 3 (Pdm3), in PG cells. From these results, we propose that Pabp regulates subcellular localization in the PG, specifically of the transcription factor Mld, in the context of ecdysone biosynthesis.
Collapse
Affiliation(s)
- Takumi Kamiyama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Wei Sun
- Laboratory of Evolutionary and Functional Genomics, School of Life Sciences, Chongqing University, Chongqing, China.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Naoki Tani
- Department of Germline Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Akira Nakamura
- Department of Germline Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
31
|
Assessing the potential of lignin nanoparticles as drug carrier: Synthesis, cytotoxicity and genotoxicity studies. Int J Biol Macromol 2020; 152:786-802. [DOI: 10.1016/j.ijbiomac.2020.02.311] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/30/2022]
|
32
|
Complexity and plasticity in honey bee phototactic behaviour. Sci Rep 2020; 10:7872. [PMID: 32398687 PMCID: PMC7217928 DOI: 10.1038/s41598-020-64782-y] [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: 11/06/2019] [Accepted: 04/21/2020] [Indexed: 11/28/2022] Open
Abstract
The ability to move towards or away from a light source, namely phototaxis, is essential for a number of species to find the right environmental niche and may have driven the appearance of simple visual systems. In this study we ask if the later evolution of more complex visual systems was accompanied by a sophistication of phototactic behaviour. The honey bee is an ideal model organism to tackle this question, as it has an elaborate visual system, demonstrates exquisite abilities for visual learning and performs phototaxis. Our data suggest that in this insect, phototaxis has wavelength specific properties and is a highly dynamical response including multiple decision steps. In addition, we show that previous experience with a light (through exposure or classical aversive conditioning) modulates the phototactic response. This plasticity is dependent on the wavelength used, with blue being more labile than green or ultraviolet. Wavelength, intensity and past experience are integrated into an overall valence for each light that determines phototactic behaviour in honey bees. Thus, our results support the idea that complex visual systems allow sophisticated phototaxis. Future studies could take advantage of these findings to better understand the neuronal circuits underlying this processing of the visual information.
Collapse
|
33
|
Imura E, Shimada-Niwa Y, Nishimura T, Hückesfeld S, Schlegel P, Ohhara Y, Kondo S, Tanimoto H, Cardona A, Pankratz MJ, Niwa R. The Corazonin-PTTH Neuronal Axis Controls Systemic Body Growth by Regulating Basal Ecdysteroid Biosynthesis in Drosophila melanogaster. Curr Biol 2020; 30:2156-2165.e5. [PMID: 32386525 DOI: 10.1016/j.cub.2020.03.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/10/2020] [Accepted: 03/19/2020] [Indexed: 12/21/2022]
Abstract
Steroid hormones play key roles in development, growth, and reproduction in various animal phyla [1]. The insect steroid hormone, ecdysteroid, coordinates growth and maturation, represented by molting and metamorphosis [2]. In Drosophila melanogaster, the prothoracicotropic hormone (PTTH)-producing neurons stimulate peak levels of ecdysteroid biosynthesis for maturation [3]. Additionally, recent studies on PTTH signaling indicated that basal levels of ecdysteroid negatively affect systemic growth prior to maturation [4-8]. However, it remains unclear how PTTH signaling is regulated for basal ecdysteroid biosynthesis. Here, we report that Corazonin (Crz)-producing neurons regulate basal ecdysteroid biosynthesis by affecting PTTH neurons. Crz belongs to gonadotropin-releasing hormone (GnRH) superfamily, implying an analogous role in growth and maturation [9]. Inhibition of Crz neuronal activity increased pupal size, whereas it hardly affected pupariation timing. This phenotype resulted from enhanced growth rate and a delay in ecdysteroid elevation during the mid-third instar larval (L3) stage. Interestingly, Crz receptor (CrzR) expression in PTTH neurons was higher during the mid- than the late-L3 stage. Silencing of CrzR in PTTH neurons increased pupal size, phenocopying the inhibition of Crz neuronal activity. When Crz neurons were optogenetically activated, a strong calcium response was observed in PTTH neurons during the mid-L3, but not the late-L3, stage. Furthermore, we found that octopamine neurons contact Crz neurons in the subesophageal zone (SEZ), transmitting signals for systemic growth. Together, our results suggest that the Crz-PTTH neuronal axis modulates ecdysteroid biosynthesis in response to octopamine, uncovering a regulatory neuroendocrine system in the developmental transition from growth to maturation.
Collapse
Affiliation(s)
- Eisuke Imura
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba 305-8572, Japan
| | - Yuko Shimada-Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 305-8577 Tsukuba, Japan.
| | | | - Sebastian Hückesfeld
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn 53115, Germany
| | - Philipp Schlegel
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn 53115, Germany
| | - Yuya Ohhara
- School of Food and Nutritional Sciences, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Shu Kondo
- Invertebrate Genetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Albert Cardona
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Michael J Pankratz
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Bonn 53115, Germany
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 305-8577 Tsukuba, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| |
Collapse
|
34
|
Asirim EZ, Humberg TH, Maier GL, Sprecher SG. Circadian and Genetic Modulation of Visually-Guided Navigation in Drosophila Larvae. Sci Rep 2020; 10:2752. [PMID: 32066794 PMCID: PMC7026142 DOI: 10.1038/s41598-020-59614-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/23/2020] [Indexed: 12/17/2022] Open
Abstract
Organisms possess an endogenous molecular clock which enables them to adapt to environmental rhythms and to synchronize their metabolism and behavior accordingly. Circadian rhythms govern daily oscillations in numerous physiological processes, and the underlying molecular components have been extensively described from fruit flies to mammals. Drosophila larvae have relatively simple nervous system compared to their adult counterparts, yet they both share a homologous molecular clock with mammals, governed by interlocking transcriptional feedback loops with highly conserved constituents. Larvae exhibit a robust light avoidance behavior, presumably enabling them to avoid predators and desiccation, and DNA-damage by exposure to ultraviolet light, hence are crucial for survival. Circadian rhythm has been shown to alter light-dark preference, however it remains unclear how distinct behavioral strategies are modulated by circadian time. To address this question, we investigate the larval visual navigation at different time-points of the day employing a computer-based tracking system, which allows detailed evaluation of distinct navigation strategies. Our results show that due to circadian modulation specific to light information processing, larvae avoid light most efficiently at dawn, and a functioning clock mechanism at both molecular and neuro-signaling level is necessary to conduct this modulation.
Collapse
Affiliation(s)
- Ece Z Asirim
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland
| | - Tim-Henning Humberg
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland
| | - G Larisa Maier
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland
| | - Simon G Sprecher
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
35
|
Delanoue R, Romero NM. Growth and Maturation in Development: A Fly's Perspective. Int J Mol Sci 2020; 21:E1260. [PMID: 32070061 PMCID: PMC7072963 DOI: 10.3390/ijms21041260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/02/2020] [Accepted: 02/10/2020] [Indexed: 01/09/2023] Open
Abstract
In mammals like humans, adult fitness is improved due to resource allocation, investing energy in the developmental growth process during the juvenile period, and in reproduction at the adult stage. Therefore, the attainment of their target body height/size co-occurs with the acquisition of maturation, implying a need for coordination between mechanisms that regulate organismal growth and maturation timing. Insects like Drosophila melanogaster also define their adult body size by the end of the juvenile larval period. Recent studies in the fly have shown evolutionary conservation of the regulatory pathways controlling growth and maturation, suggesting the existence of common coordinator mechanisms between them. In this review, we will present an overview of the significant advancements in the coordination mechanisms ensuring developmental robustness in Drosophila. We will include (i) the characterization of feedback mechanisms between maturation and growth hormones, (ii) the recognition of a relaxin-like peptide Dilp8 as a central processor coordinating juvenile regeneration and time of maturation, and (iii) the identification of a novel coordinator mechanism involving the AstA/KISS system.
Collapse
Affiliation(s)
- Renald Delanoue
- University Côte d’Azur, CNRS, Inserm, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | - Nuria M. Romero
- University Côte d’Azur, CNRS, Inserm, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
- Universitey Côte d’Azur, INRA, CNRS, Institut Sophia Agrobiotech, 06900 Sophia Antipolis, France
| |
Collapse
|
36
|
Galagovsky D, Depetris-Chauvin A, Manière G, Geillon F, Berthelot-Grosjean M, Noirot E, Alves G, Grosjean Y. Sobremesa L-type Amino Acid Transporter Expressed in Glia Is Essential for Proper Timing of Development and Brain Growth. Cell Rep 2019; 24:3156-3166.e4. [PMID: 30231999 PMCID: PMC6167638 DOI: 10.1016/j.celrep.2018.08.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 07/13/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
In Drosophila, ecdysone hormone levels determine the timing of larval development. Its production is regulated by the stereotypical rise in prothoracicotropic hormone (PTTH) levels. Additionally, ecdysone levels can also be modulated by nutrition (specifically by amino acids) through their action on Drosophila insulin-like peptides (Dilps). Moreover, in glia, amino-acid-sensitive production of Dilps regulates brain development. In this work, we describe the function of an SLC7 amino acid transporter, Sobremesa (Sbm). Larvae with reduced Sbm levels in glia remain in third instar for an additional 24 hr. These larvae show reduced brain growth with increased body size but do not show reduction in insulin signaling or production. Interestingly, Sbm downregulation in glia leads to reduced Ecdysone production and a surprising delay in the rise of PTTH levels. Our work highlights Sbm as a modulator of both brain development and the timing of larval development via an amino-acid-sensitive and Dilp-independent function of glia. Glia express the SLC7 amino acid transporter Sobremesa, which controls development Sobremesa downregulation in glia leads to contrasting effects: small brain and big body size Sobremesa downregulation results in reduced ecdysone production Sobremesa downregulation causes a delayed rise in PTTH
Collapse
Affiliation(s)
- Diego Galagovsky
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Ana Depetris-Chauvin
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France; Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Gérard Manière
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Flore Geillon
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Martine Berthelot-Grosjean
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Elodie Noirot
- Plateforme DImaCell, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Georges Alves
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Yael Grosjean
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000 Dijon, France.
| |
Collapse
|
37
|
Moss-Taylor L, Upadhyay A, Pan X, Kim MJ, O'Connor MB. Body Size and Tissue-Scaling Is Regulated by Motoneuron-Derived Activinß in Drosophila melanogaster. Genetics 2019; 213:1447-1464. [PMID: 31585954 PMCID: PMC6893369 DOI: 10.1534/genetics.119.302394] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/29/2019] [Indexed: 01/17/2023] Open
Abstract
Correct scaling of body and organ size is crucial for proper development, and the survival of all organisms. Perturbations in circulating hormones, including insulins and steroids, are largely responsible for changing body size in response to both genetic and environmental factors. Such perturbations typically produce adults whose organs and appendages scale proportionately with final size. The identity of additional factors that might contribute to scaling of organs and appendages with body size is unknown. Here, we report that loss-of-function mutations in DrosophilaActivinβ (Actβ), a member of the TGF-β superfamily, lead to the production of small larvae/pupae and undersized rare adult escapers. Morphometric measurements of escaper adult appendage size (wings and legs), as well as heads, thoraxes, and abdomens, reveal a disproportional reduction in abdominal size compared to other tissues. Similar size measurements of selected Actβ mutant larval tissues demonstrate that somatic muscle size is disproportionately smaller when compared to the fat body, salivary glands, prothoracic glands, imaginal discs, and brain. We also show that Actβ control of body size is dependent on canonical signaling through the transcription-factor dSmad2 and that it modulates the growth rate, but not feeding behavior, during the third-instar period. Tissue- and cell-specific knockdown, and overexpression studies, reveal that motoneuron-derived Actβ is essential for regulating proper body size and tissue scaling. These studies suggest that, unlike in vertebrates, where Myostatin and certain other Activin-like factors act as systemic negative regulators of muscle mass, in Drosophila, Actβ is a positive regulator of muscle mass that is directly delivered to muscles by motoneurons. We discuss the importance of these findings in coordinating proportional scaling of insect muscle mass to appendage size.
Collapse
Affiliation(s)
- Lindsay Moss-Taylor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Ambuj Upadhyay
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Xueyang Pan
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
38
|
Pan X, Neufeld TP, O'Connor MB. A Tissue- and Temporal-Specific Autophagic Switch Controls Drosophila Pre-metamorphic Nutritional Checkpoints. Curr Biol 2019; 29:2840-2851.e4. [PMID: 31422886 DOI: 10.1016/j.cub.2019.07.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/24/2019] [Accepted: 07/10/2019] [Indexed: 01/28/2023]
Abstract
Properly timed production of steroid hormones by endocrine tissues regulates juvenile-to-adult transitions in both mammals (puberty) and holometabolous insects (metamorphosis). Nutritional conditions influence the temporal control of the transition, but the mechanisms responsible are ill defined. Here we demonstrate that autophagy acts as an endocrine organ-specific, nutritionally regulated gating mechanism to help ensure productive metamorphosis in Drosophila. Autophagy in the endocrine organ is specifically stimulated by nutrient restriction at the early, but not the late, third-instar larva stage. The timing of autophagy induction correlates with the nutritional checkpoints, which inhibit precocious metamorphosis during nutrient restriction in undersized larvae. Suppression of autophagy causes dysregulated pupariation of starved larvae, which leads to pupal lethality, whereas forced autophagy induction results in developmental delay/arrest in well-fed animals. Induction of autophagy disrupts production of the steroid hormone ecdysone at the time of pupariation not by destruction of hormone biosynthetic capacity but rather by limiting the availability of the steroid hormone precursor cholesterol in the endocrine cells via a lipophagy mechanism. Interestingly, autophagy in the endocrine organ functions by interacting with the endolysosome system, yet shows multiple features not fully consistent with a canonical autophagy process. Taken together, our findings demonstrate an autophagy mechanism in endocrine cells that helps shape the nutritional checkpoints and guarantee a successful juvenile-to-adult transition in animals confronting nutritional stress.
Collapse
Affiliation(s)
- Xueyang Pan
- Department of Genetics, Cell Biology and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thomas P Neufeld
- Department of Genetics, Cell Biology and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
39
|
Mishra PK, Ekielski A, Mukherjee S, Sahu S, Chowdhury S, Mishra M, Talegaonkar S, Siddiqui L, Mishra H. Wood-Based Cellulose Nanofibrils: Haemocompatibility and Impact on the Development and Behaviour of Drosophila melanogaster. Biomolecules 2019; 9:biom9080363. [PMID: 31412664 PMCID: PMC6722666 DOI: 10.3390/biom9080363] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Wood-based cellulose nanofibrils (CNF) offer an excellent scaffold for drug-delivery formulation development. However, toxicity and haemocompatibility of the drug carrier is always an important issue. In this study, toxicity-related issues of CNF were addressed. Different doses of CNF were orally administered to Drosophila and different tests like the developmental cycle, trypan blue exclusion assay, larva crawling assay, thermal sensitivity assay, cold sensitivity assay, larval light preference test, climbing behaviour, nitroblue tetrazolium (NBT) reduction assay, adult phenotype, and adult weight were conducted to observe the impact on its development and behaviour. A haemocompatibility assay was done on the blood taken from healthy Wistar rats. In Drosophila, the abnormalities in larval development and behaviour were observed in the behavioural assays. However, the cytotoxic effect could not be confirmed by the gut staining and level of reactive oxygen species. The larvae developed into an adult without any abnormality in the phenotype. The CNF did cause loss of weight in the adult flies and did not cause much toxicity within the body since there was no phenotypic defect. Hemolysis data also suggested that CNF was safe at lower doses, as the data was well within acceptable limits. All these results suggest that cellulose nanofibres have no significant cytotoxic effects on Drosophila. However, the developmental and behavioural abnormalities suggest that CNF may act as a behavioural teratogen.
Collapse
Affiliation(s)
- Pawan Kumar Mishra
- Department of Wood Processing Technology, Mendel University in Brno, 61300 Brno, Czech Republic.
| | - Adam Ekielski
- Department of Production Management and Engineering, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Sumit Mukherjee
- Neural Developmental Biology Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha 76908, India
| | - Swetapadma Sahu
- Neural Developmental Biology Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha 76908, India
| | - Saptarshi Chowdhury
- Biotechnology Department, Heritage Institute of Technology, Kolkata 700107, West Bengal, India
| | - Monalisa Mishra
- Neural Developmental Biology Lab, Department of Life Science, National Institute of Technology, Rourkela, Odisha 76908, India
| | - Sushama Talegaonkar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Lubna Siddiqui
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Govt. of NCT of Delhi 110017, New Delhi, India
| | - Harshita Mishra
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Govt. of NCT of Delhi 110017, New Delhi, India
| |
Collapse
|
40
|
Nässel DR, Zandawala M. Recent advances in neuropeptide signaling in Drosophila, from genes to physiology and behavior. Prog Neurobiol 2019; 179:101607. [PMID: 30905728 DOI: 10.1016/j.pneurobio.2019.02.003] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
This review focuses on neuropeptides and peptide hormones, the largest and most diverse class of neuroactive substances, known in Drosophila and other animals to play roles in almost all aspects of daily life, as w;1;ell as in developmental processes. We provide an update on novel neuropeptides and receptors identified in the last decade, and highlight progress in analysis of neuropeptide signaling in Drosophila. Especially exciting is the huge amount of work published on novel functions of neuropeptides and peptide hormones in Drosophila, largely due to the rapid developments of powerful genetic methods, imaging techniques and innovative assays. We critically discuss the roles of peptides in olfaction, taste, foraging, feeding, clock function/sleep, aggression, mating/reproduction, learning and other behaviors, as well as in regulation of development, growth, metabolic and water homeostasis, stress responses, fecundity, and lifespan. We furthermore provide novel information on neuropeptide distribution and organization of peptidergic systems, as well as the phylogenetic relations between Drosophila neuropeptides and those of other phyla, including mammals. As will be shown, neuropeptide signaling is phylogenetically ancient, and not only are the structures of the peptides, precursors and receptors conserved over evolution, but also many functions of neuropeptide signaling in physiology and behavior.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden.
| | - Meet Zandawala
- Department of Zoology, Stockholm University, Stockholm, Sweden; Department of Neuroscience, Brown University, Providence, RI, USA.
| |
Collapse
|
41
|
Gong C, Ouyang Z, Zhao W, Wang J, Li K, Zhou P, Zhao T, Zheng N, Gong Z. A Neuronal Pathway that Commands Deceleration in Drosophila Larval Light-Avoidance. Neurosci Bull 2019; 35:959-968. [PMID: 30810958 DOI: 10.1007/s12264-019-00349-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 12/26/2018] [Indexed: 01/09/2023] Open
Abstract
When facing a sudden danger or aversive condition while engaged in on-going forward motion, animals transiently slow down and make a turn to escape. The neural mechanisms underlying stimulation-induced deceleration in avoidance behavior are largely unknown. Here, we report that in Drosophila larvae, light-induced deceleration was commanded by a continuous neural pathway that included prothoracicotropic hormone neurons, eclosion hormone neurons, and tyrosine decarboxylase 2 motor neurons (the PET pathway). Inhibiting neurons in the PET pathway led to defects in light-avoidance due to insufficient deceleration and head casting. On the other hand, activation of PET pathway neurons specifically caused immediate deceleration in larval locomotion. Our findings reveal a neural substrate for the emergent deceleration response and provide a new understanding of the relationship between behavioral modules in animal avoidance responses.
Collapse
Affiliation(s)
- Caixia Gong
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhenhuan Ouyang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, 310007, China
| | - Weiqiao Zhao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jie Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Kun Li
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Peipei Zhou
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ting Zhao
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 22011, USA
| | - Nenggan Zheng
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, 310007, China.
| | - Zhefeng Gong
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
42
|
Deveci D, Martin FA, Leopold P, Romero NM. AstA Signaling Functions as an Evolutionary Conserved Mechanism Timing Juvenile to Adult Transition. Curr Biol 2019; 29:813-822.e4. [PMID: 30799245 DOI: 10.1016/j.cub.2019.01.053] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/28/2018] [Accepted: 01/21/2019] [Indexed: 01/07/2023]
Abstract
The onset of sexual maturation is the result of a hormonal cascade peaking with the production of steroid hormones. In animals undergoing a program of determinate growth, sexual maturation also coincides with the attainment of adult size. The exact signals that time the onset of maturation and the mechanisms coupling growth and maturation remain elusive. Here, we show that the Drosophila neuropeptide AstA and its receptor AstAR1 act as a brain trigger for maturation and juvenile growth. We first identified AstAR1 in an RNAi-based genetic screen as a key regulator of sexual maturation. Its specific knockdown in prothoracicotropic hormone (PTTH)-producing neurons delays the onset of maturation by impairing PTTH secretion. In addition to its role in PTTH neurons, AstAR1 is required in the brain insulin-producing cells (IPCs) to promote insulin secretion and systemic growth. AstAR1 function is mediated by the AstA neuropeptide that is expressed in two bilateral neurons contacting the PTTH neurons and the IPCs. Silencing brain AstA expression delays the onset of maturation, therefore extending the growth period. However, no pupal overgrowth is observed, indicating that, in these conditions, the growth-promoting function of AstAR1 is also impaired. These data suggest that AstA/AstAR1 acts to coordinate juvenile growth with maturation. Interesting, AstA/AstAR1 is homologous to KISS/GPR54, a ligand-receptor signal required for human puberty, suggesting that an evolutionary conserved neural circuitry controls the onset of maturation.
Collapse
Affiliation(s)
- Derya Deveci
- University Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | | | - Pierre Leopold
- Institut Curie, PSL Research University, CNRS UMR3215, Inserm U934, UPMC Paris-Sorbonne, 26 Rue d'Ulm, 75005 Paris, France.
| | - Nuria M Romero
- University Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France.
| |
Collapse
|
43
|
Hormonal signaling cascades required for phototaxis switch in wandering Leptinotarsa decemlineata larvae. PLoS Genet 2019; 15:e1007423. [PMID: 30615614 PMCID: PMC6336328 DOI: 10.1371/journal.pgen.1007423] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 01/17/2019] [Accepted: 11/27/2018] [Indexed: 02/08/2023] Open
Abstract
Many animals exploit several niches sequentially during their life cycles, a fitness referred to as ontogenetic niche shift (ONS). To successfully accomplish ONS, transition between development stages is often coupled with changes in one or more primitive, instinctive behaviors. Yet, the underlining molecular mechanisms remain elusive. We show here that Leptinotarsa decemlineata larvae finish their ONS at the wandering stage by leaving the plant and pupating in soil. At middle wandering phase, larvae also switch their phototactic behavior, from photophilic at foraging period to photophobic. We find that enhancement of juvenile hormone (JH) signal delays the phototactic switch, and vise verse. Moreover, RNA interference (RNAi)-aided knockdown of LdPTTH (prothoracicotropic hormone gene) or LdTorso (PTTH receptor gene) impairs avoidance response to light, a phenotype nonrescuable by 20-hydroxyecdysone. Consequently, the RNAi beetles pupate at the soil surface or in shallow layer of soil, with most of them failing to construct pupation chambers. Furthermore, a combination of depletion of LdPTTH/LdTorso and disturbance of JH signal causes no additive effects on light avoidance response and pupation site selection. Finally, we establish that TrpA1 (transient receptor potential (TRP) cation channel) is necessary for light avoidance behavior, acting downstream of PTTH. We conclude that JH/PTTH cascade concomitantly regulates metamorphosis and the phototaxis switch, to drive ONS of the wandering beetles from plant into soil to start the immobile pupal stage. Many animals occupy distinct niches and utilize diverse resources at different development stages in order to meet stage-dependent requirements and overcome stage-specific limitations. This fitness is referred to as ontogenetic niche shift (ONS). During the preparation for ONS, animals often change one or more primitive, instinctive behaviors. Holometabolous insects, with four discrete developmental periods usually in different niches, are a suitable animal group to explore the molecular modes of these behavioral switches. Here we find that Leptinotarsa decemlineata larvae, an insect defoliator of potatoes, switch their phototactic behavior, from photophilic at feeding period to photophobic during the larval-pupal transition (wandering stage). This phototactic switch facilitates the wandering larvae to accomplish the ONS from potato plants to their pupation sites below ground. We show that JH/PTTH cascade controls the phototaxis switch, through a step in photo transduction between the photoreceptor molecule and the transient receptor potential cation channel.
Collapse
|
44
|
Humberg TH, Sprecher SG. Two Pairs of Drosophila Central Brain Neurons Mediate Larval Navigational Strategies Based on Temporal Light Information Processing. Front Behav Neurosci 2018; 12:305. [PMID: 30568583 PMCID: PMC6289971 DOI: 10.3389/fnbeh.2018.00305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/26/2018] [Indexed: 11/21/2022] Open
Abstract
Some animals are attracted by sun light, others are highly repulsed by it. Especially for slowly moving animals, such as Drosophila larvae, direct sunlight may be perceived as noxious stimulus as it increases the risk of desiccation, DNA-damaging by UV-light and exposure to predators. For several reasons, model organisms like Drosophila larvae are well-suited for investigating how light cues are translated into an appropriate behavioral output. First, many of the genetic tools, which were created for use in adult fruit flies, work also in larvae. Second, the lower number of cells in Drosophila larvae compared to adults makes this system adequate for reconstructing neural circuits. Third, the relatively simple behavioral repertoire of larvae facilitates the study of basic functions like navigation with regards to light. Larvae navigate robustly away from a light source by the use of several sophisticated behavioral strategies which are based on temporal or spatial information processing. Two central brain neurons, the NP394-neurons, are highly important for larval light avoidance. It was even reported that these cells seem to play a functional role in a putative larval light preference switch right before pupation. However, the exact function of the NP394-neurons in light navigation remains unknown. We here show that the functional role of NP394-neurons in larval light navigation is restricted to behaviors based on temporal information processing, but not for spatial navigation.
Collapse
Affiliation(s)
| | - Simon G. Sprecher
- Department of Biology, Institute of Zoology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
45
|
Mineo A, Furriols M, Casanova J. The trigger (and the restriction) of Torso RTK activation. Open Biol 2018; 8:180180. [PMID: 30977718 PMCID: PMC6303783 DOI: 10.1098/rsob.180180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/08/2018] [Indexed: 01/09/2023] Open
Abstract
The Torso pathway is an ideal model of receptor tyrosine kinase systems, in particular when addressing questions such as how receptor activity is turned on and, equally important, how it is restricted, how different outcomes can be generated from a single signal, and the extent to which gene regulation by signalling pathways relies on the relief of transcriptional repression. In this regard, we considered it pertinent to single out the fundamental notions learned from the Torso pathway beyond the specificities of this system (Furriols and Casanova 2003 EMBO J. 22, 1947-1952. ( doi:10.1093/emboj/cdg224 )). Since then, the Torso system has gained relevance and its implications beyond its original involvement in morphogenesis and into many disciplines such as oncogenesis, hormone control and neurobiology are now acknowledged. Thus, we believe that it is timely to highlight additional notions supported by new findings and to draw attention to future prospects. Given the late development of research in the field, we wish to devote this review to the events leading to the activation of the Torso receptor, the main focus of our most recent work.
Collapse
Affiliation(s)
- Alessandro Mineo
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
- Institut de Recerca Biomèdica de Barcelona, (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| | - Marc Furriols
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
- Institut de Recerca Biomèdica de Barcelona, (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
- Institut de Recerca Biomèdica de Barcelona, (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), C/Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
46
|
Hyun S. Body size regulation by maturation steroid hormones: a Drosophila perspective. Front Zool 2018; 15:44. [PMID: 30479644 PMCID: PMC6247710 DOI: 10.1186/s12983-018-0290-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/01/2018] [Indexed: 12/19/2022] Open
Abstract
The mechanism that determines the specific body size of an animal is a fundamental biological question that remains largely unanswered. This aspect is now beginning to be understood in insect models, particularly in Drosophila melanogaster, with studies highlighting the importance of nutrient-responsive growth signaling pathways involving insulin/insulin-like growth factor signaling (IIS) and target of rapamycin (TOR) (IIS/TOR). These pathways operate in animals, from insects to mammals, adjusting the growth rate in response to the nutritional condition of the organism. Organismal growth is closely coupled with the process of developmental maturation mediated by maturation steroid hormones, which is influenced greatly by environmental and nutritional conditions. Recent Drosophila studies have been revealing the mechanisms responsible for this phenomenon. In this review, I summarize some important findings about the steroid hormone regulation of Drosophila body growth, calling attention to the influence of developmental nutritional conditions on animal size determination.
Collapse
Affiliation(s)
- Seogang Hyun
- Department of Life Science, Chung-Ang University, Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea
| |
Collapse
|
47
|
Wang L, Liu X, Liu Z, Wang X, Lei C, Zhu F. Members of the neuropeptide transcriptional network in Helicoverpa armigera and their expression in response to light stress. Gene 2018; 671:67-77. [PMID: 29787823 DOI: 10.1016/j.gene.2018.05.070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/11/2018] [Accepted: 05/17/2018] [Indexed: 12/14/2022]
Abstract
Neuropeptides and peptide hormones play central roles in the regulation of various types of insect physiology and behavior. Artificial light at night, a form of environmental stress, has recently been regarded as a source of light stress on nocturnal insects. Because related genomic information is not available, molecular biological studies on the response of neuropeptides in nocturnal insects to light stress are limited. Based on the de novo sequencing of the Helicoverpa armigera head transcriptome, we obtained 124,960 unigenes. Of these, the number of unigenes annotated as neuropeptides and peptide hormones, neurotransmitter precursor processing enzymes, and neurotransmitter receptors were 34, 17, and 58, respectively. Under light stress, there were sex-specific differences in gene expression measured by qRT-PCR. The IMFamide, leucokinin and sNPF genes were differentially expressed at the mRNA level in males but not in females in response to light stress. The results provide new insights on the diversity of the neuropeptide transcriptional network of H. armigera. In addition, some neuropeptides exhibited sex-specific differential expression in response to light stress. Taken collectively, these results not only expand the catalog of known insect neuropeptides but also provide a framework for future functional studies on the physiological roles they play in the light stress response behavior of nocturnal moths.
Collapse
Affiliation(s)
- Lijun Wang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinhui Liu
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| | - Zhengxing Liu
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xiaoping Wang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| | - Chaoliang Lei
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| | - Fen Zhu
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
48
|
Shimell M, Pan X, Martin FA, Ghosh AC, Leopold P, O'Connor MB, Romero NM. Prothoracicotropic hormone modulates environmental adaptive plasticity through the control of developmental timing. Development 2018; 145:dev.159699. [PMID: 29467242 DOI: 10.1242/dev.159699] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 02/12/2018] [Indexed: 12/19/2022]
Abstract
Adult size and fitness are controlled by a combination of genetics and environmental cues. In Drosophila, growth is confined to the larval phase and final body size is impacted by the duration of this phase, which is under neuroendocrine control. The neuropeptide prothoracicotropic hormone (PTTH) has been proposed to play a central role in controlling the length of the larval phase through regulation of ecdysone production, a steroid hormone that initiates larval molting and metamorphosis. Here, we test this by examining the consequences of null mutations in the Ptth gene for Drosophila development. Loss of Ptth causes several developmental defects, including a delay in developmental timing, increase in critical weight, loss of coordination between body and imaginal disc growth, and reduced adult survival in suboptimal environmental conditions such as nutritional deprivation or high population density. These defects are caused by a decrease in ecdysone production associated with altered transcription of ecdysone biosynthetic genes. Therefore, the PTTH signal contributes to coordination between environmental cues and the developmental program to ensure individual fitness and survival.
Collapse
Affiliation(s)
- MaryJane Shimell
- Department of Genetics Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Xueyang Pan
- Department of Genetics Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Francisco A Martin
- University Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France.,Cajal Institute, Av Doctor Arce 37, 28002 Madrid, Spain
| | - Arpan C Ghosh
- Department of Genetics Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Pierre Leopold
- University Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| | - Michael B O'Connor
- Department of Genetics Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nuria M Romero
- University Côte d'Azur, CNRS, Inserm, Institute of Biology Valrose, Parc Valrose, 06108 Nice, France
| |
Collapse
|
49
|
Abstract
Unlike adult Drosophila ovaries, pupal ovaries are relatively difficult to access and examine due to their small size, translucence, and encasing within a pupal case. The challenge of dissecting pupal ovaries also lies in their physical location within the pupa: the ovaries are surrounded by fat body cells inside the pupal abdomen, and these fat cells must be removed to allow for proper antibody staining. To overcome these challenges, this protocol utilizes customized Pasteur pipets to extract fat body cells from the pupal abdomen. Moreover, a chambered coverglass is used in place of a microcentrifuge tube during the staining process to improve visibility of the pupae. However, despite these and other advantages of the tools used in this protocol, successful execution of these techniques may still involve several days of practice due to the small size of pupal ovaries. The techniques outlined in this protocol could be applied to time course experiments in which ovaries are analyzed at various stages of pupal development.
Collapse
Affiliation(s)
| | - Dorothea Godt
- Department of Cell and Systems Biology, University of Toronto
| | | |
Collapse
|
50
|
Jarabo P, Martin FA. Neurogenetics of Drosophila circadian clock: expect the unexpected. J Neurogenet 2017; 31:250-265. [DOI: 10.1080/01677063.2017.1370466] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|