1
|
Lu JM, Shang F, Ding BY, Wang L, Li QC, Wang JJ, Dou W. Characterization of two Bursicon genes and their association with wing development in the brown citrus aphid, Aphis citricidus. INSECT SCIENCE 2024; 31:1684-1696. [PMID: 38339808 DOI: 10.1111/1744-7917.13337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
The tanning hormone, Bursicon, is a neuropeptide secreted by the insect nervous system that functions as a heterodimer composed of Burs-α and Burs-β subunits. It plays a critical role in the processes of cuticle tanning and wing expansion in insects. In this study, we successfully identified the AcBurs-α and AcBurs-β genes in Aphis citricidus. The open reading frames of AcBurs-α and AcBurs-β were 480 and 417 bp in length, respectively. Both AcBurs-α and AcBurs-β exhibited 11 conserved cysteine residues. AcBurs-α and AcBurs-β were expressed during all developmental stages of A. citricidus and showed high expression levels in the winged aphids. To investigate the potential role of AcBurs-α and AcBurs-β in wing development, we employed RNA interference (RNAi) techniques. With the efficient silencing of AcBurs-α (44.90%) and AcBurs-β (52.31%), malformed wings were induced in aphids. The proportions of malformed wings were 22.50%, 25.84%, and 38.34% in dsAcBurs-α-, dsAcBur-β-, and dsAcBurs-α + dsAcBur-β-treated groups, respectively. Moreover, feeding protein kinase A inhibitors (H-89) also increased the proportion of malformed wings to 30.00%. Feeding both double-stranded RNA and inhibitors (H-89) significantly downregulated the wing development-related genes nubbin, vestigial, notch and spalt major. Silence of vestigial through RNAi also led to malformed wings. Meanwhile, the exogenous application of 3 hormones that influence wing development did not affect the expression level of AcBursicon genes. These findings indicate that AcBursicon genes plays a crucial role in wing development in A. citricidus; therefore, it represents a potential molecular target for the control of this pest through RNAi-based approaches.
Collapse
Affiliation(s)
- Jin-Ming Lu
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
- Academy of Agricultural Sciences, Southwest University, Chongqing, China
| | - Feng Shang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
- Academy of Agricultural Sciences, Southwest University, Chongqing, China
| | - Bi-Yue Ding
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
- Academy of Agricultural Sciences, Southwest University, Chongqing, China
| | - Lin Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
- Academy of Agricultural Sciences, Southwest University, Chongqing, China
| | - Qing-Chun Li
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
- Academy of Agricultural Sciences, Southwest University, Chongqing, China
| | - Jin-Jun Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
- Academy of Agricultural Sciences, Southwest University, Chongqing, China
| | - Wei Dou
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing, China
- Academy of Agricultural Sciences, Southwest University, Chongqing, China
| |
Collapse
|
2
|
Zhang Z, Li J, Wang Y, Li Z, Liu X, Zhang S. Neuropeptide Bursicon and its receptor-mediated the transition from summer-form to winter-form of Cacopsylla chinensis. eLife 2024; 13:RP97298. [PMID: 39514284 PMCID: PMC11548876 DOI: 10.7554/elife.97298] [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: 11/16/2024] Open
Abstract
Seasonal polyphenism enables organisms to adapt to environmental challenges by increasing phenotypic diversity. Cacopsylla chinensis exhibits remarkable seasonal polyphenism, specifically in the form of summer-form and winter-form, which have distinct morphological phenotypes. Previous research has shown that low temperature and the temperature receptor CcTRPM regulate the transition from summer-form to winter-form in C. chinensis by impacting cuticle content and thickness. However, the underling neuroendocrine regulatory mechanism remains largely unknown. Bursicon, also known as the tanning hormone, is responsible for the hardening and darkening of the insect cuticle. In this study, we report for the first time on the novel function of Bursicon and its receptor in the transition from summer-form to winter-form in C. chinensis. Firstly, we identified CcBurs-α and CcBurs-β as two typical subunits of Bursicon in C. chinensis, which were regulated by low temperature (10 °C) and CcTRPM. Subsequently, CcBurs-α and CcBurs-β formed a heterodimer that mediated the transition from summer-form to winter-form by influencing the cuticle chitin contents and cuticle thickness. Furthermore, we demonstrated that CcBurs-R acts as the Bursicon receptor and plays a critical role in the up-stream signaling of the chitin biosynthesis pathway, regulating the transition from summer-form to winter-form. Finally, we discovered that miR-6012 directly targets CcBurs-R, contributing to the regulation of Bursicon signaling in the seasonal polyphenism of C. chinensis. In summary, these findings reveal the novel function of the neuroendocrine regulatory mechanism underlying seasonal polyphenism and provide critical insights into the insect Bursicon and its receptor.
Collapse
Affiliation(s)
- Zhixian Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural UniversityBeijingChina
| | - Jianying Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural UniversityBeijingChina
| | - Yilin Wang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural UniversityBeijingChina
| | - Zhen Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural UniversityBeijingChina
| | - Xiaoxia Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural UniversityBeijingChina
| | - Songdou Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural UniversityBeijingChina
- Sanya Institute of China Agricultural UniversitySanya CityChina
| |
Collapse
|
3
|
Sardina F, Carsetti C, Giorgini L, Fattorini G, Cestra G, Rinaldo C. Cul-4 inhibition rescues spastin levels and reduces defects in hereditary spastic paraplegia models. Brain 2024; 147:3534-3546. [PMID: 38551087 PMCID: PMC11449140 DOI: 10.1093/brain/awae095] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 10/05/2024] Open
Abstract
Hereditary spastic paraplegias (HSPs) are degenerative motor neuron diseases characterized by progressive spasticity and weakness in the lower limbs. The most common form of HSP is due to SPG4 gene haploinsufficiency. SPG4 encodes the microtubule severing enzyme spastin. Although, there is no cure for SPG4-HSP, strategies to induce a spastin recovery are emerging as promising therapeutic approaches. Spastin protein levels are regulated by poly-ubiquitination and proteasomal-mediated degradation, in a neddylation-dependent manner. However, the molecular players involved in this regulation are unknown. Here, we show that the Cullin-4-RING E3 ubiquitin ligase complex (CRL4) regulates spastin stability. Inhibition of CRL4 increases spastin levels by preventing its poly-ubiquitination and subsequent degradation in spastin-proficient and in patient derived SPG4 haploinsufficient cells. To evaluate the role of CRL4 complex in spastin regulation in vivo, we developed a Drosophila melanogaster model of SPG4 haploinsufficiency which show alterations of synapse morphology and locomotor activity, recapitulating phenotypical defects observed in patients. Downregulation of the CRL4 complex, highly conserved in Drosophila, rescues spastin levels and the phenotypical defects observed in flies. As a proof of concept of possible pharmacological treatments, we demonstrate a recovery of spastin levels and amelioration of the SPG4-HSP-associated defects both in the fly model and in patient-derived cells by chemical inactivation of the CRL4 complex with NSC1892. Taken together, these findings show that CRL4 contributes to spastin stability regulation and that it is possible to induce spastin recovery and rescue of SPG4-HSP defects by blocking the CRL4-mediated spastin degradation.
Collapse
Affiliation(s)
- Francesca Sardina
- National Research Council (CNR), Institute of Molecular Biology and Pathology (IBPM), c/o University of Rome Sapienza, 00185 Rome, Italy
| | - Claudia Carsetti
- National Research Council (CNR), Institute of Molecular Biology and Pathology (IBPM), c/o University of Rome Sapienza, 00185 Rome, Italy
- Department of Biology and Biotechnology, University of Rome Sapienza, 00185, Rome, Italy
| | - Ludovica Giorgini
- National Research Council (CNR), Institute of Molecular Biology and Pathology (IBPM), c/o University of Rome Sapienza, 00185 Rome, Italy
| | - Gaia Fattorini
- National Research Council (CNR), Institute of Molecular Biology and Pathology (IBPM), c/o University of Rome Sapienza, 00185 Rome, Italy
- Department of Biology and Biotechnology, University of Rome Sapienza, 00185, Rome, Italy
| | - Gianluca Cestra
- National Research Council (CNR), Institute of Molecular Biology and Pathology (IBPM), c/o University of Rome Sapienza, 00185 Rome, Italy
- Department of Biology and Biotechnology, University of Rome Sapienza, 00185, Rome, Italy
- Fondazione Santa Lucia IRCCS, c/o CERC, 00179, Rome, Italy
| | - Cinzia Rinaldo
- National Research Council (CNR), Institute of Molecular Biology and Pathology (IBPM), c/o University of Rome Sapienza, 00185 Rome, Italy
| |
Collapse
|
4
|
Liao LL, Li WZ, Jin L, Li GQ. Rnai-based functional analysis of bursicon genes related to cuticle pigmentation in a ladybird beetle. JOURNAL OF INSECT PHYSIOLOGY 2024; 158:104696. [PMID: 39173874 DOI: 10.1016/j.jinsphys.2024.104696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/01/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
In arthropods, the binding of a bursicon (encoded by burs and pburs) heterodimer or homodimer to a leucine-rich repeat-containing G protein coupled receptor LGR2 (encoded by rk) can activate many physiological processes, especially cuticle pigmentation during insect ecdysis. In the current paper, we intended to ascertain whether bursicon signaling mediates body coloration in the 28-spotted larger potato ladybird, Henosepilachna vigintioctomaculata, and if so, by which way bursicon signal governs the pigmentation. The high expression of Hvburs, Hvpburs and Hvrk occurred in the young larvae, pupae and adults, especially in the head and ventral nerve cord. RNA interference (RNAi) aided knockdown of Hvburs, Hvpburs or Hvrk in the prepupae caused similar phenotypic defects. The pigmentation of the resultant adults was affected, with significantly reduced dark areas on the sternums. Moreover, the accumulated mRNA levels of two sclerotin biosynthesis genes, aspartate 1-decarboxylase gene Hvadc and N-β-alanyldopamine synthase gene Hvebony, were significantly increased in the Hvburs, Hvpburs or Hvrk RNAi beetles. Furthermore, depletion of either Hvadc or Hvebony could completely rescue the impaired coloration on the sternums of Hvpburs RNAi adult. Our results supported that bursicon heterodimer-mediated signal regulate cuticle pigmentation. The bursicon signaling may tune the ratio of melanins (dark/black, brown) to sclerotins (light yellow, colorless) exerting its regulative role in the pigmentation of H. vigintioctomaculata sternums.
Collapse
Affiliation(s)
- Lan-Lan Liao
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Wen-Ze Li
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Lin Jin
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Guo-Qing Li
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
5
|
Wu G, Ma T, Hancock CE, Gonzalez S, Aryal B, Vaz S, Chan G, Palarca-Wong M, Allen N, Chung CI, Shu X, Liu Q. Opposing GPCR signaling programs protein intake setpoint in Drosophila. Cell 2024; 187:5376-5392.e17. [PMID: 39197448 PMCID: PMC11437785 DOI: 10.1016/j.cell.2024.07.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 05/31/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024]
Abstract
Animals defend a target level for their fundamental needs, including food, water, and sleep. Deviation from the target range, or "setpoint," triggers motivated behaviors to eliminate that difference. Whether and how the setpoint itself is encoded remains enigmatic for all motivated behaviors. Employing a high-throughput feeding assay in Drosophila, we demonstrate that the protein intake setpoint is set to different values in male, virgin female, and mated female flies to meet their varying protein demands. Leveraging this setpoint variability, we found, remarkably, that the information on the intake setpoint is stored within the protein hunger neurons as the resting membrane potential. Two RFamide G protein-coupled receptor (GPCR) pathways, by tuning the resting membrane potential in opposite directions, coordinately program and adjust the protein intake setpoint. Together, our studies map the protein intake setpoint to a single trackable physiological parameter and elucidate the cellular and molecular mechanisms underlying setpoint determination and modulation.
Collapse
Affiliation(s)
- Guangyan Wu
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tianji Ma
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Clare E Hancock
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Santiago Gonzalez
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Binod Aryal
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sharon Vaz
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Gabrielle Chan
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Madison Palarca-Wong
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nick Allen
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chan-I Chung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Xiaokun Shu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Qili Liu
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
6
|
Li WZ, Wu YK, Zhang YX, Jin L, Li GQ. Behavioral events and functional analysis of bursicon signal during adult eclosion in the 28-spotted larger potato ladybird. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 203:106011. [PMID: 39084776 DOI: 10.1016/j.pestbp.2024.106011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
To accommodate growth, insects must periodically shed their exoskeletons. In Manduca sexta, Drosophila melanogaster and Tribolium castaneum, Bursicon (Burs)/ Partner of bursicon (Pburs)-LGR2 signal is an indispensable component for the proper execution of ecdysis behavior during adult eclosion. Nevertheless, the behavioral events and the roles of bursicon signaling in other insects deserve further exploration. In the current paper, we found that the pupal-adult ecdysis in Henosepilachna vigintioctomaculata could be divided into three distinct stages, preecdysis, ecdysis and postecdysis. Preecdysis behavioral sequences included abdomen twitches, dorsal-ventral contractions and air filling that function to loosen the old cuticle. Ecdysis events began with anterior-posterior contractions that gradually split the old integument along the dorsal body midline, followed by freeing of legs and mouthparts, and culminated in detachment from pupal cuticle. Postecdysis behavioral processes contained three actions: perch selection and stretching of elytra and hindwings. RNA interference for HvBurs, HvPburs or Hvrk (encoding LGR2) strongly impaired wing expansion actions, and slightly influenced preecdysis and ecdysis behaviors. The RNAi beetles failed to extend their elytra and hindwings. In addition, injected with dsrk also caused kinked femurs and tibia. Our findings establish that bursicon pathway is involved in regulation of adult eclosion behavior, especially wing expansion motor programs. Given that wings facilitate food foraging, courtship, predator avoidance, dispersal and migration, our results provide a potential target for controlling H. vigintioctomaculata.
Collapse
Affiliation(s)
- Wen-Ze Li
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yi-Kuan Wu
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yu-Xing Zhang
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Lin Jin
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Guo-Qing Li
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
7
|
Nguyen TH, Vicidomini R, Choudhury SD, Han TH, Maric D, Brody T, Serpe M. scRNA-seq data from the larval Drosophila ventral cord provides a resource for studying motor systems function and development. Dev Cell 2024; 59:1210-1230.e9. [PMID: 38569548 PMCID: PMC11078614 DOI: 10.1016/j.devcel.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/05/2023] [Accepted: 03/06/2024] [Indexed: 04/05/2024]
Abstract
The Drosophila larval ventral nerve cord (VNC) shares many similarities with the spinal cord of vertebrates and has emerged as a major model for understanding the development and function of motor systems. Here, we use high-quality scRNA-seq, validated by anatomical identification, to create a comprehensive census of larval VNC cell types. We show that the neural lineages that comprise the adult VNC are already defined, but quiescent, at the larval stage. Using fluorescence-activated cell sorting (FACS)-enriched populations, we separate all motor neuron bundles and link individual neuron clusters to morphologically characterized known subtypes. We discovered a glutamate receptor subunit required for basal neurotransmission and homeostasis at the larval neuromuscular junction. We describe larval glia and endorse the general view that glia perform consistent activities throughout development. This census represents an extensive resource and a powerful platform for future discoveries of cellular and molecular mechanisms in repair, regeneration, plasticity, homeostasis, and behavioral coordination.
Collapse
Affiliation(s)
| | | | | | | | - Dragan Maric
- Flow and Imaging Cytometry Core, NINDS, NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
8
|
Dermady APC, DeFazio DL, Hensley EM, Ruiz DL, Chavez AD, Iannone SA, Dermady NM, Grandel LV, Hill AS. Neuronal excitability modulates developmental time of Drosophila melanogaster. Dev Biol 2024; 508:38-45. [PMID: 38224932 DOI: 10.1016/j.ydbio.2024.01.006] [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: 09/11/2022] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
Developmental time is a fundamental life history trait that affects the reproductive success of animals. Developmental time is known to be regulated by many genes and environmental conditions, yet mechanistic understandings of how various cellular processes influence the developmental timing of an organism are lacking. The nervous system is known to control key processes that affect developmental time, including the release of hormones that signal transitions between developmental stages. Here we show that the excitability of neurons plays a crucial role in modulating developmental time. Genetic manipulation of neuronal excitability in Drosophila melanogaster alters developmental time, which is faster in animals with increased neuronal excitability. We find that selectively modulating the excitability of peptidergic neurons is sufficient to alter developmental time, suggesting the intriguing hypothesis that the impact of neuronal excitability on DT may be at least partially mediated by peptidergic regulation of hormone release. This effect of neuronal excitability on developmental time is seen during embryogenesis and later developmental stages. Observed phenotypic plasticity in the effect of genetically increasing neuronal excitability at different temperatures, a condition also known to modulate excitability, suggests there is an optimal level of neuronal excitability, in terms of shortening DT. Together, our data highlight a novel connection between neuronal excitability and developmental time, with broad implications related to organismal physiology and evolution.
Collapse
Affiliation(s)
- Aidan P C Dermady
- College of the Holy Cross, Department of Biology, Worcester, MA, USA
| | - Dionna L DeFazio
- College of the Holy Cross, Department of Biology, Worcester, MA, USA
| | - Emily M Hensley
- College of the Holy Cross, Department of Biology, Worcester, MA, USA
| | - Daniel L Ruiz
- College of the Holy Cross, Department of Biology, Worcester, MA, USA
| | | | - Sarah A Iannone
- College of the Holy Cross, Department of Biology, Worcester, MA, USA
| | - Niall M Dermady
- College of the Holy Cross, Department of Biology, Worcester, MA, USA
| | - Lexis V Grandel
- College of the Holy Cross, Department of Biology, Worcester, MA, USA
| | - Alexis S Hill
- College of the Holy Cross, Department of Biology, Worcester, MA, USA.
| |
Collapse
|
9
|
Zhou ZX, Dou W, Wang M, Shang F, Wang JJ. Bursicon regulates wing expansion via PKA in the oriental fruit fly, Bactrocera dorsalis. PEST MANAGEMENT SCIENCE 2024; 80:388-396. [PMID: 37708392 DOI: 10.1002/ps.7768] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/06/2023] [Accepted: 09/15/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Bursicon is a heterodimeric neuropeptide that is involved in many physiological activities such as cuticle tanning, wing expansion, reproduction and immunity in insects. In this study, the role of bursicon in the wing expansion was investigated in Bactrocera dorsalis, an important invasive insect pest in agriculture. RESULTS The cDNA sequences and deduced amino acids of bursicon genes (named BdBurs-α and BdBurs-β) were determined, and two proteins typically contained 11 cysteine residues in conserved positions that were highly conserved in other insect species. The spatiotemporal expressions of bursicon genes showed that higher expression occurred at the pupal, early adult stage and ovaries, and lower expression at the late larval stage and in wing tissue (8-day-old pupae). Dysfunction of bursicon genes by dsRNA microinjection into 5-day-old pupae reduced PKA (a downstream component of the bursicon pathway) activity and resulted in malformed adult wings. PKA inhibitor injection into 5-day-old pupae also resulted in similar phenotypes. Hematoxylin & eosin staining of the adult wing showed that RNAi and PKA inhibitor treatment reduced the thickness of the wing cuticle, which wing cuticle thickness were ≈50% thinner than in the control. Furthermore, the expression of hedgehog (Bdhh) (one of 10 tested genes related to wing development) was significantly upregulated after RNAi and PKA inhibitor application. CONCLUSION The results indicate that bursicon plays a crucial role in the wing expansion of B. dorsalis, suggesting bursicon genes have potential to be the targets for B. dorsalis control. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhi-Xiong Zhou
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Science, Southwest University, Chongqing, China
| | - Wei Dou
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Science, Southwest University, Chongqing, China
| | - Mo Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
| | - Feng Shang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Science, Southwest University, Chongqing, China
| | - Jin-Jun Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Academy of Agricultural Science, Southwest University, Chongqing, China
| |
Collapse
|
10
|
Lim-Kian-Siang G, Izawa-Ishiguro AR, Rao Y. Neurexin-1-dependent circuit activity is required for the maintenance of photoreceptor subtype identity in Drosophila. Mol Brain 2024; 17:2. [PMID: 38167109 PMCID: PMC10759516 DOI: 10.1186/s13041-023-01073-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
In the human and Drosophila color vision system, each photoreceptor neuron (cone cell in humans and R7/R8 photoreceptor cell in Drosophila) makes a stochastic decision to express a single photopigment of the same family with the exclusion of the others. While recent studies have begun to reveal the mechanisms that specify the generation of cone subtypes during development in mammals, nothing is known about how the mosaic of mutually exclusive cone subtypes is maintained in the mammalian retina. In Drosophila, recent work has led to the identification of several intrinsic factors that maintain the identity of R8 photoreceptor subtypes in adults. Whether and how extrinsic mechanisms are involved, however, remain unknown. In this study, we present evidence that supports that the Drosophila transsynaptic adhesion molecule Neurexin 1 (Dnrx-1) is required non-cell autonomously in R8p subtypes for the maintenance of R8y subtype identity. Silencing the activity of R8p subtypes caused a phenotype identical to that in dnrx-1 mutants. These results support a novel role for Nrx-1-dependent circuit activity in mediating the communication between R8 photoreceptor subtypes for maintaining the subtype identity in the retina.
Collapse
Affiliation(s)
- Gabrielle Lim-Kian-Siang
- McGill Centre for Research in Neuroscience, Montreal, Canada
- Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Arianna R Izawa-Ishiguro
- McGill Centre for Research in Neuroscience, Montreal, Canada
- Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Yong Rao
- McGill Centre for Research in Neuroscience, Montreal, Canada.
- Department of Neurology and Neurosurgery, Montreal, Canada.
- Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
- Centre for Research in Neuroscience, McGill University Health Centre, Room L7-136, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
| |
Collapse
|
11
|
Guo H, Long GJ, Liu XZ, Ma YF, Zhang MQ, Gong LL, Dewer Y, Hull JJ, Wang MM, Wang Q, He M, He P. Functional characterization of tyrosine melanin genes in the white-backed planthopper and utilization of a spray-based nanoparticle-wrapped dsRNA technique for pest control. Int J Biol Macromol 2023; 230:123123. [PMID: 36603718 DOI: 10.1016/j.ijbiomac.2022.123123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023]
Abstract
As a significant pest of rice the white-backed planthopper (WBPH) Sogatella furcifera is a focus of pest management. However, traditional chemical-based control methods risk the development of pesticide resistance as well as severe ecological repercussions. Although nanoparticle-encapsulated dsRNAs provide a promising alternative method for sustainable pest management, gene targets specific to WBPH have yet to be optimized. Genes in the tyrosine-melanin pathway impact epidermal melanization and sclerotization, two processes essential for insect development and metabolism, have been proposed as good candidate targets for pest management. Seven genes (aaNAT, black, DDC, ebony, tan, TH, and yellow-y) in this group were identified from WBPH genome and functionally characterized by using RNAi for their impact on WBPH body color, development, and mortality. Knockdown of SfDDC, Sfblack, SfaaNAT, and Sftan caused cuticles to turn black, whereas Sfyellow-y and Sfebony knockdown resulted in yellow coloration. SfTH knockdown resulted in pale-colored bodies and high mortality. Additionally, an Escherichia coli expression system for large-scale dsRNA production was coupled with star polycation nanoparticles to develop a sprayable RNAi method targeting SfTH that induced high WBPH mortality rates on rice seedlings. These findings lay the groundwork for the development of large-scale dsRNA nanoparticle sprays as a WBPH control method.
Collapse
Affiliation(s)
- Huan Guo
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Gui-Jun Long
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Xuan-Zheng Liu
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Yun-Feng Ma
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Meng-Qi Zhang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Lang-Lang Gong
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Youssef Dewer
- Phytotoxicity Research Department, Central Agricultural Pesticide Laboratory, Agricultural Research Center, 7 Nadi El-Seid Street, Dokki, 12618 Giza, Egypt
| | - J Joe Hull
- Pest Management and Biocontrol Research Unit, US Arid Land Agricultural Research Center, USDA Agricultural Research Services, Maricopa, AZ, 85138, USA
| | - Mei-Mei Wang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Qin Wang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China
| | - Ming He
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China.
| | - Peng He
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang, 550025, PR China.
| |
Collapse
|
12
|
A novel post-developmental role of the Hox genes underlies normal adult behavior. Proc Natl Acad Sci U S A 2022; 119:e2209531119. [PMID: 36454751 PMCID: PMC9894213 DOI: 10.1073/pnas.2209531119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
The molecular mechanisms underlying the stability of mature neurons and neural circuits are poorly understood. Here we explore this problem and discover that the Hox genes are a component of the genetic program that maintains normal neural function in adult Drosophila. We show that post-developmental downregulation of the Hox gene Ultrabithorax (Ubx) in adult neurons leads to substantial anomalies in flight. Mapping the cellular basis of these effects reveals that Ubx is required within a subset of dopaminergic neurons, and cell circuitry analyses in combination with optogenetics allow us to link these dopaminergic neurons to flight control. Functional imaging experiments show that Ubx is necessary for normal dopaminergic activity, and neuron-specific RNA-sequencing defines two previously uncharacterized ion channel-encoding genes as potential mediators of Ubx behavioral roles. Our study thus reveals a novel role of the Hox system in controlling adult behavior and neural function. Based on the broad evolutionary conservation of the Hox system across distantly related animal phyla, we predict that the Hox genes might play neurophysiological roles in adult forms of other species, including humans.
Collapse
|
13
|
Ze LJ, Wang P, Peng YC, Jin L, Li GQ. Silencing tyrosine hydroxylase or dopa decarboxylase gene disrupts cuticle tanning during larva-pupa-adult transformation in Henosepilachna vigintioctopunctata. PEST MANAGEMENT SCIENCE 2022; 78:3880-3893. [PMID: 35470957 DOI: 10.1002/ps.6948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/30/2022] [Accepted: 04/26/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The 28-spotted potato ladybird, Henosepilachna vigintioctopunctata, is a notorious defoliator of many solanaceous and cucurbitaceous plants. Tyrosine hydroxylase (TH) and dopa decarboxylase (DDC) are responsible for cuticle tanning pathway in insects. RESULTS We identified HvTH and HvDDC in H. vigintioctopunctata, and found that high levels of them were accumulated just before or right after molting. Injection of dsHvTH or feeding 3-iodo-tyrosine (3-IT) at the third instar larval stage repressed tanning of the larval cuticle, reduced larval feeding, inhibited larval growth, and consequently caused 100% of larval mortality. Knockdown of HvDDC at the third instar larval stage hardly affected the coloration of larval head, and partially inhibited pigmentation of larval bodies and around 80% of the HvDDC RNAi larvae developed into albino pupae and adults. Moreover, depletion of HvTH or HvDDC at the fourth instar larval stage resulted in albino pupae and adults. The HvTH or HvDDC hypomorph adults fully or partially failed to remove the larval/pupal exuviae, possessed pale and abnormal wings, and poorly tanned heads and bodies, and eventually, struggled for several days without feeding on leaves before death. CONCLUSION These results show that TH and DDC play key roles in larval and adult cuticle tanning and development in H. vigintioctopunctata. Also, these findings suggest that dopa- and dopamine-originated pigments are essential for larval and adult feeding behavior and the molting process during emergence. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Long-Ji Ze
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Pei Wang
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Ying-Chuan Peng
- Institute of Entomology, Jiangxi Agricultural University, Nanchang, China
| | - Lin Jin
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Guo-Qing Li
- Key Laboratory of Integrated Management of Crop Disease and Pests, Ministry of Education / State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
14
|
Corrales M, Cocanougher BT, Kohn AB, Wittenbach JD, Long XS, Lemire A, Cardona A, Singer RH, Moroz LL, Zlatic M. A single-cell transcriptomic atlas of complete insect nervous systems across multiple life stages. Neural Dev 2022; 17:8. [PMID: 36002881 PMCID: PMC9404646 DOI: 10.1186/s13064-022-00164-6] [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: 03/02/2022] [Accepted: 07/10/2022] [Indexed: 12/15/2022] Open
Abstract
Molecular profiles of neurons influence neural development and function but bridging the gap between genes, circuits, and behavior has been very difficult. Here we used single cell RNAseq to generate a complete gene expression atlas of the Drosophila larval central nervous system composed of 131,077 single cells across three developmental stages (1 h, 24 h and 48 h after hatching). We identify 67 distinct cell clusters based on the patterns of gene expression. These include 31 functional mature larval neuron clusters, 1 ring gland cluster, 8 glial clusters, 6 neural precursor clusters, and 13 developing immature adult neuron clusters. Some clusters are present across all stages of larval development, while others are stage specific (such as developing adult neurons). We identify genes that are differentially expressed in each cluster, as well as genes that are differentially expressed at distinct stages of larval life. These differentially expressed genes provide promising candidates for regulating the function of specific neuronal and glial types in the larval nervous system, or the specification and differentiation of adult neurons. The cell transcriptome Atlas of the Drosophila larval nervous system is a valuable resource for developmental biology and systems neuroscience and provides a basis for elucidating how genes regulate neural development and function.
Collapse
Affiliation(s)
- Marc Corrales
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA.,Department of Physiology, Development, and Neuroscience, Cambridge University, Cambridge, UK
| | - Benjamin T Cocanougher
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA.,Department of Zoology, Cambridge University, Cambridge, UK
| | - Andrea B Kohn
- Department of Neuroscience and Whitney Laboratory for Marine Biosciences, University of Florida, Gainesville/St. Augustine, FL, 32080, USA
| | - Jason D Wittenbach
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA
| | - Xi S Long
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA
| | - Andrew Lemire
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA
| | - Albert Cardona
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA.,Department of Physiology, Development, and Neuroscience, Cambridge University, Cambridge, UK.,MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, UK
| | - Robert H Singer
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA.,Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Leonid L Moroz
- Department of Neuroscience and Whitney Laboratory for Marine Biosciences, University of Florida, Gainesville/St. Augustine, FL, 32080, USA.
| | - Marta Zlatic
- Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA, USA. .,Department of Zoology, Cambridge University, Cambridge, UK. .,MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge, UK.
| |
Collapse
|
15
|
Zhang CS, Sun LL, Xie JM, Cao CW. RNAi-based functional analysis of bursicon genes related to wing expansion in gypsy moths. JOURNAL OF INSECT PHYSIOLOGY 2022; 139:104398. [PMID: 35537524 DOI: 10.1016/j.jinsphys.2022.104398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/16/2022] [Accepted: 05/04/2022] [Indexed: 06/14/2023]
Abstract
Bursicon is a heterodimeric neuropeptide composed of Burs-α and Burs-β subunits that plays an important role in cuticle tanning and wing expansion in insects. In this study, full-length cDNAs of Burs-α (LdBurs-α) and Burs-β (LdBurs-β) genes were identified in gypsy moth (Lymantria dispar) and cloned. The 480 bp and 420 bp open reading frames (ORFs) encode 159 and 129 amino acid polypeptides, respectively. LdBurs-α and LdBurs-β have 11 conserved cysteine residues, and LdBurs-α and LdBurs-β genes were expressed during all developmental stages according to quantitative reverse transcription PCR (qRT-PCR), with highest expression in the egg stage. High expression levels were also detected in the haemolymph, cuticle and head. To explore the physiological functions of LdBurs-α and LdBurs-β, the genes were knocked down in larvae and pupae using RNA interference (RNAi), and expression levels of LdBurs-α and LdBurs-β were decreased by 42.26-80.09%. Wing defects were observed in L. dispar pupae following Ldbursion silencing, with a phenotypic percentage ranging from 10.17% to 15.00%. RNAi-mediated knockdown of Ldbursicon prevented the expansion of male and female L. dispar adult wings, with malformation rates ranging from 6.38% and 30.00% to 57.69% and 69.23%, but no cuticle tanning defects were observed in pupae or adults. The results indicate that bursicon plays a key role in wing expansion in L. dispar adults, making it a potentially novel molecular target for insecticide-based control of this pest species.
Collapse
Affiliation(s)
- Chen-Shu Zhang
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Li-Li Sun
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Jia-Ming Xie
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin 150040, PR China
| | - Chuan-Wang Cao
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin 150040, PR China.
| |
Collapse
|
16
|
Honda T. Optogenetic and thermogenetic manipulation of defined neural circuits and behaviors in Drosophila. Learn Mem 2022; 29:100-109. [PMID: 35332066 PMCID: PMC8973390 DOI: 10.1101/lm.053556.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/06/2022] [Indexed: 11/25/2022]
Abstract
Neural network dynamics underlying flexible animal behaviors remain elusive. The fruit fly Drosophila melanogaster is considered an excellent model in behavioral neuroscience because of its simple neuroanatomical architecture and the availability of various genetic methods. Moreover, Drosophila larvae's transparent body allows investigators to use optical methods on freely moving animals, broadening research directions. Activating or inhibiting well-defined events in excitable cells with a fine temporal resolution using optogenetics and thermogenetics led to the association of functions of defined neural populations with specific behavioral outputs such as the induction of associative memory. Furthermore, combining optogenetics and thermogenetics with state-of-the-art approaches, including connectome mapping and machine learning-based behavioral quantification, might provide a complete view of the experience- and time-dependent variations of behavioral responses. These methodologies allow further understanding of the functional connections between neural circuits and behaviors such as chemosensory, motivational, courtship, and feeding behaviors and sleep, learning, and memory.
Collapse
Affiliation(s)
- Takato Honda
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| |
Collapse
|
17
|
Sun L, Zhang C, Yan L, Wang X, Wang Z, Cao C. Bursicon receptor gene HLGR2 as a potential RNA interference target for control of the fall webworm Hyphantria cunea. PEST MANAGEMENT SCIENCE 2022; 78:1492-1499. [PMID: 34962063 DOI: 10.1002/ps.6767] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Insect G protein-coupled receptors (GPCRs) have been identified as a new generation of attractive targets for RNA interference (RNAi)-based pest control. A functional study of the leucine-rich repeat-containing (LGR2) gene in Hyphantria cunea (HLGR2) was performed to examine whether it can be used in the molecular control of this notorious pest. RESULTS The complementary DNA (cDNA) sequence and deduced amino acids of HLGR2 were obtained and analyzed in the present study. HLGR2 is a typical GPCR and shows high structural and sequence similarity with other insect LGR2 proteins. The spatiotemporal expression profiles of HLGR2 showed that HLGR2 was highly expressed at the egg stage and tissues of head and silk gland. After RNAi of HLGR2, distinct phenotypes were observed when HLGR2 expression was suppressed, indicating that HLGR2 is essential in pupation and eclosion. HLGR2 RNAi led to a low pupation rate (45.00%), body malformation, abnormal wing expansion, failed cuticle melanization (63.33%), and high mortality rate (48.33%). Furthermore, we identified eight genes that are regulated by HLGR2. The expression of these eight genes was induced by the HLGR2 signaling pathway and correlated well with cuticle sclerotization. Unlike LGR2 in other insect species, HLGR2 was found to play a crucial role in the control of H. cunea during ecdysis and postecdysial stages. CONCLUSION HLGR2 is essential for the growth and development and wing expansion and maturation in H. cunea, suggesting HLGR2 is a promising candidate for application in RNAi-based control of this notorious agriculture-forest pest. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Lili Sun
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin, P. R. China
| | - Chenshu Zhang
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin, P. R. China
| | - Liqiong Yan
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin, P. R. China
| | - Xiaoqi Wang
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin, P. R. China
| | - Zhiying Wang
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin, P. R. China
| | - Chuanwang Cao
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, School of Forestry, Northeast Forestry University, Harbin, P. R. China
| |
Collapse
|
18
|
McClure CD, Hassan A, Aughey GN, Butt K, Estacio-Gómez A, Duggal A, Ying Sia C, Barber AF, Southall TD. An auxin-inducible, GAL4-compatible, gene expression system for Drosophila. eLife 2022; 11:e67598. [PMID: 35363137 PMCID: PMC8975555 DOI: 10.7554/elife.67598] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/27/2022] [Indexed: 01/04/2023] Open
Abstract
The ability to control transgene expression, both spatially and temporally, is essential for studying model organisms. In Drosophila, spatial control is primarily provided by the GAL4/UAS system, whilst temporal control relies on a temperature-sensitive GAL80 (which inhibits GAL4) and drug-inducible systems. However, these are not ideal. Shifting temperature can impact on many physiological and behavioural traits, and the current drug-inducible systems are either leaky, toxic, incompatible with existing GAL4-driver lines, or do not generate effective levels of expression. Here, we describe the auxin-inducible gene expression system (AGES). AGES relies on the auxin-dependent degradation of a ubiquitously expressed GAL80, and therefore, is compatible with existing GAL4-driver lines. Water-soluble auxin is added to fly food at a low, non-lethal, concentration, which induces expression comparable to uninhibited GAL4 expression. The system works in both larvae and adults, providing a stringent, non-lethal, cost-effective, and convenient method for temporally controlling GAL4 activity in Drosophila.
Collapse
Affiliation(s)
- Colin D McClure
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Amira Hassan
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Gabriel N Aughey
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Khushbakht Butt
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers, the State University of New JerseyNew BrunswickUnited States
| | | | - Aneisha Duggal
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Chee Ying Sia
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Annika F Barber
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers, the State University of New JerseyNew BrunswickUnited States
| | - Tony D Southall
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
19
|
Christesen D, Yang YT, Chen W, Batterham P, Perry T. Loss of the Dβ1 nicotinic acetylcholine receptor subunit disrupts bursicon-driven wing expansion and diminishes adult viability in Drosophila melanogaster. Genetics 2021; 219:iyab112. [PMID: 34849910 PMCID: PMC8633089 DOI: 10.1093/genetics/iyab112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/03/2021] [Indexed: 01/15/2023] Open
Abstract
Cholinergic signaling dominates the insect central nervous system, contributing to numerous fundamental pathways and behavioral circuits. However, we are only just beginning to uncover the diverse roles different cholinergic receptors may play. Historically, insect nicotinic acetylcholine receptors have received attention due to several subunits being key insecticide targets. More recently, there has been a focus on teasing apart the roles of these receptors, and their constituent subunits, in native signaling pathways. In this study, we use CRISPR-Cas9 genome editing to generate germline and somatic deletions of the Dβ1 nicotinic acetylcholine receptor subunit and investigate the consequences of loss of function in Drosophila melanogaster. Severe impacts on movement, male courtship, longevity, and wing expansion were found. Loss of Dβ1 was also associated with a reduction in transcript levels for the wing expansion hormone bursicon. Neuron-specific somatic deletion of Dβ1 in bursicon-producing neurons (CCAP-GAL4) was sufficient to disrupt wing expansion. Furthermore, CCAP-GAL4-specific expression of Dβ1 in a germline deletion background was sufficient to rescue the wing phenotype, pinpointing CCAP neurons as the neuronal subset requiring Dβ1 for the wing expansion pathway. Dβ1 is a known target of multiple commercially important insecticides, and the fitness costs exposed here explain why field-isolated target-site resistance has only been reported for amino acid replacements and not loss of function. This work reveals the importance of Dβ1-containing nicotinic acetylcholine receptors in CCAP neurons for robust bursicon-driven wing expansion.
Collapse
Affiliation(s)
- Danielle Christesen
- School of Biosciences, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ying Ting Yang
- School of Biosciences, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Wei Chen
- School of Biosciences, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Philip Batterham
- School of Biosciences, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Trent Perry
- School of Biosciences, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
20
|
Melnattur K, Kirszenblat L, Morgan E, Militchin V, Sakran B, English D, Patel R, Chan D, van Swinderen B, Shaw PJ. A conserved role for sleep in supporting Spatial Learning in Drosophila. Sleep 2021; 44:5909488. [PMID: 32959053 DOI: 10.1093/sleep/zsaa197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/18/2020] [Indexed: 01/25/2023] Open
Abstract
Sleep loss and aging impair hippocampus-dependent Spatial Learning in mammalian systems. Here we use the fly Drosophila melanogaster to investigate the relationship between sleep and Spatial Learning in healthy and impaired flies. The Spatial Learning assay is modeled after the Morris Water Maze. The assay uses a "thermal maze" consisting of a 5 × 5 grid of Peltier plates maintained at 36-37°C and a visual panorama. The first trial begins when a single tile that is associated with a specific visual cue is cooled to 25°C. For subsequent trials, the cold tile is heated, the visual panorama is rotated and the flies must find the new cold tile by remembering its association with the visual cue. Significant learning was observed with two different wild-type strains-Cs and 2U, validating our design. Sleep deprivation prior to training impaired Spatial Learning. Learning was also impaired in the classic learning mutant rutabaga (rut); enhancing sleep restored learning to rut mutants. Further, we found that flies exhibited a dramatic age-dependent cognitive decline in Spatial Learning starting at 20-24 days of age. These impairments could be reversed by enhancing sleep. Finally, we find that Spatial Learning requires dopaminergic signaling and that enhancing dopaminergic signaling in aged flies restored learning. Our results are consistent with the impairments seen in rodents and humans. These results thus demonstrate a critical conserved role for sleep in supporting Spatial Learning, and suggest potential avenues for therapeutic intervention during aging.
Collapse
Affiliation(s)
- Krishna Melnattur
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO
| | - Leonie Kirszenblat
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia.,RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Ellen Morgan
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO
| | - Valentin Militchin
- Department of Otolaryngology, Washington University School of Medicine, St Louis, MO
| | - Blake Sakran
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO
| | - Denis English
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO
| | - Rushi Patel
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO
| | - Dorothy Chan
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO
| | - Bruno van Swinderen
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Paul J Shaw
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
21
|
Lund VK, Lycas MD, Schack A, Andersen RC, Gether U, Kjaerulff O. Rab2 drives axonal transport of dense core vesicles and lysosomal organelles. Cell Rep 2021; 35:108973. [PMID: 33852866 DOI: 10.1016/j.celrep.2021.108973] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 02/10/2021] [Accepted: 03/19/2021] [Indexed: 12/18/2022] Open
Abstract
Fast axonal transport of neuropeptide-containing dense core vesicles (DCVs), endolysosomal organelles, and presynaptic components is critical for maintaining neuronal functionality. How the transport of DCVs is orchestrated remains an important unresolved question. The small GTPase Rab2 mediates DCV biogenesis and endosome-lysosome fusion. Here, we use Drosophila to demonstrate that Rab2 also plays a critical role in bidirectional axonal transport of DCVs, endosomes, and lysosomal organelles, most likely by controlling molecular motors. We further show that the lysosomal motility factor Arl8 is required as well for axonal transport of DCVs, but unlike Rab2, it is also critical for DCV exit from cell bodies into axons. We also provide evidence that the upstream regulators of Rab2 and Arl8, Ema and BORC, activate these GTPases during DCV transport. Our results uncover the mechanisms underlying axonal transport of DCVs and reveal surprising parallels between the regulation of DCV and lysosomal motility.
Collapse
Affiliation(s)
- Viktor Karlovich Lund
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Matthew Domenic Lycas
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Anders Schack
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Rita Chan Andersen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Ulrik Gether
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Ole Kjaerulff
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark.
| |
Collapse
|
22
|
Xu Y, Wei W, Lin G, Yan S, Zhang J, Shen J, Wang D. The Ras/MAPK pathway is required for regenerative growth of wing discs in the black cutworm Agrotis ypsilon. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 131:103552. [PMID: 33577967 DOI: 10.1016/j.ibmb.2021.103552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
Regeneration is a common phenomenon in various organisms by which tissues restore the damaged or naturally detached parts. In insects, appendage regeneration takes place during the embryonic, larval and pupal stages for individual survival. The wing disc of black cutworm Agrotis ypsilon has the capacity of regeneration after ablation, but understanding of molecular mechanisms in wing disc regeneration is still limited. After ablation of partial or whole wing discs before the fifth instar larval stage, the adult wings appeared to be normal. In the last two larval stages, ablation of the left wing disc led to smaller corresponding adult wing. Cell proliferation was reduced in the ablated wing disc but was gradually recovered two days post ablation. Transcriptome analysis found that genes in the mitogen-activated protein kinase (MAPK) pathway were upregulated. Repression of gene expression in this pathway, including Ras oncogene at 64B (Ras64B), Downstream of raf1 (Dsor1), and cAMP-dependent protein kinase catalytic subunit 3 (Pka-C3) by RNA interference after ablation, led to diminishment of both adult wings, suggesting that the MAPK signaling is essential for wing growth. Additionally, cell proliferation was still decelerated by injecting Ras64B, Dsor, or Pka-C3 dsRNA two days after ablation, indicating that the MAPK signaling-regulated cell proliferation is essential for growth. These results provide molecular clues to the regulation of cell proliferation during regeneration in lepidopteran insects.
Collapse
Affiliation(s)
- Yuanyuan Xu
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Wei Wei
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Guangze Lin
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Shuo Yan
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Junzheng Zhang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Jie Shen
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| | - Dan Wang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
23
|
Parallel Synaptic Acetylcholine Signals Facilitate Large Monopolar Cell Repolarization and Modulate Visual Behavior in Drosophila. J Neurosci 2021; 41:2164-2176. [PMID: 33468565 DOI: 10.1523/jneurosci.2388-20.2021] [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: 09/13/2020] [Revised: 12/03/2020] [Accepted: 01/03/2021] [Indexed: 11/21/2022] Open
Abstract
Appropriate termination of the photoresponse in image-forming photoreceptors and downstream neurons is critical for an animal to achieve high temporal resolution. Although the cellular and molecular mechanisms of termination in image-forming photoreceptors have been extensively studied in Drosophila, the underlying mechanism of termination in their downstream large monopolar cells remains less explored. Here, we show that synaptic ACh signaling, from both amacrine cells (ACs) and L4 neurons, facilitates the rapid repolarization of L1 and L2 neurons. Intracellular recordings in female flies show that blocking synaptic ACh output from either ACs or L4 neurons leads to slow repolarization of L1 and L2 neurons. Genetic and electrophysiological studies in both male and female flies determine that L2 neurons express ACh receptors and directly receive ACh signaling. Moreover, our results demonstrate that synaptic ACh signaling from both ACs and L4 neurons simultaneously facilitates ERG termination. Finally, visual behavior studies in both male and female flies show that synaptic ACh signaling, from either ACs or L4 neurons to L2 neurons, is essential for the optomotor response of the flies in high-frequency light stimulation. Our study identifies parallel synaptic ACh signaling for repolarization of L1 and L2 neurons and demonstrates that synaptic ACh signaling facilitates L1 and L2 neuron repolarization to maintain the optomotor response of the fly on high-frequency light stimulation.SIGNIFICANCE STATEMENT The image-forming photoreceptor downstream neurons receive multiple synaptic inputs from image-forming photoreceptors and various types of interneurons. It remains largely unknown how these synaptic inputs modulate the neural activity and function of image-forming photoreceptor downstream neurons. We show that parallel synaptic ACh signaling from both amacrine cells and L4 neurons facilitates rapid repolarization of large monopolar cells in Drosophila and maintains the optomotor response of the fly on high-frequency light stimulation. This work is one of the first reports showing how parallel synaptic signaling modulates the activity of large monopolar cells and motion vision simultaneously.
Collapse
|
24
|
Srivastava P, Kane A, Harrison C, Levin M. A Meta-Analysis of Bioelectric Data in Cancer, Embryogenesis, and Regeneration. Bioelectricity 2021; 3:42-67. [PMID: 34476377 DOI: 10.1089/bioe.2019.0034] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Developmental bioelectricity is the study of the endogenous role of bioelectrical signaling in all cell types. Resting potentials and other aspects of ionic cell physiology are known to be important regulatory parameters in embryogenesis, regeneration, and cancer. However, relevant quantitative measurement and genetic phenotyping data are distributed throughout wide-ranging literature, hampering experimental design and hypothesis generation. Here, we analyze published studies on bioelectrics and transcriptomic and genomic/phenotypic databases to provide a novel synthesis of what is known in three important aspects of bioelectrics research. First, we provide a comprehensive list of channelopathies-ion channel and pump gene mutations-in a range of important model systems with developmental patterning phenotypes, illustrating the breadth of channel types, tissues, and phyla (including man) in which bioelectric signaling is a critical endogenous aspect of embryogenesis. Second, we perform a novel bioinformatic analysis of transcriptomic data during regeneration in diverse taxa that reveals an electrogenic protein to be the one common factor specifically expressed in regeneration blastemas across Kingdoms. Finally, we analyze data on distinct Vmem signatures in normal and cancer cells, revealing a specific bioelectrical signature corresponding to some types of malignancies. These analyses shed light on fundamental questions in developmental bioelectricity and suggest new avenues for research in this exciting field.
Collapse
Affiliation(s)
- Pranjal Srivastava
- Rye High School, Rye, New York, USA; Current Affiliation: College of Chemistry, University of California, Berkeley, Berkeley, California, USA
| | - Anna Kane
- Department of Biology, Allen Discovery Center, Tufts University, Medford, Massachusetts, USA
| | - Christina Harrison
- Department of Biology, Allen Discovery Center, Tufts University, Medford, Massachusetts, USA
| | - Michael Levin
- Department of Biology, Allen Discovery Center, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
25
|
Emmons-Bell M, Hariharan IK. Membrane potential regulates Hedgehog signalling in the Drosophila wing imaginal disc. EMBO Rep 2021; 22:e51861. [PMID: 33629503 PMCID: PMC8024891 DOI: 10.15252/embr.202051861] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 01/02/2023] Open
Abstract
While the membrane potential of cells has been shown to be patterned in some tissues, specific roles for membrane potential in regulating signalling pathways that function during development are still being established. In the Drosophila wing imaginal disc, Hedgehog (Hh) from posterior cells activates a signalling pathway in anterior cells near the boundary which is necessary for boundary maintenance. Here, we show that membrane potential is patterned in the wing disc. Anterior cells near the boundary, where Hh signalling is most active, are more depolarized than posterior cells across the boundary. Elevated expression of the ENaC channel Ripped Pocket (Rpk), observed in these anterior cells, requires Hh. Antagonizing Rpk reduces depolarization and Hh signal transduction. Using genetic and optogenetic manipulations, in both the wing disc and the salivary gland, we show that membrane depolarization promotes membrane localization of Smoothened and augments Hh signalling, independently of Patched. Thus, membrane depolarization and Hh‐dependent signalling mutually reinforce each other in cells immediately anterior to the compartment boundary.
Collapse
Affiliation(s)
- Maya Emmons-Bell
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| |
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: 9.2] [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
|
Vásquez-Procopio J, Rajpurohit S, Missirlis F. Cuticle darkening correlates with increased body copper content in Drosophila melanogaster. Biometals 2020; 33:293-303. [PMID: 33026606 PMCID: PMC7538679 DOI: 10.1007/s10534-020-00245-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/29/2020] [Indexed: 12/18/2022]
Abstract
Insect epidermal cells secrete a cuticle that serves as an exoskeleton providing mechanical rigidity to each individual, but also insulation, camouflage or communication within their environment. Cuticle deposition and hardening (sclerotization) and pigment synthesis are parallel processes requiring tyrosinase activity, which depends on an unidentified copper-dependent enzyme component in Drosophila melanogaster. We determined the metallomes of fly strains selected for lighter or darker cuticles in a laboratory evolution experiment, asking whether any specific element changed in abundance in concert with pigment deposition. The results showed a correlation between total iron content and strength of pigmentation, which was further corroborated by ferritin iron quantification. To ask if the observed increase in iron body content along with increased pigment deposition could be generalizable, we crossed yellow and ebony alleles causing light and dark pigmentation, respectively, into similar genetic backgrounds and measured their metallomes. Iron remained unaffected in the various mutants providing no support for a causative link between pigmentation and iron content. In contrast, the combined analysis of both experiments suggested instead a correlation between pigment deposition and total copper body content, possibly due to increased demand for epidermal tyrosinase activity.
Collapse
Affiliation(s)
- Johana Vásquez-Procopio
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, Zacatenco, Mexico City, Mexico
| | - Subhash Rajpurohit
- Division of Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Commerce Six Road, Navrangpura, Ahmedabad, Gujarat, India
| | - Fanis Missirlis
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, Zacatenco, Mexico City, Mexico.
| |
Collapse
|
28
|
TRPV4 disrupts mitochondrial transport and causes axonal degeneration via a CaMKII-dependent elevation of intracellular Ca 2. Nat Commun 2020; 11:2679. [PMID: 32471994 PMCID: PMC7260201 DOI: 10.1038/s41467-020-16411-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 05/01/2020] [Indexed: 12/14/2022] Open
Abstract
The cation channel transient receptor potential vanilloid 4 (TRPV4) is one of the few identified ion channels that can directly cause inherited neurodegeneration syndromes, but the molecular mechanisms are unknown. Here, we show that in vivo expression of a neuropathy-causing TRPV4 mutant (TRPV4R269C) causes dose-dependent neuronal dysfunction and axonal degeneration, which are rescued by genetic or pharmacological blockade of TRPV4 channel activity. TRPV4R269C triggers increased intracellular Ca2+ through a Ca2+/calmodulin-dependent protein kinase II (CaMKII)-mediated mechanism, and CaMKII inhibition prevents both increased intracellular Ca2+ and neurotoxicity in Drosophila and cultured primary mouse neurons. Importantly, TRPV4 activity impairs axonal mitochondrial transport, and TRPV4-mediated neurotoxicity is modulated by the Ca2+-binding mitochondrial GTPase Miro. Our data highlight an integral role for CaMKII in neuronal TRPV4-associated Ca2+ responses, the importance of tightly regulated Ca2+ dynamics for mitochondrial axonal transport, and the therapeutic promise of TRPV4 antagonists for patients with TRPV4-related neurodegenerative diseases. Mutations in the TRPV4 channel cause inherited neurodegeneration syndromes, but the molecular mechanisms are unknown. Here the authors reveal that TRPV4 activation causes dose-dependent, CaMKII-mediated neuronal dysfunction and axonal degeneration via disruption of mitochondrial axonal transport.
Collapse
|
29
|
Maccallini P, Bavasso F, Scatolini L, Bucciarelli E, Noviello G, Lisi V, Palumbo V, D'Angeli S, Cacchione S, Cenci G, Ciapponi L, Wakefield JG, Gatti M, Raffa GD. Intimate functional interactions between TGS1 and the Smn complex revealed by an analysis of the Drosophila eye development. PLoS Genet 2020; 16:e1008815. [PMID: 32453722 PMCID: PMC7289441 DOI: 10.1371/journal.pgen.1008815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/11/2020] [Accepted: 04/30/2020] [Indexed: 11/27/2022] Open
Abstract
Trimethylguanosine synthase 1 (TGS1) is a conserved enzyme that mediates formation of the trimethylguanosine cap on several RNAs, including snRNAs and telomerase RNA. Previous studies have shown that TGS1 binds the Survival Motor Neuron (SMN) protein, whose deficiency causes spinal muscular atrophy (SMA). Here, we analyzed the roles of the Drosophila orthologs of the human TGS1 and SMN genes. We show that the Drosophila TGS1 protein (dTgs1) physically interacts with all subunits of the Drosophila Smn complex (Smn, Gem2, Gem3, Gem4 and Gem5), and that a human TGS1 transgene rescues the mutant phenotype caused by dTgs1 loss. We demonstrate that both dTgs1 and Smn are required for viability of retinal progenitor cells and that downregulation of these genes leads to a reduced eye size. Importantly, overexpression of dTgs1 partially rescues the eye defects caused by Smn depletion, and vice versa. These results suggest that the Drosophila eye model can be exploited for screens aimed at the identification of genes and drugs that modify the phenotypes elicited by Tgs1 and Smn deficiency. These modifiers could help to understand the molecular mechanisms underlying SMA pathogenesis and devise new therapies for this genetic disease. We explored the functional relationships between TGS1 and SMN using Drosophila as model organism. TGS1 is an enzyme that modifies the structure of the 5’-end of several RNAs, including telomerase RNA and the small nuclear RNAs (snRNAs) that are required for messenger RNA maturation. The SMN protein regulates snRNAs biogenesis and mutations in human SMN cause Spinal Muscular Atrophy (SMA), a devastating disorder characterized by neurodegeneration, progressive paralysis and death. We show that mutations in the Drosophila TGS1 (dTgs1) gene cause lethality, which is rescued by a human TGS1 transgene. We also show that the dTgs1 protein physically interacts with all subunits of the Smn complex, and that downregulation of either dTgs1 or Smn leads to a reduced Drosophila eye size. Notably, overexpression of dTgs1 partially rescues the eye defects caused by Smn knockdown, and vice versa, indicating that these genes cooperate in eye development. These results suggest that the eye model can be exploited for screens aimed at detection of chemical and genetic modifiers of the eye mutant phenotype elicited by dTgs1 and Smn deficiency, providing new clues about SMA pathogenesis and potential therapies.
Collapse
Affiliation(s)
- Paolo Maccallini
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | - Francesca Bavasso
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | - Livia Scatolini
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | | | - Gemma Noviello
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | - Veronica Lisi
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | - Valeria Palumbo
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | - Simone D'Angeli
- Dipartimento di Biologia Ambientale, Sapienza University of Rome, Rome, Italy
| | - Stefano Cacchione
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | - Giovanni Cenci
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
- Fondazione Cenci Bolognetti, Istituto Pasteur, Rome, Italy
| | - Laura Ciapponi
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
| | - James G. Wakefield
- Biosciences/Living Systems Institute, College of Life and Environmental Sciences, University of Exeter, United Kingdom
| | - Maurizio Gatti
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
- Istituto di Biologia e Patologia Molecolari (IBPM) del CNR, Rome, Italy
- * E-mail: (MG); (GDR)
| | - Grazia Daniela Raffa
- Dipartimento di Biologia e Biotecnologie “C Darwin”, Sapienza University of Rome, Rome, Italy
- * E-mail: (MG); (GDR)
| |
Collapse
|
30
|
Lee G, Sehgal R, Wang Z, Park JH. Ultraspiracle-independent anti-apoptotic function of ecdysone receptors is required for the survival of larval peptidergic neurons via suppression of grim expression in Drosophila melanogaster. Apoptosis 2020; 24:256-268. [PMID: 30637539 DOI: 10.1007/s10495-019-01514-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In Drosophila melanogaster a significant number of heterogenous larval neurons in the central nervous system undergo metamorphosis-associated programmed cell death, termed metamorphoptosis. Interestingly distinct groups of doomed larval neurons are eliminated at different metamorphic phases. Although ecdysone hormonal signaling via nuclear ecdysone receptors (EcRs) is known to orchestrate the neuronal metamorphoptosis, little is known about how this signaling controls such diverse neuronal responses. Crustacean cardioactive peptide (CCAP)-producing neurons in the ventral nerve cord are developmentally programmed to die shortly after adult emergence. In this study, we show that disruption of endogenous EcR function by ectopic expression of dominant negative forms of EcRs (EcRDN) causes premature death of larval CCAP neurons in a caspase-dependent manner. This event is rescued by co-expression of individual EcR isoforms. Furthermore, larval CCAP neurons are largely normal in ecr mutants lacking either EcR-A or EcR-B isoforms, suggesting that EcR isoforms redundantly function to protect larval CCAP neurons. Of surprise, a role of Ultraspiracle (Usp), a canonical partner of EcR, is dispensable in the protection of CCAP neurons, whereas both EcR and Usp are required for inducing metamorphoptosis of vCrz neurons shortly after prepupal formation. As a downstream, grim is an essential cell death gene for the EcRDN-mediated CCAP neuronal death, while either hid or rpr function is dispensable. Together, our results suggest that Usp-independent EcR actions protect CCAP neurons from their premature death by repressing grim expression until their normally scheduled apoptosis at post-emergence. Our studies highlight two opposite roles played by EcR function for metamorphoptosis of two different peptidergic neuronal groups, proapoptotic (vCrz) versus antiapoptotic (CCAP), and propose that distinct death timings of doomed larval neurons are determined by differential signaling mechanisms involving EcR.
Collapse
Affiliation(s)
- Gyunghee Lee
- Department of Biochemistry and Cellular and Molecular Biology and NeuroNet Research Center, University of Tennessee, Knoxville, TN, 37996, USA
| | - Ritika Sehgal
- Department of Biochemistry and Cellular and Molecular Biology and NeuroNet Research Center, University of Tennessee, Knoxville, TN, 37996, USA
| | - Zixing Wang
- UT-ORNL Graduate School of Genome Science and Technology Program, University of Tennessee, Knoxville, TN, 37996, USA
| | - Jae H Park
- Department of Biochemistry and Cellular and Molecular Biology and NeuroNet Research Center, University of Tennessee, Knoxville, TN, 37996, USA. .,UT-ORNL Graduate School of Genome Science and Technology Program, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
31
|
Flaven-Pouchon J, Alvarez JV, Rojas C, Ewer J. The tanning hormone, bursicon, does not act directly on the epidermis to tan the Drosophila exoskeleton. BMC Biol 2020; 18:17. [PMID: 32075655 PMCID: PMC7029472 DOI: 10.1186/s12915-020-0742-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/24/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In insects, continuous growth requires the periodic replacement of the exoskeleton. Once the remains of the exoskeleton from the previous stage have been shed during ecdysis, the new one is rapidly sclerotized (hardened) and melanized (pigmented), a process collectively known as tanning. The rapid tanning that occurs after ecdysis is critical for insect survival, as it reduces desiccation, and gives the exoskeleton the rigidity needed to support the internal organs and to provide a solid anchor for the muscles. This rapid postecdysial tanning is triggered by the "tanning hormone", bursicon. Since bursicon is released into the hemolymph, it has naturally been assumed that it would act on the epidermal cells to cause the tanning of the overlying exoskeleton. RESULTS Here we investigated the site of bursicon action in Drosophila by examining the consequences on tanning of disabling the bursicon receptor (encoded by the rickets gene) in different tissues. To our surprise, we found that rapid tanning does not require rickets function in the epidermis but requires it instead in peptidergic neurons of the ventral nervous system (VNS). Although we were unable to identify the signal that is transmitted from the VNS to the epidermis, we show that neurons that express the Drosophila insulin-like peptide ILP7, but not the ILP7 peptide itself, are involved. In addition, we found that some of the bursicon targets involved in melanization are different from those that cause sclerotization. CONCLUSIONS Our findings show that bursicon does not act directly on the epidermis to cause the tanning of the overlying exoskeleton but instead requires an intermediary messenger produced by peptidergic neurons within the central nervous system. Thus, this work has uncovered an unexpected layer of control in a process that is critical for insect survival, which will significantly alter the direction of future research aimed at understanding how rapid postecdysial tanning occurs.
Collapse
Affiliation(s)
| | - Javier V Alvarez
- Instituto de Neurociencia, Universidad de Valparaíso, Valparaiso, Chile
| | - Candy Rojas
- Instituto de Neurociencia, Universidad de Valparaíso, Valparaiso, Chile
| | - John Ewer
- Instituto de Neurociencia, Universidad de Valparaíso, Valparaiso, Chile.
| |
Collapse
|
32
|
Amourda C, Saunders TE. The mirtron miR-1010 functions in concert with its host gene SKIP to balance elevation of nAcRβ2. Sci Rep 2020; 10:1688. [PMID: 32015391 PMCID: PMC6997181 DOI: 10.1038/s41598-020-58655-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/19/2020] [Indexed: 02/02/2023] Open
Abstract
Mirtrons are non-canonical miRNAs arising by splicing and debranching from short introns. A plethora of introns have been inferred by computational analyses as potential mirtrons. Yet, few have been experimentally validated and their functions, particularly in relation to their host genes, remain poorly understood. Here, we found that Drosophila larvae lacking either the mirtron miR-1010 or its binding site in the nicotinic acetylcholine receptor β2 (nAcRβ2) 3′UTR fail to grow properly and pupariate. Increase of cortical nAcRβ2 mediated by neural activity elevates the level of intracellular Ca2+, which in turn activates CaMKII and, further downstream, the transcription factor Adf-1. We show that miR-1010 downregulates nAcRβ2. We reveal that Adf-1 initiates the expression of SKIP, the host gene of miR-1010. Preventing synaptic potentials from overshooting their optimal range requires both SKIP to temper synaptic potentials (incoherent feedforward loop) and miR-1010 to reduce nAcRβ2 mRNA levels (negative feedback loop). Our results demonstrate how a mirtron, in coordination with its host gene, contributes to maintaining appropriate receptor levels, which in turn may play a role in maintaining homeostasis.
Collapse
Affiliation(s)
- Christopher Amourda
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore. .,MRC London Institute of Medical Science, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| | - Timothy E Saunders
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore. .,Department of Biological Sciences, National University of Singapore, Singapore, Singapore. .,Institute of Molecular and Cell Biology, A*Star, Proteos, Singapore.
| |
Collapse
|
33
|
Krishnaswamy S, Huang HW, Marchal IS, Ryoo HD, Sigurdsson EM. Neuronally expressed anti-tau scFv prevents tauopathy-induced phenotypes in Drosophila models. Neurobiol Dis 2020; 137:104770. [PMID: 31982516 DOI: 10.1016/j.nbd.2020.104770] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/10/2020] [Accepted: 01/23/2020] [Indexed: 01/20/2023] Open
Abstract
We have derived single-chain variable fragments (scFv) from tau antibody hybridomas and previously shown their promise as imaging diagnostic agents. Here, we examined the therapeutic potential of anti-tau scFv in transgenic Drosophila models that express in neurons wild-type (WT) human tau (htau) or the human tauopathy mutation R406W. scFv expressing flies were crossed with the tauopathy flies and analyzed. Overall, the survival curves differed significantly (p < .0001). Control flies not expressing htau survived the longest, whereas R406W expressing flies had the shortest lifespan, which was greatly prolonged by co-expressing the anti-tau scFv (p < .0001). Likewise, htau WT expressing flies had a moderately short lifespan, which was prolonged by co-expressing the anti-tau scFv (p < .01). In addition, the htau expression impaired wing expansion after eclosion (p < .0001), and caused progressive abdomen expansion (p < .0001). These features were more severe in htau R406W flies than in htau WT flies. Importantly, both phenotypes were prevented by co-expression of the anti-tau scFv (p < .01-0.0001). Lastly, brain analyses revealed scFv-mediated tau clearance (p < .05-0.01), and its prevention of tau-mediated neurotoxicity (p < .05-0.001). In summary, these findings support the therapeutic potential of an anti-tau scFv, including as gene therapies, and the use of Drosophila models for such screening.
Collapse
Affiliation(s)
- Senthilkumar Krishnaswamy
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, United States of America
| | - Huai-Wei Huang
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, United States of America
| | - Isabella S Marchal
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, United States of America
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, United States of America.
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, United States of America; Department of Psychiatry, New York University School of Medicine, New York, NY 10016, United States of America; Neuroscience Institute, New York University School of Medicine, New York, NY 10016, United States of America.
| |
Collapse
|
34
|
Simon E, de la Puebla SF, Guerrero I. Drosophila Zic family member odd-paired is needed for adult post-ecdysis maturation. Open Biol 2019; 9:190245. [PMID: 31847787 PMCID: PMC6936260 DOI: 10.1098/rsob.190245] [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] [Indexed: 01/05/2023] Open
Abstract
Specific neuropeptides regulate in arthropods the shedding of the old cuticle (ecdysis) followed by maturation of the new cuticle. In Drosophila melanogaster, the last ecdysis occurs at eclosion from the pupal case, with a post-eclosion behavioural sequence that leads to wing extension, cuticle stretching and tanning. These events are highly stereotyped and are controlled by a subset of crustacean cardioactive peptide (CCAP) neurons through the expression of the neuropeptide Bursicon (Burs). We have studied the role of the transcription factor Odd-paired (Opa) during the post-eclosion period. We report that opa is expressed in the CCAP neurons of the central nervous system during various steps of the ecdysis process and in peripheral CCAP neurons innerving the larval muscles involved in adult ecdysis. We show that its downregulation alters Burs expression in the CCAP neurons. Ectopic expression of Opa, or the vertebrate homologue Zic2, in the CCAP neurons also affects Burs expression, indicating an evolutionary functional conservation. Finally, our results show that, independently of its role in Burs regulation, Opa prevents death of CCAP neurons during larval development.
Collapse
Affiliation(s)
- Eléanor Simon
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Sergio Fernández de la Puebla
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Isabel Guerrero
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
35
|
Masuzzo A, Manière G, Viallat-Lieutaud A, Avazeri É, Zugasti O, Grosjean Y, Kurz CL, Royet J. Peptidoglycan-dependent NF-κB activation in a small subset of brain octopaminergic neurons controls female oviposition. eLife 2019; 8:50559. [PMID: 31661076 PMCID: PMC6819134 DOI: 10.7554/elife.50559] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/30/2019] [Indexed: 12/26/2022] Open
Abstract
When facing microbes, animals engage in behaviors that lower the impact of the infection. We previously demonstrated that internal sensing of bacterial peptidoglycan reduces Drosophila female oviposition via NF-κB pathway activation in some neurons (Kurz et al., 2017). Although we showed that the neuromodulator octopamine is implicated, the identity of the involved neurons, as well as the physiological mechanism blocking egg-laying, remained unknown. In this study, we identified few ventral nerve cord and brain octopaminergic neurons expressing an NF-κB pathway component. We functionally demonstrated that NF-κB pathway activation in the brain, but not in the ventral nerve cord octopaminergic neurons, triggers an egg-laying drop in response to infection. Furthermore, we demonstrated via calcium imaging that the activity of these neurons can be directly modulated by peptidoglycan and that these cells do not control other octopamine-dependent behaviors such as female receptivity. This study shows that by sensing peptidoglycan and hence activating NF-κB cascade, a couple of brain neurons modulate a specific octopamine-dependent behavior to adapt female physiology status to their infectious state.
Collapse
Affiliation(s)
- Ambra Masuzzo
- Aix-Marseille Université, CNRS, IBDM, Marseille, France
| | - Gérard Manière
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | | | | | | | - Yaël Grosjean
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, Dijon, France
| | | | - Julien Royet
- Aix-Marseille Université, CNRS, IBDM, Marseille, France
| |
Collapse
|
36
|
Postnatal Increases in Axonal Conduction Velocity of an Identified Drosophila Interneuron Require Fast Sodium, L-Type Calcium and Shaker Potassium Channels. eNeuro 2019; 6:ENEURO.0181-19.2019. [PMID: 31253715 PMCID: PMC6709211 DOI: 10.1523/eneuro.0181-19.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 11/21/2022] Open
Abstract
During early postnatal life, speed up of signal propagation through many central and peripheral neurons has been associated with an increase in axon diameter or/and myelination. Especially in unmyelinated axons postnatal adjustments of axonal membrane conductances is potentially a third mechanism but solid evidence is lacking. Here, we show that axonal action potential (AP) conduction velocity in the Drosophila giant fiber (GF) interneuron, which is required for fast long-distance signal conduction through the escape circuit, is increased by 80% during the first day of adult life. Genetic manipulations indicate that this postnatal increase in AP conduction velocity in the unmyelinated GF axon is likely owed to adjustments of ion channel expression or properties rather than axon diameter increases. Specifically, targeted RNAi knock-down of either Para fast voltage-gated sodium, Shaker potassium (Kv1 homologue), or surprisingly, L-type like calcium channels counteracts postnatal increases in GF axonal conduction velocity. By contrast, the calcium-dependent potassium channel Slowpoke (BK) is not essential for postnatal speeding, although it also significantly increases conduction velocity. Therefore, we identified multiple ion channels that function to support fast axonal AP conduction velocity, but only a subset of these are regulated during early postnatal life to maximize conduction velocity. Despite its large diameter (∼7 µm) and postnatal regulation of multiple ionic conductances, mature GF axonal conduction velocity is still 20-60 times slower than that of vertebrate Aβ sensory axons and α motoneurons, thus unraveling the limits of long-range information transfer speed through invertebrate circuits.
Collapse
|
37
|
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: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
This review focuses on neuropeptides and peptide hormones, the largest and most diverse class of neuroactive substances, known in Drosophila and other animals to play roles in almost all aspects of daily life, as w;1;ell as in developmental processes. We provide an update on novel neuropeptides and receptors identified in the last decade, and highlight progress in analysis of neuropeptide signaling in Drosophila. Especially exciting is the huge amount of work published on novel functions of neuropeptides and peptide hormones in Drosophila, largely due to the rapid developments of powerful genetic methods, imaging techniques and innovative assays. We critically discuss the roles of peptides in olfaction, taste, foraging, feeding, clock function/sleep, aggression, mating/reproduction, learning and other behaviors, as well as in regulation of development, growth, metabolic and water homeostasis, stress responses, fecundity, and lifespan. We furthermore provide novel information on neuropeptide distribution and organization of peptidergic systems, as well as the phylogenetic relations between Drosophila neuropeptides and those of other phyla, including mammals. As will be shown, neuropeptide signaling is phylogenetically ancient, and not only are the structures of the peptides, precursors and receptors conserved over evolution, but also many functions of neuropeptide signaling in physiology and behavior.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden.
| | - Meet Zandawala
- Department of Zoology, Stockholm University, Stockholm, Sweden; Department of Neuroscience, Brown University, Providence, RI, USA.
| |
Collapse
|
38
|
Steyaert J, Scheveneels W, Vanneste J, Van Damme P, Robberecht W, Callaerts P, Bogaert E, Van Den Bosch L. FUS-induced neurotoxicity in Drosophila is prevented by downregulating nucleocytoplasmic transport proteins. Hum Mol Genet 2018; 27:4103-4116. [PMID: 30379317 PMCID: PMC6240733 DOI: 10.1093/hmg/ddy303] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/14/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative diseases characterized by the progressive loss of specific groups of neurons. Due to clinical, genetic and pathological overlap, both diseases are considered as the extremes of one disease spectrum and in a number of ALS and FTD patients, fused in sarcoma (FUS) aggregates are present. Even in families with a monogenetic disease cause, a striking variability is observed in disease presentation. This suggests the presence of important modifying genes. The identification of disease-modifying genes will contribute to defining clear therapeutic targets and to understanding the pathways involved in motor neuron death. In this study, we established a novel in vivo screening platform in which new modifying genes of FUS toxicity can be identified. Expression of human FUS induced the selective apoptosis of crustacean cardioactive peptide (CCAP) neurons from the ventral nerve cord of fruit flies. No defects in the development of these neurons were observed nor were the regulatory CCAP neurons from the brain affected. We used the number of CCAP neurons from the ventral nerve cord as an in vivo read-out for FUS toxicity in neurons. Via a targeted screen, we discovered a potent modifying role of proteins involved in nucleocytoplasmic transport. Downregulation of Nucleoporin 154 and Exportin1 (XPO1) prevented FUS-induced neurotoxicity. Moreover, we show that XPO1 interacted with FUS. Silencing XPO1 significantly reduced the propensity of FUS to form inclusions upon stress. Taken together, our findings point to an important role of nucleocytoplasmic transport proteins in FUS-induced ALS/FTD.
Collapse
Affiliation(s)
- Jolien Steyaert
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Wendy Scheveneels
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Joni Vanneste
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Wim Robberecht
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Callaerts
- Department of Human Genetics, Laboratory of Behavioral and Developmental Genetics, KU Leuven, Leuven, Belgium
| | - Elke Bogaert
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| |
Collapse
|
39
|
Wolff T, Rubin GM. Neuroarchitecture of the Drosophila central complex: A catalog of nodulus and asymmetrical body neurons and a revision of the protocerebral bridge catalog. J Comp Neurol 2018; 526:2585-2611. [PMID: 30084503 PMCID: PMC6283239 DOI: 10.1002/cne.24512] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/17/2022]
Abstract
The central complex, a set of neuropils in the center of the insect brain, plays a crucial role in spatial aspects of sensory integration and motor control. Stereotyped neurons interconnect these neuropils with one another and with accessory structures. We screened over 5,000 Drosophila melanogaster GAL4 lines for expression in two neuropils, the noduli (NO) of the central complex and the asymmetrical body (AB), and used multicolor stochastic labeling to analyze the morphology, polarity, and organization of individual cells in a subset of the GAL4 lines that showed expression in these neuropils. We identified nine NO and three AB cell types and describe them here. The morphology of the NO neurons suggests that they receive input primarily in the lateral accessory lobe and send output to each of the six paired noduli. We demonstrate that the AB is a bilateral structure which exhibits asymmetry in size between the left and right bodies. We show that the AB neurons directly connect the AB to the central complex and accessory neuropils, that they target both the left and right ABs, and that one cell type preferentially innervates the right AB. We propose that the AB be considered a central complex neuropil in Drosophila. Finally, we present highly restricted GAL4 lines for most identified protocerebral bridge, NO, and AB cell types. These lines, generated using the split-GAL4 method, will facilitate anatomical studies, behavioral assays, and physiological experiments.
Collapse
Affiliation(s)
- Tanya Wolff
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia
| | - Gerald M Rubin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia
| |
Collapse
|
40
|
Tabuchi M, Monaco JD, Duan G, Bell B, Liu S, Liu Q, Zhang K, Wu MN. Clock-Generated Temporal Codes Determine Synaptic Plasticity to Control Sleep. Cell 2018; 175:1213-1227.e18. [PMID: 30318147 DOI: 10.1016/j.cell.2018.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/31/2018] [Accepted: 09/10/2018] [Indexed: 10/28/2022]
Abstract
Neurons use two main schemes to encode information: rate coding (frequency of firing) and temporal coding (timing or pattern of firing). While the importance of rate coding is well established, it remains controversial whether temporal codes alone are sufficient for controlling behavior. Moreover, the molecular mechanisms underlying the generation of specific temporal codes are enigmatic. Here, we show in Drosophila clock neurons that distinct temporal spike patterns, dissociated from changes in firing rate, encode time-dependent arousal and regulate sleep. From a large-scale genetic screen, we identify the molecular pathways mediating the circadian-dependent changes in ionic flux and spike morphology that rhythmically modulate spike timing. Remarkably, the daytime spiking pattern alone is sufficient to drive plasticity in downstream arousal neurons, leading to increased firing of these cells. These findings demonstrate a causal role for temporal coding in behavior and define a form of synaptic plasticity triggered solely by temporal spike patterns.
Collapse
Affiliation(s)
- Masashi Tabuchi
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joseph D Monaco
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Grace Duan
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Benjamin Bell
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sha Liu
- VIB Center for Brain and Disease Research and Department of Neuroscience, KU Leuven, Leuven, 3000, Belgium
| | - Qili Liu
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kechen Zhang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mark N Wu
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
41
|
Abstract
Since the founding of Drosophila genetics by Thomas Hunt Morgan and his colleagues over 100 years ago, the experimental induction of mosaicism has featured prominently in its recognition as an unsurpassed genetic model organism. The use of genetic mosaics has facilitated the discovery of a wide variety of developmental processes, identified specific cell lineages, allowed the study of recessive embryonic lethal mutations, and demonstrated the existence of cell competition. Here, we discuss how genetic mosaicism in Drosophila became an invaluable research tool that revolutionized developmental biology. We describe the prevailing methods used to produce mosaic animals, and highlight advantages and disadvantages of each genetic system. We cover methods ranging from simple "twin-spot" analysis to more sophisticated systems of multicolor labeling.
Collapse
|
42
|
Hill VM, O’Connor RM, Sissoko GB, Irobunda IS, Leong S, Canman JC, Stavropoulos N, Shirasu-Hiza M. A bidirectional relationship between sleep and oxidative stress in Drosophila. PLoS Biol 2018; 16:e2005206. [PMID: 30001323 PMCID: PMC6042693 DOI: 10.1371/journal.pbio.2005206] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 06/04/2018] [Indexed: 01/02/2023] Open
Abstract
Although sleep appears to be broadly conserved in animals, the physiological functions of sleep remain unclear. In this study, we sought to identify a physiological defect common to a diverse group of short-sleeping Drosophila mutants, which might provide insight into the function and regulation of sleep. We found that these short-sleeping mutants share a common phenotype of sensitivity to acute oxidative stress, exhibiting shorter survival times than controls. We further showed that increasing sleep in wild-type flies using genetic or pharmacological approaches increases survival after oxidative challenge. Moreover, reducing oxidative stress in the neurons of wild-type flies by overexpression of antioxidant genes reduces the amount of sleep. Together, these results support the hypothesis that a key function of sleep is to defend against oxidative stress and also point to a reciprocal role for reactive oxygen species (ROS) in neurons in the regulation of sleep.
Collapse
Affiliation(s)
- Vanessa M. Hill
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, United States of America
| | - Reed M. O’Connor
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, United States of America
| | | | | | - Stephen Leong
- Columbia University, New York, New York, United States of America
| | - Julie C. Canman
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, United States of America
| | - Nicholas Stavropoulos
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, New York, United States of America
| | - Mimi Shirasu-Hiza
- Department of Genetics and Development, Columbia University Medical Center, New York, New York, United States of America
| |
Collapse
|
43
|
Davie K, Janssens J, Koldere D, De Waegeneer M, Pech U, Kreft Ł, Aibar S, Makhzami S, Christiaens V, Bravo González-Blas C, Poovathingal S, Hulselmans G, Spanier KI, Moerman T, Vanspauwen B, Geurs S, Voet T, Lammertyn J, Thienpont B, Liu S, Konstantinides N, Fiers M, Verstreken P, Aerts S. A Single-Cell Transcriptome Atlas of the Aging Drosophila Brain. Cell 2018; 174:982-998.e20. [PMID: 29909982 PMCID: PMC6086935 DOI: 10.1016/j.cell.2018.05.057] [Citation(s) in RCA: 469] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/30/2018] [Accepted: 05/25/2018] [Indexed: 02/06/2023]
Abstract
The diversity of cell types and regulatory states in the brain, and how these change during aging, remains largely unknown. We present a single-cell transcriptome atlas of the entire adult Drosophila melanogaster brain sampled across its lifespan. Cell clustering identified 87 initial cell clusters that are further subclustered and validated by targeted cell-sorting. Our data show high granularity and identify a wide range of cell types. Gene network analyses using SCENIC revealed regulatory heterogeneity linked to energy consumption. During aging, RNA content declines exponentially without affecting neuronal identity in old brains. This single-cell brain atlas covers nearly all cells in the normal brain and provides the tools to study cellular diversity alongside other Drosophila and mammalian single-cell datasets in our unique single-cell analysis platform: SCope (http://scope.aertslab.org). These results, together with SCope, allow comprehensive exploration of all transcriptional states of an entire aging brain. A single-cell atlas of the adult fly brain during aging Network inference reveals regulatory states related to oxidative phosphorylation Cell identity is retained during aging despite exponential decline of gene expression SCope: An online tool to explore and compare single-cell datasets across species
Collapse
Affiliation(s)
- Kristofer Davie
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Jasper Janssens
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Duygu Koldere
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Maxime De Waegeneer
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Uli Pech
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Neurosciences, KU Leuven, Leuven 3000, Belgium
| | - Łukasz Kreft
- VIB Bioinformatics Core, VIB, Ghent 9052, Belgium
| | - Sara Aibar
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Samira Makhzami
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Valerie Christiaens
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Carmen Bravo González-Blas
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | | | - Gert Hulselmans
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Katina I Spanier
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Thomas Moerman
- ESAT, KU Leuven, Leuven 3001, Belgium; Smart Applications and Innovation Services, IMEC, Leuven 3001, Belgium
| | | | - Sarah Geurs
- Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | - Thierry Voet
- Department of Human Genetics KU Leuven, Leuven 3000, Belgium
| | | | | | - Sha Liu
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Neurosciences, KU Leuven, Leuven 3000, Belgium
| | | | - Mark Fiers
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Neurosciences, KU Leuven, Leuven 3000, Belgium
| | - Patrik Verstreken
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Neurosciences, KU Leuven, Leuven 3000, Belgium
| | - Stein Aerts
- VIB Center for Brain & Disease Research, KU Leuven, Leuven 3000, Belgium; Department of Human Genetics KU Leuven, Leuven 3000, Belgium.
| |
Collapse
|
44
|
Nässel DR. Substrates for Neuronal Cotransmission With Neuropeptides and Small Molecule Neurotransmitters in Drosophila. Front Cell Neurosci 2018; 12:83. [PMID: 29651236 PMCID: PMC5885757 DOI: 10.3389/fncel.2018.00083] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/08/2018] [Indexed: 01/11/2023] Open
Abstract
It has been known for more than 40 years that individual neurons can produce more than one neurotransmitter and that neuropeptides often are colocalized with small molecule neurotransmitters (SMNs). Over the years much progress has been made in understanding the functional consequences of cotransmission in the nervous system of mammals. There are also some excellent invertebrate models that have revealed roles of coexpressed neuropeptides and SMNs in increasing complexity, flexibility, and dynamics in neuronal signaling. However, for the fly Drosophila there are surprisingly few functional studies on cotransmission, although there is ample evidence for colocalization of neuroactive compounds in neurons of the CNS, based both on traditional techniques and novel single cell transcriptome analysis. With the hope to trigger interest in initiating cotransmission studies, this review summarizes what is known about Drosophila neurons and neuronal circuits where different neuropeptides and SMNs are colocalized. Coexistence of neuroactive substances has been recorded in different neuron types such as neuroendocrine cells, interneurons, sensory cells and motor neurons. Some of the circuits highlighted here are well established in the analysis of learning and memory, circadian clock networks regulating rhythmic activity and sleep, as well as neurons and neuroendocrine cells regulating olfaction, nociception, feeding, metabolic homeostasis, diuretic functions, reproduction, and developmental processes. One emerging trait is the broad role of short neuropeptide F in cotransmission and presynaptic facilitation in a number of different neuronal circuits. This review also discusses the functional relevance of coexisting peptides in the intestine. Based on recent single cell transcriptomics data, it is likely that the neuronal systems discussed in this review are just a fraction of the total set of circuits where cotransmission occurs in Drosophila. Thus, a systematic search for colocalized neuroactive compounds in further neurons in anatomically defined circuits is of interest for the near future.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| |
Collapse
|
45
|
Selcho M, Mühlbauer B, Hensgen R, Shiga S, Wegener C, Yasuyama K. Anatomical characterization of PDF-tri neurons and peptidergic neurons associated with eclosion behavior in Drosophila. J Comp Neurol 2018; 526:1307-1328. [DOI: 10.1002/cne.24408] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Mareike Selcho
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter; University of Würzburg; Würzburg D-97074 Germany
| | - Barbara Mühlbauer
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter; University of Würzburg; Würzburg D-97074 Germany
| | - Ronja Hensgen
- Animal Physiology, Department of Biology; Philipps-University Marburg; Marburg D-35032 Germany
| | - Sakiko Shiga
- Department of Biology and Geosciences, Graduate School of Science; Osaka City University; Osaka 558-8585 Japan
| | - Christian Wegener
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter; University of Würzburg; Würzburg D-97074 Germany
| | - Kouji Yasuyama
- Department of Natural Sciences; Kawasaki Medical School; Kurashiki 701-0192 Japan
| |
Collapse
|
46
|
Chen D, Dale RK, Lei EP. Shep regulates Drosophila neuronal remodeling by controlling transcription of its chromatin targets. Development 2018; 145:dev.154047. [PMID: 29158441 DOI: 10.1242/dev.154047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 11/07/2017] [Indexed: 11/20/2022]
Abstract
Neuronal remodeling is crucial for formation of the mature nervous system and disruption of this process can lead to neuropsychiatric diseases. Global gene expression changes in neurons during remodeling as well as the factors that regulate these changes remain poorly defined. To elucidate this process, we performed RNA-seq on isolated Drosophila larval and pupal neurons and found upregulated synaptic signaling and downregulated gene expression regulators as a result of normal neuronal metamorphosis. We further tested the role of alan shepard (shep), which encodes an evolutionarily conserved RNA-binding protein required for proper neuronal remodeling. Depletion of shep in neurons prevents the execution of metamorphic gene expression patterns, and shep-regulated genes correspond to Shep chromatin and/or RNA-binding targets. Reduced expression of a Shep-inhibited target gene that we identified, brat, is sufficient to rescue neuronal remodeling defects of shep knockdown flies. Our results reveal direct regulation of transcriptional programs by Shep to regulate neuronal remodeling during metamorphosis.
Collapse
Affiliation(s)
- Dahong Chen
- Nuclear Organization and Gene Expression Section, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan K Dale
- Nuclear Organization and Gene Expression Section, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elissa P Lei
- Nuclear Organization and Gene Expression Section, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Diao F, Elliott AD, Diao F, Shah S, White BH. Neuromodulatory connectivity defines the structure of a behavioral neural network. eLife 2017; 6:29797. [PMID: 29165248 PMCID: PMC5720592 DOI: 10.7554/elife.29797] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
Neural networks are typically defined by their synaptic connectivity, yet synaptic wiring diagrams often provide limited insight into network function. This is due partly to the importance of non-synaptic communication by neuromodulators, which can dynamically reconfigure circuit activity to alter its output. Here, we systematically map the patterns of neuromodulatory connectivity in a network that governs a developmentally critical behavioral sequence in Drosophila. This sequence, which mediates pupal ecdysis, is governed by the serial release of several key factors, which act both somatically as hormones and within the brain as neuromodulators. By identifying and characterizing the functions of the neuronal targets of these factors, we find that they define hierarchically organized layers of the network controlling the pupal ecdysis sequence: a modular input layer, an intermediate central pattern generating layer, and a motor output layer. Mapping neuromodulatory connections in this system thus defines the functional architecture of the network. Why do animals behave the way they do? Behavior occurs in response to signals from the environment, such as those indicating food or danger, or signals from the body, such as those indicating hunger or thirst. The nervous system detects these signals and triggers an appropriate response, such as seeking food or fleeing a threat. But because much of the nervous system takes part in generating these responses, it can make it difficult to understand how even simple behaviors come about. One behavior that has been studied extensively is molting in insects. Molting enables insects to grow and develop, and involves casting off the outer skeleton of the previous developmental stage. To do this, the insect performs a series of repetitive movements, known as an ecdysis sequence. In the fruit fly, the pupal ecdysis sequence consists of three distinct patterns rhythmic abdominal movement. A hormone called ecdysis triggering hormone, or ETH for short, initiates this sequence by triggering the release of two further hormones, Bursicon and CCAP. All three hormones act on the nervous system to coordinate molting behavior, but exactly how they do so is unclear. Diao et al. have now used genetic tools called Trojan exons to identify the neurons of fruit flies on which these hormones act. Trojan exons are short sequences of DNA that can be inserted into non-coding regions of a target gene to mark or manipulate the cells that express it. When a cell uses its copy of the target gene to make a protein, it also makes the product encoded by the Trojan exon. Using this technique, Diao et al. identified three sets of neurons that produce receptor proteins that recognize the molting hormones. Neurons with ETH receptors start the molting process by activating neurons that make Bursicon and CCAP. Neurons with Bursicon receptors then generate motor rhythms within the nervous system. Finally, neurons with CCAP receptors respond to these rhythms and produce the abdominal movements of the ecdysis sequence. Many other animal behaviors depend on substances like ETH, Bursicon and CCAP, which act within the brain to change the activity of neurons and circuits. The work of Diao et al. suggests that identifying the sites at which such substances act can help reveal the circuits that govern complex behaviors.
Collapse
Affiliation(s)
- Feici Diao
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, United States
| | - Amicia D Elliott
- National Institute of General Medical Sciences, National Institutes of Health, Bethesda, United States
| | - Fengqiu Diao
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, United States
| | - Sarav Shah
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, United States
| | - Benjamin H White
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, United States
| |
Collapse
|
48
|
Direct Sensing of Nutrients via a LAT1-like Transporter in Drosophila Insulin-Producing Cells. Cell Rep 2017; 17:137-148. [PMID: 27681427 PMCID: PMC5055474 DOI: 10.1016/j.celrep.2016.08.093] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/28/2016] [Accepted: 08/29/2016] [Indexed: 12/22/2022] Open
Abstract
Dietary leucine has been suspected to play an important role in insulin release, a hormone that controls satiety and metabolism. The mechanism by which insulin-producing cells (IPCs) sense leucine and regulate insulin secretion is still poorly understood. In Drosophila, insulin-like peptides (DILP2 and DILP5) are produced by brain IPCs and are released in the hemolymph after leucine ingestion. Using Ca2+-imaging and ex vivo cultured larval brains, we demonstrate that IPCs can directly sense extracellular leucine levels via minidiscs (MND), a leucine transporter. MND knockdown in IPCs abolished leucine-dependent changes, including loss of DILP2 and DILP5 in IPC bodies, consistent with the idea that MND is necessary for leucine-dependent DILP release. This, in turn, leads to a strong increase in hemolymph sugar levels and reduced growth. GDH knockdown in IPCs also reduced leucine-dependent DILP release, suggesting that nutrient sensing is coupled to the glutamate dehydrogenase pathway. IPCs directly sense extracellular leucine levels via minidiscs (MND) MND knockdown in IPCs abolishes loss of DILP2 and DILP5 This leads to a strong increase in hemolymph sugar levels and reduces growth GDH knockdown in IPCs reduces leucine-dependent DILP release
Collapse
|
49
|
Qian Y, Cao Y, Deng B, Yang G, Li J, Xu R, Zhang D, Huang J, Rao Y. Sleep homeostasis regulated by 5HT2b receptor in a small subset of neurons in the dorsal fan-shaped body of drosophila. eLife 2017; 6:26519. [PMID: 28984573 PMCID: PMC5648528 DOI: 10.7554/elife.26519] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 10/05/2017] [Indexed: 12/14/2022] Open
Abstract
Our understanding of the molecular mechanisms underlying sleep homeostasis is limited. We have taken a systematic approach to study neural signaling by the transmitter 5-hydroxytryptamine (5-HT) in drosophila. We have generated knockout and knockin lines for Trh, the 5-HT synthesizing enzyme and all five 5-HT receptors, making it possible for us to determine their expression patterns and to investigate their functional roles. Loss of the Trh, 5HT1a or 5HT2b gene decreased sleep time whereas loss of the Trh or 5HT2b gene diminished sleep rebound after sleep deprivation. 5HT2b expression in a small subset of, probably a single pair of, neurons in the dorsal fan-shaped body (dFB) is functionally essential: elimination of the 5HT2b gene from these neurons led to loss of sleep homeostasis. Genetic ablation of 5HT2b neurons in the dFB decreased sleep and impaired sleep homeostasis. Our results have shown that serotonergic signaling in specific neurons is required for the regulation of sleep homeostasis.
Collapse
Affiliation(s)
- Yongjun Qian
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Biomembrane and Membrane Biology, PKU-IDG/McGovern Institute For Brain Research, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Yue Cao
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Biomembrane and Membrane Biology, PKU-IDG/McGovern Institute For Brain Research, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China
| | - Bowen Deng
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Biomembrane and Membrane Biology, PKU-IDG/McGovern Institute For Brain Research, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Guang Yang
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Biomembrane and Membrane Biology, PKU-IDG/McGovern Institute For Brain Research, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China
| | - Jiayun Li
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Biomembrane and Membrane Biology, PKU-IDG/McGovern Institute For Brain Research, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China
| | - Rui Xu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Dandan Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Juan Huang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Yi Rao
- Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Biomembrane and Membrane Biology, PKU-IDG/McGovern Institute For Brain Research, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| |
Collapse
|
50
|
Gu T, Zhao T, Kohli U, Hewes RS. The large and small SPEN family proteins stimulate axon outgrowth during neurosecretory cell remodeling in Drosophila. Dev Biol 2017; 431:226-238. [PMID: 28916169 DOI: 10.1016/j.ydbio.2017.09.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/08/2017] [Accepted: 09/09/2017] [Indexed: 11/16/2022]
Abstract
Split ends (SPEN) is the founding member of a well conserved family of nuclear proteins with critical functions in transcriptional regulation and the post-transcriptional processing and nuclear export of transcripts. In animals, the SPEN proteins fall into two size classes that perform either complementary or antagonistic functions in different cellular contexts. Here, we show that the two Drosophila representatives of this family, SPEN and Spenito (NITO), regulate metamorphic remodeling of the CCAP/bursicon neurosecretory cells. CCAP/bursicon cell-targeted overexpression of SPEN had no effect on the larval morphology or the pruning back of the CCAP/bursicon cell axons at the onset of metamorphosis. During the subsequent outgrowth phase of metamorphic remodeling, overexpression of either SPEN or NITO strongly inhibited axon extension, axon branching, peripheral neuropeptide accumulation, and soma growth. Cell-targeted loss-of-function alleles for both spen and nito caused similar reductions in axon outgrowth, indicating that the absolute levels of SPEN and NITO activity are critical to support the developmental plasticity of these neurons. Although nito RNAi did not affect SPEN protein levels, the phenotypes produced by SPEN overexpression were suppressed by nito RNAi. We propose that SPEN and NITO function additively or synergistically in the CCAP/bursicon neurons to regulate multiple aspects of neurite outgrowth during metamorphic remodeling.
Collapse
Affiliation(s)
- Tingting Gu
- Department of Biology, University of Oklahoma, Norman, OK 73019, USA
| | - Tao Zhao
- Department of Biology, University of Oklahoma, Norman, OK 73019, USA
| | - Uday Kohli
- Department of Biology, University of Oklahoma, Norman, OK 73019, USA
| | - Randall S Hewes
- Department of Biology, University of Oklahoma, Norman, OK 73019, USA.
| |
Collapse
|