1
|
Yin C, Morita T, Parrish JZ. A cell atlas of the larval Aedes aegypti ventral nerve cord. Neural Dev 2024; 19:2. [PMID: 38297398 PMCID: PMC10829479 DOI: 10.1186/s13064-023-00178-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/28/2023] [Indexed: 02/02/2024] Open
Abstract
Mosquito-borne diseases account for nearly 1 million human deaths annually, yet we have a limited understanding of developmental events that influence host-seeking behavior and pathogen transmission in mosquitoes. Mosquito-borne pathogens are transmitted during blood meals, hence adult mosquito behavior and physiology have been intensely studied. However, events during larval development shape adult traits, larvae respond to many of the same sensory cues as adults, and larvae are susceptible to infection by many of the same disease-causing agents as adults. Hence, a better understanding of larval physiology will directly inform our understanding of physiological processes in adults. Here, we use single cell RNA sequencing (scRNA-seq) to provide a comprehensive view of cellular composition in the Aedes aegypti larval ventral nerve cord (VNC), a central hub of sensory inputs and motor outputs which additionally controls multiple aspects of larval physiology. We identify more than 35 VNC cell types defined in part by neurotransmitter and neuropeptide expression. We also explore diversity among monoaminergic and peptidergic neurons that likely control key elements of larval physiology and developmental timing, and identify neuroblasts and immature neurons, providing a view of neuronal differentiation in the VNC. Finally, we find that larval cell composition, number, and position are preserved in the adult abdominal VNC, suggesting studies of larval VNC form and function will likely directly inform our understanding adult mosquito physiology. Altogether, these studies provide a framework for targeted analysis of VNC development and neuronal function in Aedes aegypti larvae.
Collapse
Affiliation(s)
- Chang Yin
- Department of Biology, University of Washington, Seattle, WA, 98195, USA
- Division of Education, Marine Biological Laboratory, 7 MBL Street, Woods Hole, MA, 02543, USA
| | - Takeshi Morita
- Division of Education, Marine Biological Laboratory, 7 MBL Street, Woods Hole, MA, 02543, USA
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY, 10065, USA
- Howard Hughes Medical Institute, New York, NY, 10065, USA
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, WA, 98195, USA.
- Division of Education, Marine Biological Laboratory, 7 MBL Street, Woods Hole, MA, 02543, USA.
| |
Collapse
|
2
|
Malhotra P, Basu S. The Intricate Role of Ecdysis Triggering Hormone Signaling in Insect Development and Reproductive Regulation. INSECTS 2023; 14:711. [PMID: 37623421 PMCID: PMC10455322 DOI: 10.3390/insects14080711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/10/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
Insect growth is interrupted by molts, during which the insect develops a new exoskeleton. The exoskeleton confers protection and undergoes shedding between each developmental stage through an evolutionarily conserved and ordered sequence of behaviors, collectively referred to as ecdysis. Ecdysis is triggered by Ecdysis triggering hormone (ETH) synthesized and secreted from peripheral Inka cells on the tracheal surface and plays a vital role in the orchestration of ecdysis in insects and possibly in other arthropod species. ETH synthesized by Inka cells then binds to ETH receptor (ETHR) present on the peptidergic neurons in the central nervous system (CNS) to facilitate synthesis of various other neuropeptides involved in ecdysis. The mechanism of ETH function on ecdysis has been well investigated in holometabolous insects such as moths Manduca sexta and Bombyx mori, fruit fly Drosophila melanogaster, the yellow fever mosquito Aedes aegypti and beetle Tribolium castaneum etc. In contrast, very little information is available about the role of ETH in sequential and gradual growth and developmental changes associated with ecdysis in hemimetabolous insects. Recent studies have identified ETH precursors and characterized functional and biochemical features of ETH and ETHR in a hemimetabolous insect, desert locust, Schistocerca gregaria. Recently, the role of ETH in Juvenile hormone (JH) mediated courtship short-term memory (STM) retention and long-term courtship memory regulation and retention have also been investigated in adult male Drosophila. Our review provides a novel synthesis of ETH signaling cascades and responses in various insects triggering diverse functions in adults and juvenile insects including their development and reproductive regulation and might allow researchers to develop sustainable pest management strategies by identifying novel compounds and targets.
Collapse
Affiliation(s)
| | - Saumik Basu
- Department of Entomology, Washington State University, Pullman, WA 99164, USA;
| |
Collapse
|
3
|
Metaxakis A, Pavlidis M, Tavernarakis N. Neuronal atg1 Coordinates Autophagy Induction and Physiological Adaptations to Balance mTORC1 Signalling. Cells 2023; 12:2024. [PMID: 37626835 PMCID: PMC10453232 DOI: 10.3390/cells12162024] [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: 07/11/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The mTORC1 nutrient-sensing pathway integrates metabolic and endocrine signals into the brain to evoke physiological responses to food deprivation, such as autophagy. Nevertheless, the impact of neuronal mTORC1 activity on neuronal circuits and organismal metabolism remains obscure. Here, we show that mTORC1 inhibition acutely perturbs serotonergic neurotransmission via proteostatic alterations evoked by the autophagy inducer atg1. Neuronal ATG1 alters the intracellular localization of the serotonin transporter, which increases the extracellular serotonin and stimulates the 5HTR7 postsynaptic receptor. 5HTR7 enhances food-searching behaviour and ecdysone-induced catabolism in Drosophila. Along similar lines, the pharmacological inhibition of mTORC1 in zebrafish also stimulates food-searching behaviour via serotonergic activity. These effects occur in parallel with neuronal autophagy induction, irrespective of the autophagic activity and the protein synthesis reduction. In addition, ectopic neuronal atg1 expression enhances catabolism via insulin pathway downregulation, impedes peptidergic secretion, and activates non-cell autonomous cAMP/PKA. The above exert diverse systemic effects on organismal metabolism, development, melanisation, and longevity. We conclude that neuronal atg1 aligns neuronal autophagy induction with distinct physiological modulations, to orchestrate a coordinated physiological response against reduced mTORC1 activity.
Collapse
Affiliation(s)
- Athanasios Metaxakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
| | - Michail Pavlidis
- Department of Biology, University of Crete, 71409 Heraklion, Crete, Greece;
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Crete, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, 71110 Heraklion, Crete, Greece
| |
Collapse
|
4
|
Piñeiro M, Mena W, Ewer J, Orio P. Extracting temporal relationships between weakly coupled peptidergic and motoneuronal signaling: Application to Drosophila ecdysis behavior. PLoS Comput Biol 2021; 17:e1008933. [PMID: 34910730 PMCID: PMC8716061 DOI: 10.1371/journal.pcbi.1008933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/29/2021] [Accepted: 11/14/2021] [Indexed: 11/18/2022] Open
Abstract
Neuromodulators, such as neuropeptides, can regulate and reconfigure neural circuits to alter their output, affecting in this way animal physiology and behavior. The interplay between the activity of neuronal circuits, their modulation by neuropeptides, and the resulting behavior, is still poorly understood. Here, we present a quantitative framework to study the relationships between the temporal pattern of activity of peptidergic neurons and of motoneurons during Drosophila ecdysis behavior, a highly stereotyped motor sequence that is critical for insect growth. We analyzed, in the time and frequency domains, simultaneous intracellular calcium recordings of peptidergic CCAP (crustacean cardioactive peptide) neurons and motoneurons obtained from isolated central nervous systems throughout fictive ecdysis behavior induced ex vivo by Ecdysis triggering hormone. We found that the activity of both neuronal populations is tightly coupled in a cross-frequency manner, suggesting that CCAP neurons modulate the frequency of motoneuron firing. To explore this idea further, we used a probabilistic logistic model to show that calcium dynamics in CCAP neurons can predict the oscillation of motoneurons, both in a simple model and in a conductance-based model capable of simulating many features of the observed neural dynamics. Finally, we developed an algorithm to quantify the motor behavior observed in videos of pupal ecdysis, and compared their features to the patterns of neuronal calcium activity recorded ex vivo. We found that the motor activity of the intact animal is more regular than the motoneuronal activity recorded from ex vivo preparations during fictive ecdysis behavior; the analysis of the patterns of movement also allowed us to identify a new post-ecdysis phase.
Collapse
Affiliation(s)
- Miguel Piñeiro
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Wilson Mena
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Department of Neuroscience, Institut Pasteur, Paris, France
| | - John Ewer
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- * E-mail: (JE); (PO)
| | - Patricio Orio
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
- Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- * E-mail: (JE); (PO)
| |
Collapse
|
5
|
Saad F, Hipfner DR. Extensive crosstalk of G protein-coupled receptors with the Hedgehog signalling pathway. Development 2021; 148:dev189258. [PMID: 33653875 PMCID: PMC10656458 DOI: 10.1242/dev.189258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/19/2021] [Indexed: 12/23/2022]
Abstract
Hedgehog (Hh) ligands orchestrate tissue patterning and growth by acting as morphogens, dictating different cellular responses depending on ligand concentration. Cellular sensitivity to Hh ligands is influenced by heterotrimeric G protein activity, which controls production of the second messenger 3',5'-cyclic adenosine monophosphate (cAMP). cAMP in turn activates Protein kinase A (PKA), which functions as an inhibitor and (uniquely in Drosophila) as an activator of Hh signalling. A few mammalian Gαi- and Gαs-coupled G protein-coupled receptors (GPCRs) have been shown to influence Sonic hedgehog (Shh) responses in this way. To determine whether this is a more-general phenomenon, we carried out an RNAi screen targeting GPCRs in Drosophila. RNAi-mediated depletion of more than 40% of GPCRs tested either decreased or increased Hh responsiveness in the developing Drosophila wing, closely matching the effects of Gαs and Gαi depletion, respectively. Genetic analysis indicated that the orphan GPCR Mthl5 lowers cAMP levels to attenuate Hh responsiveness. Our results identify Mthl5 as a new Hh signalling pathway modulator in Drosophila and suggest that many GPCRs may crosstalk with the Hh pathway in mammals.
Collapse
Affiliation(s)
- Farah Saad
- Institut de recherches cliniques de Montréal, 110 Pine Avenue West, Montreal H2W 1R7, QC, Canada
- Department of Biology, McGill University, Montreal H3A 1B1, QC, Canada
| | - David R. Hipfner
- Institut de recherches cliniques de Montréal, 110 Pine Avenue West, Montreal H2W 1R7, QC, Canada
- Department of Biology, McGill University, Montreal H3A 1B1, QC, Canada
- Département de médecine, Université de Montréal, Montreal H3C 3J7, QC, Canada
| |
Collapse
|
6
|
Kong H, Jing W, Yuan L, Dong C, Zheng M, Tian Z, Hou Q, Cheng Y, Zhang L, Jiang X, Luo L. Bursicon mediates antimicrobial peptide gene expression to enhance crowded larval prophylactic immunity in the oriental armyworm, Mythimna separata. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 115:103896. [PMID: 33075371 DOI: 10.1016/j.dci.2020.103896] [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: 05/21/2020] [Revised: 10/10/2020] [Accepted: 10/10/2020] [Indexed: 06/11/2023]
Abstract
It has been reported that a high population density alters insect prophylactic immunity. Bursicon plays a key role in the prophylactic immunity of newly emerged adults. In this paper, full-length cDNAs encoding the alpha and beta subunits of bursicon in Mythimna separata larvae (Msburs α and Msburs β) were identified. The cDNAs of Msburs α and Msburs β contain open reading frames (ORFs) encoding 145- and 139-amino acid residue proteins, respectively. Multiple alignment sequences and phylogenetic analysis indicated that Msbursicons (Msburs α and Msburs β) are orthologous to bursicons in other lepidopterans. The Msbursicons were expressed throughout all developmental states with higher relative expression during the egg, pupae, and adult stages. Msbursicons (Msburs α and Msburs β) were highly expressed in the ventral nerve cord and brain relative to other tested tissues. Msbursicon expression of larvae subject to high-density treatment (10 larvae per jar) was significantly increased compared with that of the larvae subject to low-density treatment (1 larva per jar) in the whole fourth and fifth instar stages. The trend in the expression of the antimicrobial peptide (AMP) genes cecropin C and defensin in the test stage was accorded and delayed with increased expression of bursicons. Silencing Msburs α (or Msburs β) expression by dsRNA injection in larvae subject to high-density treatment significantly decreased the expression levels of the cecropin C and defensin genes. Recombinant Msbursicon homodimers significantly induced the expression of the cecropin C and defensin genes. There was a notable decrease in the survival rate of the Msburs α (or Msburs β or Mscecropin C or Msdefensin) knockdown larvae infected by Beauveria thuringiensis. Our findings provide the first insights into how larval density mediates AMP gene expression, which subsequently affects the prophylactic immunity of insects under high-density conditions.
Collapse
Affiliation(s)
- Hailong Kong
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, PR China.
| | - Wanghui Jing
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, PR China
| | - Lin Yuan
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, PR China
| | - Chuanlei Dong
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, PR China
| | - Minyuan Zheng
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, PR China
| | - Zhen Tian
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, PR China
| | - Qiuli Hou
- College of Horticulture and Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou, Jiangsu Province, 225009, PR China
| | - Yunxia Cheng
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, PR China
| | - Lei Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, PR China
| | - Xingfu Jiang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, PR China.
| | - Lizhi Luo
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Yuanmingyuan West Road, No. 2, Beijing, 100193, PR China
| |
Collapse
|
7
|
Ogienko AA, Andreyeva EN, Omelina ES, Oshchepkova AL, Pindyurin AV. Molecular and cytological analysis of widely-used Gal4 driver lines for Drosophila neurobiology. BMC Genet 2020; 21:96. [PMID: 33092520 PMCID: PMC7583314 DOI: 10.1186/s12863-020-00895-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 11/13/2022] Open
Abstract
Background The Drosophila central nervous system (CNS) is a convenient model system for the study of the molecular mechanisms of conserved neurobiological processes. The manipulation of gene activity in specific cell types and subtypes of the Drosophila CNS is frequently achieved by employing the binary Gal4/UAS system. However, many Gal4 driver lines available from the Bloomington Drosophila Stock Center (BDSC) and commonly used in Drosophila neurobiology are still not well characterized. Among these are three lines with Gal4 driven by the elav promoter (BDSC #8760, #8765, and #458), one line with Gal4 driven by the repo promoter (BDSC #7415), and the 69B-Gal4 line (BDSC #1774). For most of these lines, the exact insertion sites of the transgenes and the detailed expression patterns of Gal4 are not known. This study is aimed at filling these gaps. Results We have mapped the genomic location of the Gal4-bearing P-elements carried by the BDSC lines #8760, #8765, #458, #7415, and #1774. In addition, for each of these lines, we have analyzed the Gal4-driven GFP expression pattern in the third instar larval CNS and eye-antennal imaginal discs. Localizations of the endogenous Elav and Repo proteins were used as markers of neuronal and glial cells, respectively. Conclusions We provide a mini-atlas of the spatial activity of Gal4 drivers that are widely used for the expression of UAS–target genes in the Drosophila CNS. The data will be helpful for planning experiments with these drivers and for the correct interpretation of the results.
Collapse
Affiliation(s)
- Anna A Ogienko
- Institute of Molecular and Cellular Biology, Siberian Branch of RAS, Novosibirsk, 630090, Russia
| | - Evgeniya N Andreyeva
- Institute of Molecular and Cellular Biology, Siberian Branch of RAS, Novosibirsk, 630090, Russia
| | - Evgeniya S Omelina
- Institute of Molecular and Cellular Biology, Siberian Branch of RAS, Novosibirsk, 630090, Russia
| | - Anastasiya L Oshchepkova
- Institute of Molecular and Cellular Biology, Siberian Branch of RAS, Novosibirsk, 630090, Russia.,Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of RAS, Novosibirsk, 630090, Russia
| | - Alexey V Pindyurin
- Institute of Molecular and Cellular Biology, Siberian Branch of RAS, Novosibirsk, 630090, Russia. .,Novosibirsk State University, Novosibirsk, 630090, Russia.
| |
Collapse
|
8
|
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
|
9
|
Miguel L, Frebourg T, Campion D, Lecourtois M. Moderate Overexpression of Tau in Drosophila Exacerbates Amyloid-β-Induced Neuronal Phenotypes and Correlates with Tau Oligomerization. J Alzheimers Dis 2020; 74:637-647. [PMID: 32065789 DOI: 10.3233/jad-190906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is neuropathologically defined by two key hallmarks: extracellular senile plaques composed primarily of amyloid-β (Aβ) peptide and intraneuronal neurofibrillary tangles, containing abnormally hyperphosphorylated tau protein. The tau protein is encoded by the MAPT gene. Recently, the H1 and H2 haplotypes of the MAPT gene were associated with AD risk. The minor MAPT H2 haplotype has been linked with a decreased risk of developing late-onset AD (LOAD). MAPT haplotypes show different levels of MAPT/Tau expression with H1 being ∼1.5-fold more expressed than H2, suggesting that MAPT expression level could be related to LOAD risk. In this study, we investigated whether this moderate difference in MAPT/Tau expression could influence Aβ-induced toxicity in vivo. We show that modest overexpression of tau protein in Drosophila exacerbates neuronal phenotypes in AβPP/BACE1 flies. The exacerbation of neuronal defects correlates with the accumulation of insoluble dTau oligomers, suggesting that the moderate difference in level of tau expression observed between H1 and H2 haplotypes could influence Aβ toxicity through the production of oligomeric tau insoluble species.
Collapse
Affiliation(s)
- Laetitia Miguel
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Thierry Frebourg
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Dominique Campion
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Centre Hospitalier du Rouvray, Sotteville-Lès-Rouen, France
| | - Magalie Lecourtois
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| |
Collapse
|
10
|
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
|
11
|
Ding K, Han Y, Seid TW, Buser C, Karigo T, Zhang S, Dickman DK, Anderson DJ. Imaging neuropeptide release at synapses with a genetically engineered reporter. eLife 2019; 8:e46421. [PMID: 31241464 PMCID: PMC6609332 DOI: 10.7554/elife.46421] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/25/2019] [Indexed: 12/26/2022] Open
Abstract
Research on neuropeptide function has advanced rapidly, yet there is still no spatio-temporally resolved method to measure the release of neuropeptides in vivo. Here we introduce Neuropeptide Release Reporters (NPRRs): novel genetically-encoded sensors with high temporal resolution and genetic specificity. Using the Drosophila larval neuromuscular junction (NMJ) as a model, we provide evidence that NPRRs recapitulate the trafficking and packaging of native neuropeptides, and report stimulation-evoked neuropeptide release events as real-time changes in fluorescence intensity, with sub-second temporal resolution.
Collapse
Affiliation(s)
- Keke Ding
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Yifu Han
- Department of NeurobiologyUniversity of Southern CaliforniaLos AngelesUnited States
- Neuroscience Graduate ProgramUniversity of Southern CaliforniaLos AngelesUnited States
| | - Taylor W Seid
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | | | - Tomomi Karigo
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Shishuo Zhang
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Dion K Dickman
- Department of NeurobiologyUniversity of Southern CaliforniaLos AngelesUnited States
| | - David J Anderson
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
- Howard Hughes Medical Institute, California Institute of TechnologyPasadenaUnited States
- Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of TechnologyPasadenaUnited States
| |
Collapse
|
12
|
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
|
13
|
Khan AM, Ashfaq M, Khan AA, Naseem MT, Mansoor S. Evaluation of potential RNA-interference-target genes to control cotton mealybug, Phenacoccus solenopsis (Hemiptera: Pseudococcuidae). INSECT SCIENCE 2018; 25:778-786. [PMID: 28316131 DOI: 10.1111/1744-7917.12455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 02/10/2017] [Accepted: 02/22/2017] [Indexed: 05/20/2023]
Abstract
RNA interference (RNAi) of vital insect genes is a potential tool for targeted pest control. However, selection of the right target genes is a challenge because the RNAi efficacy is known to vary among insect species. Cotton mealybug, Phenacoccus solenopsis, is a phloem-feeding economically important crop pest. We evaluated the RNAi of 2 vital genes, Bursicon (PsBur) and V-ATPase (PsV-ATPase) as potential targets in P. solenopsis for its control. PCR fragments of PsBur and PsV-ATPase were amplified using cDNA synthesized from the total RNA. The PCR amplicons were cloned into Potato virus X (PVX) to develop recombinant PVX for the inoculation of Nicotiana tabacum plants for bioassays with healthy P. solenopsis. Reverse-transcription-polymerase chain reaction (RT-PCR) was used to validate the expression of transgenes in the recombinant-PVX-inoculated plants (treated), and suppression of the target genes in the mealybugs exposed to them. The RT-PCR confirmed the expression of transgenes in the treated plants. Mealybug individuals on treated plants either died or showed physical deformities. Further, the population of mealybug was significantly reduced by feeding on N. tabacum expressing RNAi triggers against PsBur and PsV-ATPase. The results conclude that RNAi is activated in P. solenopsis by feeding on N. tabacum expressing RNAi triggering elements of PsBur and PsV-ATPase genes through recombinant PVX vector. Further, V-ATPase and Bursicon genes are potential targets for RNAi-mediated control of P. solenopsis.
Collapse
Affiliation(s)
- Arif M Khan
- National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Muhammad Ashfaq
- National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan
- Biodiversity Institute of Ontario, University of Guelph, Guelph, Ontario, Canada
| | - Azhar A Khan
- College of Agriculture, Bahauddin Zakariya University, Bahadur Campus, Layyah, Pakistan
| | - Muhammad T Naseem
- National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan
| | - Shahid Mansoor
- National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan
| |
Collapse
|
14
|
Iacobucci GJ, Gunawardena S. Ethanol stimulates the in vivo axonal movement of neuropeptide dense-core vesicles in Drosophila motor neurons. J Neurochem 2017; 144:466-482. [PMID: 28960313 DOI: 10.1111/jnc.14230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/10/2017] [Accepted: 09/18/2017] [Indexed: 01/01/2023]
Abstract
Proper neuronal function requires essential biological cargoes to be packaged within membranous vesicles and transported, intracellularly, through the extensive outgrowth of axonal and dendritic fibers. The precise spatiotemporal movement of these cargoes is vital for neuronal survival and, thus, is highly regulated. In this study we test how the axonal movement of a neuropeptide-containing dense-core vesicle (DCV) responds to alcohol stressors. We found that ethanol induces a strong anterograde bias in vesicle movement. Low doses of ethanol stimulate the anterograde movement of neuropeptide-DCV while high doses inhibit bi-directional movement. This process required the presence of functional kinesin-1 motors as reduction in kinesin prevented the ethanol-induced stimulation of the anterograde movement of neuropeptide-DCV. Furthermore, expression of inactive glycogen synthase kinase 3 (GSK-3β) also prevented ethanol-induced stimulation of neuropeptide-DCV movement, similar to pharmacological inhibition of GSK-3β with lithium. Conversely, inhibition of PI3K/AKT signaling with wortmannin led to a partial prevention of ethanol-stimulated transport of neuropeptide-DCV. Taken together, we conclude that GSK-3β signaling mediates the stimulatory effects of ethanol. Therefore, our study provides new insight into the physiological response of the axonal movement of neuropeptide-DCV to exogenous stressors. Cover Image for this Issue: doi: 10.1111/jnc.14165.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biological Sciences, the State University of New York at Buffalo, Buffalo, New York, USA
| | - Shermali Gunawardena
- Department of Biological Sciences, the State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
15
|
Activity Induces Fmr1-Sensitive Synaptic Capture of Anterograde Circulating Neuropeptide Vesicles. J Neurosci 2017; 36:11781-11787. [PMID: 27852784 DOI: 10.1523/jneurosci.2212-16.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/22/2016] [Accepted: 09/26/2016] [Indexed: 12/21/2022] Open
Abstract
Synaptic neuropeptide and neurotrophin stores are maintained by constitutive bidirectional capture of dense-core vesicles (DCVs) as they circulate in and out of the nerve terminal. Activity increases DCV capture to rapidly replenish synaptic neuropeptide stores following release. However, it is not known whether this is due to enhanced bidirectional capture. Here experiments at the Drosophila neuromuscular junction, where DCVs contain neuropeptides and a bone morphogenic protein, show that activity-dependent replenishment of synaptic neuropeptides following release is evident after inhibiting the retrograde transport with the dynactin disruptor mycalolide B or photobleaching DCVs entering a synaptic bouton by retrograde transport. In contrast, photobleaching anterograde transport vesicles entering a bouton inhibits neuropeptide replenishment after activity. Furthermore, tracking of individual DCVs moving through boutons shows that activity selectively increases capture of DCVs undergoing anterograde transport. Finally, upregulating fragile X mental retardation 1 protein (Fmr1, also called FMRP) acts independently of futsch/MAP-1B to abolish activity-dependent, but not constitutive, capture. Fmr1 also reduces presynaptic neuropeptide stores without affecting activity-independent delivery and evoked release. Therefore, presynaptic motoneuron neuropeptide storage is increased by a vesicle capture mechanism that is distinguished from constitutive bidirectional capture by activity dependence, anterograde selectivity, and Fmr1 sensitivity. These results show that activity recruits a separate mechanism than used at rest to stimulate additional synaptic capture of DCVs for future release of neuropeptides and neurotrophins. SIGNIFICANCE STATEMENT Synaptic release of neuropeptides and neurotrophins depends on presynaptic accumulation of dense-core vesicles (DCVs). At rest, DCVs are captured bidirectionally as they circulate through Drosophila motoneuron terminals by anterograde and retrograde transport. Here we show that activity stimulates further synaptic capture that is distinguished from basal capture by its selectivity for anterograde DCVs and its inhibition by overexpression of the fragile X retardation protein Fmr1. Fmr1 dramatically lowers DCV numbers in synaptic boutons. Therefore, activity-dependent anterograde capture is a major determinant of presynaptic peptide stores.
Collapse
|
16
|
Tao J, Bulgari D, Deitcher DL, Levitan ES. Limited distal organelles and synaptic function in extensive monoaminergic innervation. J Cell Sci 2017; 130:2520-2529. [PMID: 28600320 DOI: 10.1242/jcs.201111] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/05/2017] [Indexed: 12/20/2022] Open
Abstract
Organelles such as neuropeptide-containing dense-core vesicles (DCVs) and mitochondria travel down axons to supply synaptic boutons. DCV distribution among en passant boutons in small axonal arbors is mediated by circulation with bidirectional capture. However, it is not known how organelles are distributed in extensive arbors associated with mammalian dopamine neuron vulnerability, and with volume transmission and neuromodulation by monoamines and neuropeptides. Therefore, we studied presynaptic organelle distribution in Drosophila octopamine neurons that innervate ∼20 muscles with ∼1500 boutons. Unlike in smaller arbors, distal boutons in these arbors contain fewer DCVs and mitochondria, although active zones are present. Absence of vesicle circulation is evident by proximal nascent DCV delivery, limited impact of retrograde transport and older distal DCVs. Traffic studies show that DCV axonal transport and synaptic capture are not scaled for extensive innervation, thus limiting distal delivery. Activity-induced synaptic endocytosis and synaptic neuropeptide release are also reduced distally. We propose that limits in organelle transport and synaptic capture compromise distal synapse maintenance and function in extensive axonal arbors, thereby affecting development, plasticity and vulnerability to neurodegenerative disease.
Collapse
Affiliation(s)
- Juan Tao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Dinara Bulgari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David L Deitcher
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14853, USA
| | - Edwin S Levitan
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
17
|
Di Giorgio ML, Esposito A, Maccallini P, Micheli E, Bavasso F, Gallotta I, Vernì F, Feiguin F, Cacchione S, McCabe BD, Di Schiavi E, Raffa GD. WDR79/TCAB1 plays a conserved role in the control of locomotion and ameliorates phenotypic defects in SMA models. Neurobiol Dis 2017; 105:42-50. [PMID: 28502804 DOI: 10.1016/j.nbd.2017.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/19/2017] [Accepted: 05/10/2017] [Indexed: 12/22/2022] Open
Abstract
SMN (Survival Motor Neuron) deficiency is the predominant cause of spinal muscular atrophy (SMA), a severe neurodegenerative disorder that can lead to progressive paralysis and death. Although SMN is required in every cell for proper RNA metabolism, the reason why its loss is especially critical in the motor system is still unclear. SMA genetic models have been employed to identify several modifiers that can ameliorate the deficits induced by SMN depletion. Here we focus on WDR79/TCAB1, a protein important for the biogenesis of several RNA species that has been shown to physically interact with SMN in human cells. We show that WDR79 depletion results in locomotion defects in both Drosophila and Caenorhabditis elegans similar to those elicited by SMN depletion. Consistent with this observation, we find that SMN overexpression rescues the WDR79 loss-of-function phenotype in flies. Most importantly, we also found that WDR79 overexpression ameliorates the locomotion defects induced by SMN depletion in both flies and worms. Our results collectively suggest that WDR79 and SMN play evolutionarily conserved cooperative functions in the nervous system and suggest that WDR79/TCAB1 may have the potential to modify SMA pathogenesis.
Collapse
Affiliation(s)
- Maria Laura Di Giorgio
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Rome, Italy
| | | | - Paolo Maccallini
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Rome, Italy
| | - Emanuela Micheli
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Rome, Italy
| | - Francesca Bavasso
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Rome, Italy
| | - Ivan Gallotta
- Institute of Genetics and Biophysics - ABT, CNR, Naples, Italy
| | - Fiammetta Vernì
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Rome, Italy
| | - Fabian Feiguin
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Stefano Cacchione
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Rome, Italy
| | | | - Elia Di Schiavi
- Institute of Genetics and Biophysics - ABT, CNR, Naples, Italy; Institute of Bioscience and Bioresources, CNR, Naples, Italy
| | - Grazia Daniela Raffa
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Rome, Italy.
| |
Collapse
|
18
|
Song Y, Villeneuve DL, Toyota K, Iguchi T, Tollefsen KE. Ecdysone Receptor Agonism Leading to Lethal Molting Disruption in Arthropods: Review and Adverse Outcome Pathway Development. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:4142-4157. [PMID: 28355071 PMCID: PMC6135102 DOI: 10.1021/acs.est.7b00480] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Molting is critical for growth, development, reproduction, and survival in arthropods. Complex neuroendocrine pathways are involved in the regulation of molting and may potentially become targets of environmental endocrine disrupting chemicals (EDCs). Based on several known ED mechanisms, a wide range of pesticides has been developed to combat unwanted organisms in food production activities such as agriculture and aquaculture. Meanwhile, these chemicals may also pose hazards to nontarget species by causing molting defects, and thus potentially affecting the health of the ecosystems. The present review summarizes the available knowledge on molting-related endocrine regulation and chemically mediated disruption in arthropods (with special focus on insects and crustaceans), to identify research gaps and develop a mechanistic model for assessing environmental hazards of these compounds. Based on the review, multiple targets of EDCs in the molting processes were identified and the link between mode of action (MoA) and adverse effects characterized to inform future studies. An adverse outcome pathway (AOP) describing ecdysone receptor agonism leading to incomplete ecdysis associated mortality was developed according to the OECD guideline and subjected to weight of evidence considerations by evolved Bradford Hill Criteria. This review proposes the first invertebrate ED AOP and may serve as a knowledge foundation for future environmental studies and AOP development.
Collapse
Affiliation(s)
- You Song
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway
- Corresponding Author: Knut Erik Tollefsen, Norwegian Institute for Water Research (NIVA), Gaustadalléen 21, NO-0349 Oslo, Norway. Tlf.: 02348, Fax: (+47) 22 18 52 00, , You Song, Norwegian Institute for Water Research (NIVA), Gaustadalléen 21, NO-0349 Oslo, Norway. Tlf.: 02348, Fax: (+47) 22 18 52 00,
| | | | - Kenji Toyota
- Environmental Genomics Group, School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Taisen Iguchi
- Department of Basic Biology, Faculty of Life Science, SOKENDAI (Graduate University for Advanced Studies), Okazaki Institute for Integrative Bioscience, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
- Graduate School of Nanobioscience, Yokohama City University, Yokohama 236-0027, Japan
| | - Knut Erik Tollefsen
- Norwegian Institute for Water Research (NIVA), Section of Ecotoxicology and Risk Assessment, Gaustadalléen 21, N-0349 Oslo, Norway
- Norwegian University of Life Sciences (NMBU), Faculty of Environmental Science and Technology, Department of Environmental Sciences (IMV). P.O. Box 5003, N-1432 Ås, Norway
- Corresponding Author: Knut Erik Tollefsen, Norwegian Institute for Water Research (NIVA), Gaustadalléen 21, NO-0349 Oslo, Norway. Tlf.: 02348, Fax: (+47) 22 18 52 00, , You Song, Norwegian Institute for Water Research (NIVA), Gaustadalléen 21, NO-0349 Oslo, Norway. Tlf.: 02348, Fax: (+47) 22 18 52 00,
| |
Collapse
|
19
|
|
20
|
Costa CP, Elias-Neto M, Falcon T, Dallacqua RP, Martins JR, Bitondi MMG. RNAi-Mediated Functional Analysis of Bursicon Genes Related to Adult Cuticle Formation and Tanning in the Honeybee, Apis mellifera. PLoS One 2016; 11:e0167421. [PMID: 27907116 PMCID: PMC5132263 DOI: 10.1371/journal.pone.0167421] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/14/2016] [Indexed: 11/18/2022] Open
Abstract
Bursicon is a heterodimeric neurohormone that acts through a G protein-coupled receptor named rickets (rk), thus inducing an increase in cAMP and the activation of tyrosine hydroxylase, the rate-limiting enzyme in the cuticular tanning pathway. In insects, the role of bursicon in the post-ecdysial tanning of the adult cuticle and wing expansion is well characterized. Here we investigated the roles of the genes encoding the bursicon subunits during the adult cuticle development in the honeybee, Apis mellifera. RNAi-mediated knockdown of AmBurs α and AmBurs β bursicon genes prevented the complete formation and tanning (melanization/sclerotization) of the adult cuticle. A thinner, much less tanned cuticle was produced, and ecdysis toward adult stage was impaired. Consistent with these results, the knockdown of bursicon transcripts also interfered in the expression of genes encoding its receptor, AmRk, structural cuticular proteins, and enzymes in the melanization/sclerotization pathway, thus evidencing roles for bursicon in adult cuticle formation and tanning. Moreover, the expression of AmBurs α, AmBurs β and AmRk is contingent on the declining ecdysteroid titer that triggers the onset of adult cuticle synthesis and deposition. The search for transcripts of AmBurs α, AmBurs β and candidate targets in RNA-seq libraries prepared with brains and integuments strengthened our data on transcript quantification through RT-qPCR. Together, our results support our premise that bursicon has roles in adult cuticle formation and tanning, and are in agreement with other recent studies pointing for roles during the pharate-adult stage, in addition to the classical post-ecdysial ones.
Collapse
Affiliation(s)
- Claudinéia Pereira Costa
- Departamento de Genética; Faculdade de Medicina de Ribeirão Preto; Universidade de São Paulo; Ribeirão Preto, SP, Brazil
- Departamento de Biologia; Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto; Universidade de São Paulo; Ribeirão Preto, SP, Brazil
| | - Moysés Elias-Neto
- Departamento de Biologia; Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto; Universidade de São Paulo; Ribeirão Preto, SP, Brazil
| | - Tiago Falcon
- Departamento de Genética; Faculdade de Medicina de Ribeirão Preto; Universidade de São Paulo; Ribeirão Preto, SP, Brazil
| | - Rodrigo Pires Dallacqua
- Centro de Ciências Biológicas e da Saúde; Universidade Federal de Mato Grosso do Sul; Campo Grande, MS, Brazil
| | - Juliana Ramos Martins
- Departamento de Genética; Faculdade de Medicina de Ribeirão Preto; Universidade de São Paulo; Ribeirão Preto, SP, Brazil
| | - Marcia Maria Gentile Bitondi
- Departamento de Biologia; Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto; Universidade de São Paulo; Ribeirão Preto, SP, Brazil
- * E-mail:
| |
Collapse
|
21
|
Anllo L, Schüpbach T. Signaling through the G-protein-coupled receptor Rickets is important for polarity, detachment, and migration of the border cells in Drosophila. Dev Biol 2016; 414:193-206. [PMID: 27130192 PMCID: PMC4887387 DOI: 10.1016/j.ydbio.2016.04.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/08/2016] [Accepted: 04/24/2016] [Indexed: 01/25/2023]
Abstract
Cell migration plays crucial roles during development. An excellent model to study coordinated cell movements is provided by the migration of border cell clusters within a developing Drosophila egg chamber. In a mutagenesis screen, we isolated two alleles of the gene rickets (rk) encoding a G-protein-coupled receptor. The rk alleles result in border cell migration defects in a significant fraction of egg chambers. In rk mutants, border cells are properly specified and express the marker Slbo. Yet, analysis of both fixed as well as live samples revealed that some single border cells lag behind the main border cell cluster during migration, or, in other cases, the entire border cell cluster can remain tethered to the anterior epithelium as it migrates. These defects are observed significantly more often in mosaic border cell clusters, than in full mutant clusters. Reduction of the Rk ligand, Bursicon, in the border cell cluster also resulted in migration defects, strongly suggesting that Rk signaling is utilized for communication within the border cell cluster itself. The mutant border cell clusters show defects in localization of the adhesion protein E-cadherin, and apical polarity proteins during migration. E-cadherin mislocalization occurs in mosaic clusters, but not in full mutant clusters, correlating well with the rk border cell migration phenotype. Our work has identified a receptor with a previously unknown role in border cell migration that appears to regulate detachment and polarity of the border cell cluster coordinating processes within the cells of the cluster themselves.
Collapse
Affiliation(s)
- Lauren Anllo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Trudi Schüpbach
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
22
|
He X, Zhou S, St. Armour GE, Mackay TFC, Anholt RRH. Epistatic partners of neurogenic genes modulate Drosophila olfactory behavior. GENES, BRAIN, AND BEHAVIOR 2016; 15:280-90. [PMID: 26678546 PMCID: PMC4841442 DOI: 10.1111/gbb.12279] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/02/2015] [Accepted: 12/04/2015] [Indexed: 02/04/2023]
Abstract
The extent to which epistasis affects the genetic architecture of complex traits is difficult to quantify, and identifying variants in natural populations with epistatic interactions is challenging. Previous studies in Drosophila implicated extensive epistasis between variants in genes that affect neural connectivity and contribute to natural variation in olfactory response to benzaldehyde. In this study, we implemented a powerful screen to quantify the extent of epistasis as well as identify candidate interacting variants using 203 inbred wild-derived lines with sequenced genomes of the Drosophila melanogaster Genetic Reference Panel (DGRP). We crossed the DGRP lines to P[GT1]-element insertion mutants in Sema-5c and neuralized (neur), two neurodevelopmental loci which affect olfactory behavior, and to their coisogenic wild-type control. We observed significant variation in olfactory responses to benzaldehyde among F1 genotypes and for the DGRP line by mutant genotype interactions for both loci, showing extensive nonadditive genetic variation. We performed genome-wide association analyses to identify the candidate modifier loci. None of these polymorphisms were in or near the focal genes; therefore, epistasis is the cause of the nonadditive genetic variance. Candidate genes could be placed in interaction networks. Several candidate modifiers are associated with neural development. Analyses of mutants of candidate epistatic partners with neur (merry-go-round (mgr), prospero (pros), CG10098, Alhambra (Alh) and CG12535) and Sema-5c (CG42540 and bruchpilot (brp)) showed aberrant olfactory responses compared with coisogenic controls. Thus, integrating genome-wide analyses of natural variants with mutations at defined genomic locations in a common coisogenic background can unmask specific epistatic modifiers of behavioral phenotypes.
Collapse
Affiliation(s)
- X. He
- Department of EntomologySouth China Agricultural UniversityGuangzhouChina
| | - S. Zhou
- Department of Biological SciencesProgram in Genetics and W. M. Keck Center for Behavioral BiologyRaleighNCUSA
| | - G. E. St. Armour
- Department of Biological SciencesProgram in Genetics and W. M. Keck Center for Behavioral BiologyRaleighNCUSA
| | - T. F. C. Mackay
- Department of Biological SciencesProgram in Genetics and W. M. Keck Center for Behavioral BiologyRaleighNCUSA
| | - R. R. H. Anholt
- Department of Biological SciencesProgram in Genetics and W. M. Keck Center for Behavioral BiologyRaleighNCUSA
| |
Collapse
|
23
|
Phadnis N, Baker EP, Cooper JC, Frizzell KA, Hsieh E, de la Cruz AFA, Shendure J, Kitzman JO, Malik HS. An essential cell cycle regulation gene causes hybrid inviability in Drosophila. Science 2016; 350:1552-5. [PMID: 26680200 DOI: 10.1126/science.aac7504] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Speciation, the process by which new biological species arise, involves the evolution of reproductive barriers, such as hybrid sterility or inviability between populations. However, identifying hybrid incompatibility genes remains a key obstacle in understanding the molecular basis of reproductive isolation. We devised a genomic screen, which identified a cell cycle-regulation gene as the cause of male inviability in hybrids resulting from a cross between Drosophila melanogaster and D. simulans. Ablation of the D. simulans allele of this gene is sufficient to rescue the adult viability of hybrid males. This dominantly acting cell cycle regulator causes mitotic arrest and, thereby, inviability of male hybrid larvae. Our genomic method provides a facile means to accelerate the identification of hybrid incompatibility genes in other model and nonmodel systems.
Collapse
Affiliation(s)
- Nitin Phadnis
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA.
| | - EmilyClare P Baker
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jacob C Cooper
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Emily Hsieh
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Jay Shendure
- Genome Sciences, University of Washington, Seattle, WA 98195, USA. Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Jacob O Kitzman
- Genome Sciences, University of Washington, Seattle, WA 98195, USA. Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Harmit S Malik
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. Howard Hughes Medical Institute, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
24
|
Fernandez-Nicolas A, Belles X. CREB-binding protein contributes to the regulation of endocrine and developmental pathways in insect hemimetabolan pre-metamorphosis. Biochim Biophys Acta Gen Subj 2015; 1860:508-15. [PMID: 26706852 DOI: 10.1016/j.bbagen.2015.12.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/10/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND CREB-binding protein (CBP) is a promiscuous transcriptional co-regulator. In insects, CBP has been studied in the fly Drosophila melanogaster, where it is known as Nejire. Studies in D. melanogaster have revealed that Nejire is involved in the regulation of many pathways during embryo development, especially in anterior/posterior polarity, through Hedgehog and Wingless signaling, and in dorsal/ventral patterning, through TGF-ß signaling. Regarding post-embryonic development, Nejire influences histone acetyl transferase activity on the ecdysone signaling pathway. METHODS AND RESULTS Functional genomics studies using RNAi have shown that CBP contributes to the regulation of feeding and ecdysis during the pre-metamorphic nymphal instar of the cockroach Blattella germanica and is involved in TGF-ß, ecdysone, and MEKRE93 pathways, contributing to the activation of Kr-h1 and E93 expression. In D. melanogaster, Nejire's involvement in the ecdysone pathway in pre-metamorphic stages is conserved, whereas the TGF-ß pathway has only been described in the embryo. CBP role in ecdysis pathway and in the activation of Kr-h1 and E93 expression is described here for the first time. CONCLUSIONS Studies in D. melanogaster may have been suggestive that CBP functions in insects are concentrated in the embryo. Results obtained in B. germanica indicate, however, that CBP have diverse and important functions in post-embryonic development and metamorphosis, especially regarding endocrine signaling. GENERAL SIGNIFICANCE Further research into a higher diversity of models will probably reveal that the multiple post-embryonic roles of CBP observed in B. germanica are general in insects.
Collapse
Affiliation(s)
- Ana Fernandez-Nicolas
- Institut de Biologia Evolutiva (CSIC-UPF), Passeig Marítim de la Barceloneta 37, 08003 Barcelona, Spain
| | - Xavier Belles
- Institut de Biologia Evolutiva (CSIC-UPF), Passeig Marítim de la Barceloneta 37, 08003 Barcelona, Spain.
| |
Collapse
|
25
|
Dong S, Zhang H, Chen X, Stanley D, Yu X, Song Q. The neuropeptide bursicon acts in cuticle metabolism. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2015; 89:87-97. [PMID: 25821138 DOI: 10.1002/arch.21227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Bursicon is a heterodimeric neuropeptide formed of bursicon α (burs α) and bursicon β (burs β) that controls cuticle tanning and wing expansion in insects. Burs α-α and burs β-β homodimers are also formed; they act via an unknown receptor to induce expression of prophylactic immune and stress genes during molting. Based on the hypothesis that burs β-β and/or bursicon influence expression of additional genes acting after the molt, we prepared and sequenced six Drosophila cDNA libraries from groups of flies separately injected with burs β-β, bursicon, or blank control. Compared to the control, the burs β-β treatments led to upregulation (by at least 1.5-fold) of 262 genes at 0.5 h postinjection (PI) and 298 genes at 1 h PI; 323 genes at 0.5 h PI and 269 genes at 1h PI were downregulated (by at least 0.67). Similar changes were recorded following bursicon injections. Of these genes, expression of seven transcripts encoding cuticle proteins was upregulated and three downregulated by burs β-β; expression of nine transcripts encoding cuticle proteins were upregulated and four downregulated following bursicon treatments. Expression of dozens of genes involved in chitin metabolism was altered by the experimental treatments. We recorded parallel changes in expression of selected genes by transcriptome and qPCR analysis. These findings support our hypothesis that burs β-β and bursicon influence expression of additional genes acting after the molt. We report that burs β-β and bursicon act in cuticle synthesis and degradation by regulating the expression of cuticular protein and chitin metabolizing related genes.
Collapse
Affiliation(s)
- Shengzhang Dong
- Division of Plant Sciences, University of Missouri, Columbia, Missouri and Zhejiang Provincial Key Laboratory of Bio-metrology and Inspection & Quarantine, College of Life Sciences, China JiLiang University, Hangzhou, China
| | - Hongwei Zhang
- Division of Plant Sciences, University of Missouri, Columbia, Missouri
| | - Xi Chen
- Division of Plant Sciences, University of Missouri, Columbia, Missouri
| | - David Stanley
- Biological Control of Insects Research Laboratory, USDA/Agricultural Research Service, Columbia, Missouri
| | - Xiaoping Yu
- Zhejiang Provincial Key Laboratory of Bio-metrology and Inspection & Quarantine, College of Life Sciences, China JiLiang University, Hangzhou, China
| | - Qisheng Song
- Division of Plant Sciences, University of Missouri, Columbia, Missouri
| |
Collapse
|
26
|
Vandersmissen HP, Van Hiel MB, Van Loy T, Vleugels R, Vanden Broeck J. Silencing D. melanogaster lgr1 impairs transition from larval to pupal stage. Gen Comp Endocrinol 2014; 209:135-47. [PMID: 25157788 DOI: 10.1016/j.ygcen.2014.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 08/10/2014] [Accepted: 08/14/2014] [Indexed: 12/27/2022]
Abstract
G protein-coupled receptors (GPCRs) play key roles in a wide diversity of physiological processes and signalling pathways. The leucine-rich repeats containing GPCRs (LGRs) are a subfamily that is well-conserved throughout most metazoan phyla and have important regulatory roles in vertebrates. Here, we report on the critical role of Drosophila melanogaster LGR1, the fruit fly homologue of the vertebrate glycoprotein hormone receptors, in development as a factor involved in the regulation of pupariation. Transcript profiling revealed that lgr1 transcripts are most abundant in third instar larvae and adult flies. The tissues displaying the highest transcript levels were the hindgut, the rectum and the salivary glands. Knockdown using RNA interference (RNAi) demonstrated that white pupa formation was severely suppressed in D. melanogaster lgr1 RNAi larvae. Associated with this developmental defect was a reduced ecdysteroid titer, which is in line with significantly reduced transcript levels detected for the Halloween genes shadow (sad) and spookier (spok) in the third instar lgr1 RNAi larvae compared to the control condition.
Collapse
Affiliation(s)
| | - Matthias Boris Van Hiel
- KU Leuven, Animal Physiology and Neurobiology, Naamsestraat 59, PO Box 2465, Leuven, Belgium.
| | - Tom Van Loy
- KU Leuven, Animal Physiology and Neurobiology, Naamsestraat 59, PO Box 2465, Leuven, Belgium.
| | - Rut Vleugels
- KU Leuven, Animal Physiology and Neurobiology, Naamsestraat 59, PO Box 2465, Leuven, Belgium.
| | - Jozef Vanden Broeck
- KU Leuven, Animal Physiology and Neurobiology, Naamsestraat 59, PO Box 2465, Leuven, Belgium.
| |
Collapse
|
27
|
Crimpy enables discrimination of presynaptic and postsynaptic pools of a BMP at the Drosophila neuromuscular junction. Dev Cell 2014; 31:586-98. [PMID: 25453556 DOI: 10.1016/j.devcel.2014.10.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 09/07/2014] [Accepted: 10/03/2014] [Indexed: 01/22/2023]
Abstract
Distinct pools of the bone morphogenetic protein (BMP) Glass bottom boat (Gbb) control structure and function of the Drosophila neuromuscular junction. Specifically, motoneuron-derived Gbb regulates baseline neurotransmitter release, whereas muscle-derived Gbb regulates neuromuscular junction growth. Yet how cells differentiate between these ligand pools is not known. Here we present evidence that the neuronal Gbb-binding protein Crimpy (Cmpy) permits discrimination of pre- and postsynaptic ligand by serving sequential functions in Gbb signaling. Cmpy first delivers Gbb to dense core vesicles (DCVs) for activity-dependent release from presynaptic terminals. In the absence of Cmpy, Gbb is no longer associated with DCVs and is not released by activity. Electrophysiological analyses demonstrate that Cmpy promotes Gbb's proneurotransmission function. Surprisingly, the Cmpy ectodomain is itself released upon DCV exocytosis, arguing that Cmpy serves a second function in BMP signaling. In addition to trafficking Gbb to DCVs, we propose that Gbb/Cmpy corelease from presynaptic terminals defines a neuronal protransmission signal.
Collapse
|
28
|
Harwood BN, Draper I, Kopin AS. Targeted inactivation of the rickets receptor in muscle compromises Drosophila viability. ACTA ACUST UNITED AC 2014; 217:4091-8. [PMID: 25278473 DOI: 10.1242/jeb.110098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Bursicon is a hormone that modulates wing expansion, cuticle hardening and melanization in Drosophila melanogaster. Bursicon activity is mediated through its cognate G protein-coupled receptor (GPCR), rickets. We have developed a membrane-tethered bursicon construct that enables spatial modulation of rickets-mediated physiology in transgenic flies. Ubiquitous expression of tethered bursicon throughout development results in arrest at the pupal stage. The few organisms that eclose fail to undergo wing expansion. These phenotypes suggest that expression of tethered bursicon inhibits rickets-mediated function. Consistent with this hypothesis, we show in vitro that sustained stimulation of rickets by tethered bursicon leads to receptor desensitization. Furthermore, tissue-specific expression of the tethered bursicon inhibitor unraveled a critical role for rickets in a subset of adult muscles. Taken together, our findings highlight the utility of membrane-tethered inhibitors as important genetic/pharmacological tools to dissect the tissue-specific roles of GPCRs in vivo.
Collapse
Affiliation(s)
- Benjamin N Harwood
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, 800 Washington St, Box 7703, Boston, MA 02111, USA Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
| | - Isabelle Draper
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, 800 Washington St, Box 7703, Boston, MA 02111, USA
| | - Alan S Kopin
- Tufts Medical Center, Molecular Cardiology Research Institute, Molecular Pharmacology Research Center, 800 Washington St, Box 7703, Boston, MA 02111, USA Program in Genetics, Sackler School of Graduate Biomedical Sciences, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
29
|
Weiss LC, Laforsch C, Ioannidou I, Herbert Z, Tollrian R. Daphnia longicephala neuropeptides: morphological description of crustacean cardioactive peptide (CCAP) and periviscerokinins in the Ctenodaphnia central nervous system. Neuropeptides 2014; 48:287-93. [PMID: 25069697 DOI: 10.1016/j.npep.2014.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/12/2014] [Accepted: 06/10/2014] [Indexed: 11/18/2022]
Abstract
The publication of the Daphnia genome has driven research in this ecologically relevant model organism in many directions. However, information on this organism's physiology and the relevant controlling factors is limited. In this regard, especially neuropeptides are important biochemical regulators that control a variety of cellular processes, which in combination influence physiological conditions and allow the adaptation of the internal physiological state to external conditions. Thus, neuropeptides are prime in understanding an organism's physiology. We here aimed to detect and describe the distribution of evolutionary conserved neuropeptides including the crustacean cardioactive peptide (CCAP) and peptides of the family periviscerokinins (PVKs) in the central nervous system and the periphery of the Daphnia longicephala head region. We were able to identify a large pair of CCAP immunoreactive cells within central nervous system. In addition, in the periphery we found CCAP immunoreactive cells in the epidermis of the head with processes indicating cuticular secretion. Furthermore, we were able to identify and describe a complex neuronal circuit of PVK neuropeptides in the central nervous system. The data obtained in this study will provide important background information for future investigations aiming to unravel the cellular, neuronal and physiological pathways in a highly adaptive organism such as Daphnia.
Collapse
Affiliation(s)
- Linda C Weiss
- Dept. Animal Ecology, Evolution and Biodiversity, Ruhr University Bochum, Universitätsstr. 150, 447801 Bochum, Germany.
| | - Christian Laforsch
- Department of Animal Ecology I and BayCEER, University of Bayreuth, Universitätsstr. 30, 95440 Bayreuth, Germany
| | - Ioanna Ioannidou
- Dept. Animal Ecology, Evolution and Biodiversity, Ruhr University Bochum, Universitätsstr. 150, 447801 Bochum, Germany
| | - Zsofia Herbert
- Department of Biology II, Ludwig-Maximilians-University Munich, Grosshaderner Str. 2, 82152 Planegg-Martinsried, Germany
| | - Ralph Tollrian
- Dept. Animal Ecology, Evolution and Biodiversity, Ruhr University Bochum, Universitätsstr. 150, 447801 Bochum, Germany
| |
Collapse
|
30
|
Lawton KJ, Wassmer TL, Deitcher DL. Conserved role of Drosophila melanogaster FoxP in motor coordination and courtship song. Behav Brain Res 2014; 268:213-21. [PMID: 24747661 DOI: 10.1016/j.bbr.2014.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 03/12/2014] [Accepted: 04/07/2014] [Indexed: 01/03/2023]
Abstract
FoxP2 is a highly conserved vertebrate transcription factor known for its importance in human speech and language production. Disruption of FoxP2 in several vertebrate models indicates a conserved functional role for this gene in both sound production and motor coordination. Although FoxP2 is known to be strongly expressed in brain regions important for motor coordination, little is known about FoxP2's role in the nervous system. The recent discovery of the well-conserved Drosophila melanogaster homolog, FoxP, provides an opportunity to study the role of this crucial gene in an invertebrate model. We hypothesized that, like FoxP2, Drosophila FoxP is important for behaviors requiring fine motor coordination. We used targeted RNA interference to reduce expression of FoxP and assayed the effects on a variety of adult behaviors. Male flies with reduced FoxP expression exhibit decreased levels of courtship behavior, altered pulse-song structure, and sex-specific motor impairments in walking and flight. Acute disruption of synaptic activity in FoxP expressing neurons using a temperature-sensitive shibire allele dramatically impaired motor coordination. Utilizing a GFP reporter to visualize FoxP in the fly brain reveals expression in relatively few neurons in distributed clusters within the larval and adult CNS, including distinct labeling of the adult protocerebral bridge - a section of the insect central complex known to be important for motor coordination and thought to be homologous to areas of the vertebrate basal ganglia. Our results establish the necessity of this gene in motor coordination in an invertebrate model and suggest a functional homology with vertebrate FoxP2.
Collapse
Affiliation(s)
- Kristy J Lawton
- Department of Neurobiology and Behavior, Cornell University, 215 Tower Rd., Mudd Hall, Ithaca, NY 14853, USA.
| | - Taryn L Wassmer
- Department of Neurobiology and Behavior, Cornell University, 215 Tower Rd., Mudd Hall, Ithaca, NY 14853, USA
| | - David L Deitcher
- Department of Neurobiology and Behavior, Cornell University, 215 Tower Rd., Mudd Hall, Ithaca, NY 14853, USA
| |
Collapse
|
31
|
Vesicle capture, not delivery, scales up neuropeptide storage in neuroendocrine terminals. Proc Natl Acad Sci U S A 2014; 111:3597-601. [PMID: 24550480 DOI: 10.1073/pnas.1322170111] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neurons vary in their capacity to produce, store, and release neuropeptides packaged in dense-core vesicles (DCVs). Specifically, neurons used for cotransmission have terminals that contain few DCVs and many small synaptic vesicles, whereas neuroendocrine neuron terminals contain many DCVs. Although the mechanistic basis for presynaptic variation is unknown, past research demonstrated transcriptional control of neuropeptide synthesis suggesting that supply from the soma limits presynaptic neuropeptide accumulation. Here neuropeptide release is shown to scale with presynaptic neuropeptide stores in identified Drosophila cotransmitting and neuroendocrine terminals. However, the dramatic difference in DCV number in these terminals occurs with similar anterograde axonal transport and DCV half-lives. Thus, differences in presynaptic neuropeptide stores are not explained by DCV delivery from the soma or turnover. Instead, greater neuropeptide accumulation in neuroendocrine terminals is promoted by dramatically more efficient presynaptic DCV capture. Greater capture comes with tradeoffs, however, as fewer uncaptured DCVs are available to populate distal boutons and replenish neuropeptide stores following release. Finally, expression of the Dimmed transcription factor in cotransmitting neurons increases presynaptic DCV capture. Therefore, DCV capture in the terminal is genetically controlled and determines neuron-specific variation in peptidergic function.
Collapse
|
32
|
Cho KH, Daubnerová I, Park Y, Zitnan D, Adams ME. Secretory competence in a gateway endocrine cell conferred by the nuclear receptor βFTZ-F1 enables stage-specific ecdysone responses throughout development in Drosophila. Dev Biol 2013; 385:253-62. [PMID: 24247008 DOI: 10.1016/j.ydbio.2013.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/15/2013] [Accepted: 11/05/2013] [Indexed: 12/24/2022]
Abstract
Hormone-induced changes in gene expression initiate periodic molts and metamorphosis during insect development. Successful execution of these developmental steps depends upon successive phases of rising and falling 20-hydroxyecdysone (20E) levels, leading to a cascade of nuclear receptor-driven transcriptional activity that enables stage- and tissue-specific responses to the steroid. Among the cellular processes associated with declining steroids is acquisition of secretory competence in endocrine Inka cells, the source of ecdysis triggering hormones (ETHs). We show here that Inka cell secretory competence is conferred by the orphan nuclear receptor βFTZ-F1. Selective RNA silencing of βftz-f1 in Inka cells prevents ETH release, causing developmental arrest at all stages. Affected larvae display buttoned-up, the ETH-null phenotype characterized by double mouthparts, absence of ecdysis behaviors, and failure to shed the old cuticle. During the mid-prepupal period, individuals fail to translocate the air bubble, execute head eversion and elongate incipient wings and legs. Those that escape to the adult stage are defective in wing expansion and cuticle sclerotization. Failure to release ETH in βftz-f1 silenced animals is indicated by persistent ETH immunoreactivity in Inka cells. Arrested larvae are rescued by precisely-timed ETH injection or Inka cell-targeted βFTZ-F1 expression. Moreover, premature βftz-f1 expression in these cells also results in developmental arrest. The Inka cell therefore functions as a "gateway cell", whose secretion of ETH serves as a key downstream physiological output enabling stage-specific responses to 20E that are required to advance through critical developmental steps. This secretory function depends on transient and precisely timed βFTZ-F1 expression late in the molt as steroids decline.
Collapse
Affiliation(s)
- Kook-Ho Cho
- Departments of Entomology and Cell Biology & Neuroscience, 2103 Biological Sciences Building, University of California, Riverside, CA 92521, USA
| | - Ivana Daubnerová
- Institute of Zoology, Slovak Academy of Sciences, Dubravska cesta 9, 84506 Bratislava, Slovakia
| | - Yoonseong Park
- Department of Entomology, 123 Waters Hall, Kansas State University, Manhattan, KS 66506, USA
| | - Dusan Zitnan
- Institute of Zoology, Slovak Academy of Sciences, Dubravska cesta 9, 84506 Bratislava, Slovakia
| | - Michael E Adams
- Departments of Entomology and Cell Biology & Neuroscience, 2103 Biological Sciences Building, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
33
|
Lee D, Orchard I, Lange AB. Evidence for a conserved CCAP-signaling pathway controlling ecdysis in a hemimetabolous insect, Rhodnius prolixus. Front Neurosci 2013; 7:207. [PMID: 24204330 PMCID: PMC3817380 DOI: 10.3389/fnins.2013.00207] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 10/16/2013] [Indexed: 12/03/2022] Open
Abstract
A vital feature in the success of Ecdysozoa is their ability to shed their exoskeleton (a process called ecdysis) such that they can grow or change their morphology. In holometabolous insects, these behaviors are orchestrated by the sequential actions of neuropeptides, one of which is crustacean cardioactive peptide (CCAP). Little is known about the control of ecdysis in hemimetabolous insects. Here, we report that CCAP is essential for successful ecdysis in the hemimetabolous insect, Rhodnius prolixus; the vector of Chagas disease. The first indication of CCAP's involvement in ecdysis was the observation of decreased staining intensity of CCAP-containing neurons immediately following ecdysis, indicative of the release of CCAP. The critical importance of the CCAP signaling pathway was further demonstrated by knockdown (as determined by qPCR and immunohistochemistry) of the CCAP and CCAPR transcripts utilizing dsRNA. This technique reduced the staining intensity of CCAP-containing neurons, and knocked down the transcript levels by up to 92%, with lethal consequences to the insect. Insects with these transcripts knocked down had very high mortality (up to 84%), typically at the expected time of the ecdysis sequence, or had ecdysis extremely delayed. This is the first report of the susceptibility of R. prolixus to dsRNA knockdown of neuropeptide and receptor transcripts, and the data clearly demonstrates the conserved nature of the CCAP signaling pathway in ecdysis between holometabolous and hemimetabolous insects.
Collapse
Affiliation(s)
- Dohee Lee
- Department of Biology, University of Toronto Mississauga Mississauga, ON, Canada
| | | | | |
Collapse
|
34
|
Karsai G, Pollák E, Wacker M, Vömel M, Selcho M, Berta G, Nachman RJ, Isaac RE, Molnár L, Wegener C. Diverse in- and output polarities and high complexity of local synaptic and non-synaptic signaling within a chemically defined class of peptidergic Drosophila neurons. Front Neural Circuits 2013; 7:127. [PMID: 23914156 PMCID: PMC3729985 DOI: 10.3389/fncir.2013.00127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/12/2013] [Indexed: 12/31/2022] Open
Abstract
Peptidergic neurons are not easily integrated into current connectomics concepts, since their peptide messages can be distributed via non-synaptic paracrine signaling or volume transmission. Moreover, the polarity of peptidergic interneurons in terms of in- and out-put sites can be hard to predict and is very little explored. We describe in detail the morphology and the subcellular distribution of fluorescent vesicle/dendrite markers in CCAP neurons (NCCAP), a well defined set of peptidergic neurons in the Drosophila larva. NCCAP can be divided into five morphologically distinct subsets. In contrast to other subsets, serial homologous interneurons in the ventral ganglion show a mixed localization of in- and output markers along ventral neurites that defy a classification as dendritic or axonal compartments. Ultrastructurally, these neurites contain both pre- and postsynaptic sites preferably at varicosities. A significant portion of the synaptic events are due to reciprocal synapses. Peptides are mostly non-synaptically or parasynaptically released, and dense-core vesicles and synaptic vesicle pools are typically well separated. The responsiveness of the NCCAP to ecdysis-triggering hormone may be at least partly dependent on a tonic synaptic inhibition, and is independent of ecdysteroids. Our results reveal a remarkable variety and complexity of local synaptic circuitry within a chemically defined set of peptidergic neurons. Synaptic transmitter signaling as well as peptidergic paracrine signaling and volume transmission from varicosities can be main signaling modes of peptidergic interneurons depending on the subcellular region. The possibility of region-specific variable signaling modes should be taken into account in connectomic studies that aim to dissect the circuitry underlying insect behavior and physiology, in which peptidergic neurons act as important regulators.
Collapse
Affiliation(s)
- Gergely Karsai
- Department of Comparative Anatomy and Developmental Biology, Institute of Biology, Faculty of Science, University of Pécs Pécs, Hungary ; Neurobiology and Genetics, Biocenter, Theodor-Boveri-Institute, University of Würzburg Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Boseman A, Nowlin K, Ashraf S, Yang J, LaJeunesse D. Ultrastructural analysis of wild type and mutant Drosophila melanogaster using helium ion microscopy. Micron 2013; 51:26-35. [DOI: 10.1016/j.micron.2013.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 06/12/2013] [Accepted: 06/15/2013] [Indexed: 10/26/2022]
|
36
|
Webster SG, Wilcockson DC, Sharp JH. Bursicon and neuropeptide cascades during the ecdysis program of the shore crab, Carcinus maenas. Gen Comp Endocrinol 2013; 182:54-64. [PMID: 23247273 DOI: 10.1016/j.ygcen.2012.11.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 11/21/2012] [Accepted: 11/23/2012] [Indexed: 10/27/2022]
Abstract
Very little is known regarding the release patterns of neuropeptides involved in ecdysis of crustaceans compared to insects. In particular, the dynamics of release of the insect cuticle hardening hormone bursicon, which has only recently been discovered in crustaceans, is unknown. Bursicon has not previously been identified as a circulating neurohormone in these animals. Since patterns of release were likely to be ephemeral, bursicon, as well as two other neurohormones involved in the ecdysis program in crustaceans, crustacean cardioactive peptide (CCAP) and crustacean hyperglycaemic hormone (CHH) were measured in single haemolymph samples in Carcinus maenas. For bursicon, an ultrasensitive time resolved-fluoroimmunoassay (TR-FIA) was developed, which firstly involved its characterisation by HPLC, bioassay and immunoassay. Simultaneous measurement of three neurohormones was performed at unparalleled levels of resolution, which has not previously been reported in any invertebrate. Additionally, expression patterns and architecture of neurones expressing both bursicon and CCAP were determined in the CNS during the moult cycle. Bursicon and CCAP are released in a massive surge, likely a single global exocytotic event on emergence, just after release of CHH. Despite co-localisation of CCAP and bursicon in neurones of the CNS, observations suggest that differential packaging of CCAP can occur in the pericardial organs in a small population of secretory boutons, thus accounting for observations showing release of some CCAP during the penultimate stages of the ecdysis program. The results obtained vividly illustrate the dynamism of neuropeptide cascades occurring during crustacean ecdysis, and also allow proposal of a hypothesis of its endocrine control.
Collapse
|
37
|
Harwood BN, Fortin JP, Gao K, Chen C, Beinborn M, Kopin AS. Membrane tethered bursicon constructs as heterodimeric modulators of the Drosophila G protein-coupled receptor rickets. Mol Pharmacol 2013; 83:814-21. [PMID: 23340494 DOI: 10.1124/mol.112.081570] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The study of complex heterodimeric peptide ligands has been hampered by a paucity of pharmacological tools. To facilitate such investigations, we have explored the utility of membrane tethered ligands (MTLs). Feasibility of this recombinant approach was explored with a focus on Drosophila bursicon, a heterodimeric cystine-knot protein that activates the G protein-coupled receptor rickets (rk). Rk/bursicon signaling is an evolutionarily conserved pathway in insects required for wing expansion, cuticle hardening, and melanization during development. We initially engineered two distinct MTL constructs, each composed of a type II transmembrane domain, a peptide linker, and a C terminal extracellular ligand that corresponded to either the α or β bursicon subunit. Coexpression of the two complementary bursicon MTLs triggered rk-mediated signaling in vitro. We were then able to generate functionally active bursicon MTLs in which the two subunits were fused into a single heterodimeric peptide, oriented as either α-β or β-α. Carboxy-terminal deletion of 32 amino acids in the β-α MTL construct resulted in loss of agonist activity. Coexpression of this construct with rk inhibited receptor-mediated signaling by soluble bursicon. We have thus generated membrane-anchored bursicon constructs that can activate or inhibit rk signaling. These probes can be used in future studies to explore the tissue and/or developmental stage-dependent effects of bursicon in the genetically tractable Drosophila model organism. In addition, our success in generating functionally diverse bursicon MTLs offers promise that such technology can be broadly applied to other complex ligands, including the family of mammalian cystine-knot proteins.
Collapse
Affiliation(s)
- Benjamin N Harwood
- Molecular Pharmacology Research Center, Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts 02111, USA
| | | | | | | | | | | |
Collapse
|
38
|
Genetic analysis of ecdysis behavior in Drosophila reveals partially overlapping functions of two unrelated neuropeptides. J Neurosci 2012; 32:6819-29. [PMID: 22593051 DOI: 10.1523/jneurosci.5301-11.2012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Ecdysis behavior allows insects to shed their old exoskeleton at the end of every molt. It is controlled by a suite of interacting hormones and neuropeptides, and has served as a useful behavior for understanding how bioactive peptides regulate CNS function. Previous findings suggest that crustacean cardioactive peptide (CCAP) activates the ecdysis motor program; the hormone bursicon is believed to then act downstream of CCAP to inflate, pigment, and harden the exoskeleton of the next stage. However, the exact roles of these signaling molecules in regulating ecdysis remain unclear. Here we use a genetic approach to investigate the functions of CCAP and bursicon in Drosophila ecdysis. We show that null mutants in CCAP express no apparent defects in ecdysis and postecdysis, producing normal adults. By contrast, a substantial fraction of flies genetically null for one of the two subunits of bursicon [encoded by the partner of bursicon gene (pburs)] show severe defects in ecdysis, with escaper adults exhibiting the expected failures in wing expansion and exoskeleton pigmentation and hardening. Furthermore, flies lacking both CCAP and bursicon show much more severe defects at ecdysis than do animals null for either neuropeptide alone. Our results show that the functions thought to be subserved by CCAP are partially effected by bursicon, and that bursicon plays an important and heretofore undescribed role in ecdysis behavior itself. These findings have important implications for understanding the regulation of this vital insect behavior and the mechanisms by which hormones and neuropeptides control the physiology and behavior of animals.
Collapse
|
39
|
Drosophila models of tauopathies: what have we learned? Int J Alzheimers Dis 2012; 2012:970980. [PMID: 22701808 PMCID: PMC3373119 DOI: 10.1155/2012/970980] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 04/08/2012] [Indexed: 01/10/2023] Open
Abstract
Aggregates of the microtubule-associated protein Tau are neuropathological hallmark lesions in Alzheimer's disease (AD) and related primary tauopathies. In addition, Tau is genetically implicated in a number of human neurodegenerative disorders including frontotemporal dementia (FTD) and Parkinson's disease (PD). The exact mechanism by which Tau exerts its neurotoxicity is incompletely understood. Here, we give an overview of how studies using the genetic model organism Drosophila over the past decade have contributed to the molecular understanding of Tau neurotoxicity. We compare the different available readouts for Tau neurotoxicity in flies and review the molecular pathways in which Tau has been implicated. Finally, we emphasize that the integration of genome-wide approaches in human or mice with high-throughput genetic validation in Drosophila is a fruitful approach.
Collapse
|
40
|
|
41
|
Wong MY, Zhou C, Shakiryanova D, Lloyd TE, Deitcher DL, Levitan ES. Neuropeptide delivery to synapses by long-range vesicle circulation and sporadic capture. Cell 2012; 148:1029-38. [PMID: 22385966 DOI: 10.1016/j.cell.2011.12.036] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 10/13/2011] [Accepted: 12/16/2011] [Indexed: 12/11/2022]
Abstract
Neurotransmission requires anterograde axonal transport of dense core vesicles (DCVs) containing neuropeptides and active zone components from the soma to nerve terminals. However, it is puzzling how one-way traffic could uniformly supply sequential release sites called en passant boutons. Here, Drosophila neuropeptide-containing DCVs are tracked in vivo for minutes with a new method called simultaneous photobleaching and imaging (SPAIM). Surprisingly, anterograde DCVs typically bypass proximal boutons to accumulate initially in the most distal bouton. Then, excess distal DCVs undergo dynactin-dependent retrograde transport back through proximal boutons into the axon. Just before re-entering the soma, DCVs again reverse for another round of anterograde axonal transport. While circulating over long distances, both anterograde and retrograde DCVs are captured sporadically in en passant boutons. Therefore, vesicle circulation, which includes long-range retrograde transport and inefficient bidirectional capture, overcomes the limitations of one-way anterograde transport to uniformly supply release sites with DCVs.
Collapse
Affiliation(s)
- Man Yan Wong
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
42
|
An S, Dong S, Wang Q, Li S, Gilbert LI, Stanley D, Song Q. Insect neuropeptide bursicon homodimers induce innate immune and stress genes during molting by activating the NF-κB transcription factor Relish. PLoS One 2012; 7:e34510. [PMID: 22470576 PMCID: PMC3314635 DOI: 10.1371/journal.pone.0034510] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 03/06/2012] [Indexed: 11/19/2022] Open
Abstract
Background Bursicon is a heterodimer neuropeptide composed of two cystine knot proteins, bursicon α (burs α) and bursicon β (burs β), that elicits cuticle tanning (melanization and sclerotization) through the Drosophila leucine-rich repeats-containing G protein-coupled receptor 2 (DLGR2). Recent studies show that both bursicon subunits also form homodimers. However, biological functions of the homodimers have remained unknown until now. Methodology/Principal Findings In this report, we show in Drosophila melanogaster that both bursicon homodimers induced expression of genes encoding antimicrobial peptides (AMPs) in neck-ligated adults following recombinant homodimer injection and in larvae fat body after incubation with recombinant homodimers. These AMP genes were also up-regulated in 24 h old unligated flies (when the endogenous bursicon level is low) after injection of recombinant homodimers. Up-regulation of AMP genes by the homodimers was accompanied by reduced bacterial populations in fly assay preparations. The induction of AMP expression is via activation of the NF-κB transcription factor Relish in the immune deficiency (Imd) pathway. The influence of bursicon homodimers on immune function does not appear to act through the heterodimer receptor DLGR2, i.e. novel receptors exist for the homodimers. Conclusions/Significance Our results reveal a mechanism of CNS-regulated prophylactic innate immunity during molting via induced expression of genes encoding AMPs and genes of the Turandot family. Turandot genes are also up-regulated by a broader range of extreme insults. From these data we infer that CNS-generated bursicon homodimers mediate innate prophylactic immunity to both stress and infection during the vulnerable molting cycle.
Collapse
Affiliation(s)
- Shiheng An
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Shengzhang Dong
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Qian Wang
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Sheng Li
- Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Lawrence I. Gilbert
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - David Stanley
- USDA/Agricultural Research Service, Biological Control of Insects Research Laboratory, Columbia, Missouri, United States of America
| | - Qisheng Song
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
43
|
Hughes AL. Amino acid sequence coevolution in the insect bursicon ligand-receptor system. Mol Phylogenet Evol 2012; 63:617-24. [PMID: 22373512 DOI: 10.1016/j.ympev.2012.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 02/01/2012] [Accepted: 02/07/2012] [Indexed: 11/24/2022]
Abstract
The pattern of amino acid residue replacement in the components of the bursicon signaling system (involving the BURSα/BURSβ heterodimer and its receptor BURSrec) was reconstructed across a phylogeny of 17 insect species, in order to test for the co-occurrence of replacements at sets of individual sites. Sets of three or more branches with perfectly concordant changes occurred to a greater extent than expected by chance, given the observed level of amino acid change. The latter sites (SPC sites) were found to have distinctive characteristics: (1) the mean number of changes was significantly lower at SPC sites than that at other sites with multiple changes; (2) SPC sites had a significantly greater tendency toward parallel amino acid changes than other sites with multiple changes, but no greater tendency toward convergent changes; and (3) parallel changes tended to involve relatively similar amino acids, as indicated by relatively low mean chemical distances. The results implicated functional constraint, permitting only a limited subset of amino acids in a given site, as a major factor in causing both parallel amino acid replacement and coordinated amino acid changes in different sites of the same protein and of interacting proteins in this system.
Collapse
Affiliation(s)
- Austin L Hughes
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29205, USA.
| |
Collapse
|
44
|
Caers J, Verlinden H, Zels S, Vandersmissen HP, Vuerinckx K, Schoofs L. More than two decades of research on insect neuropeptide GPCRs: an overview. Front Endocrinol (Lausanne) 2012; 3:151. [PMID: 23226142 PMCID: PMC3510462 DOI: 10.3389/fendo.2012.00151] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/14/2012] [Indexed: 11/30/2022] Open
Abstract
This review focuses on the state of the art on neuropeptide receptors in insects. Most of these receptors are G protein-coupled receptors (GPCRs) and are involved in the regulation of virtually all physiological processes during an insect's life. More than 20 years ago a milestone in invertebrate endocrinology was achieved with the characterization of the first insect neuropeptide receptor, i.e., the Drosophila tachykinin-like receptor. However, it took until the release of the Drosophila genome in 2000 that research on neuropeptide receptors boosted. In the last decade a plethora of genomic information of other insect species also became available, leading to a better insight in the functions and evolution of the neuropeptide signaling systems and their intracellular pathways. It became clear that some of these systems are conserved among all insect species, indicating that they fulfill crucial roles in their physiological processes. Meanwhile, other signaling systems seem to be lost in several insect orders or species, suggesting that their actions were superfluous in those insects, or that other neuropeptides have taken over their functions. It is striking that the deorphanization of neuropeptide GPCRs gets much attention, but the subsequent unraveling of the intracellular pathways they elicit, or their physiological functions are often hardly examined. Especially in insects besides Drosophila this information is scarce if not absent. And although great progress made in characterizing neuropeptide signaling systems, even in Drosophila several predicted neuropeptide receptors remain orphan, awaiting for their endogenous ligand to be determined. The present review gives a précis of the insect neuropeptide receptor research of the last two decades. But it has to be emphasized that the work done so far is only the tip of the iceberg and our comprehensive understanding of these important signaling systems will still increase substantially in the coming years.
Collapse
Affiliation(s)
| | | | | | | | | | - Liliane Schoofs
- *Correspondence: Liliane Schoofs, Department of Biology, Research Group of Functional Genomics and Proteomics, Naamsestraat 59, KU Leuven, 3000 Leuven, Belgium. e-mail:
| |
Collapse
|
45
|
Gatto CL, Broadie K. Fragile X mental retardation protein is required for programmed cell death and clearance of developmentally-transient peptidergic neurons. Dev Biol 2011; 356:291-307. [PMID: 21596027 PMCID: PMC3143227 DOI: 10.1016/j.ydbio.2011.05.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 04/22/2011] [Accepted: 05/03/2011] [Indexed: 01/19/2023]
Abstract
Fragile X syndrome (FXS), caused by loss of fragile X mental retardation 1 (FMR1) gene function, is the most common heritable cause of intellectual disability and autism spectrum disorders. The FMR1 product (FMRP) is an RNA-binding protein best established to function in activity-dependent modulation of synaptic connections. In the Drosophila FXS disease model, loss of functionally-conserved dFMRP causes synaptic overgrowth and overelaboration in pigment dispersing factor (PDF) peptidergic neurons in the adult brain. Here, we identify a very different component of PDF neuron misregulation in dfmr1 mutants: the aberrant retention of normally developmentally-transient PDF tritocerebral (PDF-TRI) neurons. In wild-type animals, PDF-TRI neurons in the central brain undergo programmed cell death and complete, processive clearance within days of eclosion. In the absence of dFMRP, a defective apoptotic program leads to constitutive maintenance of these peptidergic neurons. We tested whether this apoptotic defect is circuit-specific by examining crustacean cardioactive peptide (CCAP) and bursicon circuits, which are similarly developmentally-transient and normally eliminated immediately post-eclosion. In dfmr1 null mutants, CCAP/bursicon neurons also exhibit significantly delayed clearance dynamics, but are subsequently eliminated from the nervous system, in contrast to the fully persistent PDF-TRI neurons. Thus, the requirement of dFMRP for the retention of transitory peptidergic neurons shows evident circuit specificity. The novel defect of impaired apoptosis and aberrant neuron persistence in the Drosophila FXS model suggests an entirely new level of "pruning" dysfunction may contribute to the FXS disease state.
Collapse
Affiliation(s)
- Cheryl L Gatto
- Departments of Biological Sciences and Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | |
Collapse
|
46
|
Bai H, Zhu F, Shah K, Palli SR. Large-scale RNAi screen of G protein-coupled receptors involved in larval growth, molting and metamorphosis in the red flour beetle. BMC Genomics 2011; 12:388. [PMID: 21806814 PMCID: PMC3163568 DOI: 10.1186/1471-2164-12-388] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Accepted: 08/01/2011] [Indexed: 02/08/2023] Open
Abstract
Background The G protein-coupled receptors (GPCRs) belong to the largest superfamily of integral cell membrane proteins and play crucial roles in physiological processes including behavior, development and reproduction. Because of their broad and diverse roles in cellular signaling, GPCRs are the therapeutic targets for many prescription drugs. However, there is no commercial pesticide targeting insect GPCRs. In this study, we employed functional genomics methods and used the red flour beetle, Tribolium castaneum, as a model system to study the physiological roles of GPCRs during the larval growth, molting and metamorphosis. Results A total of 111 non-sensory GPCRs were identified in the T. castaneum genome. Thirty-nine of them were not reported previously. Large-scale RNA interference (RNAi) screen was used to study the function of all these GPCRs during immature stages. Double-stranded RNA (dsRNA)-mediated knockdown in the expression of genes coding for eight GPCRs caused severe developmental arrest and ecdysis failure (with more than 90% mortality after dsRNA injection). These GPCRs include dopamine-2 like receptor (TC007490/D2R) and latrophilin receptor (TC001872/Cirl). The majority of larvae injected with TC007490/D2R dsRNA died during larval stage prior to entering pupal stage, suggesting that this GPCR is essential for larval growth and development. Conclusions The results from our study revealed the physiological roles of some GPCRs in T. castaneum. These findings could help in development of novel pesticides targeting these GPCRs.
Collapse
Affiliation(s)
- Hua Bai
- Department of Entomology, S-225 Agriculture Science Bldg, N,, University of Kentucky, Lexington, KY 40546, USA
| | | | | | | |
Collapse
|
47
|
Honegger HW, Estévez-Lao TY, Hillyer JF. Bursicon-expressing neurons undergo apoptosis after adult ecdysis in the mosquito Anopheles gambiae. JOURNAL OF INSECT PHYSIOLOGY 2011; 57:1017-1022. [PMID: 21554887 DOI: 10.1016/j.jinsphys.2011.04.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 04/22/2011] [Accepted: 04/25/2011] [Indexed: 05/30/2023]
Abstract
Neuropeptides are important regulators of diverse processes during development. The insect neuropeptide bursicon, a 30 kDa heterodimer, controls the hardening of the new cuticle after the shedding of the old one (ecdysis) and the inflation and maturation of adult wings. Given this specific functional role, its expression should only be required transiently because adult insects no longer undergo ecdysis. Here we report the transient expression of bursicon in the mosquito, Anopheles gambiae. Quantitative RT-PCR revealed that transcription of the bursicon monomers, burs and pburs, steadily increases through the larval stages, peaks in the black pupa stage, and decreases to below detectable levels by 8 h after adult ecdysis (eclosion). Immunohistochemistry on the adult nervous system showed that bursicon is co-expressed with crustacean cardioactive peptide (CCAP) in specific neurons of the abdominal ganglia, but that labeling intensity wanes by 14 h post-eclosion. Finally, detection of disintegrating DNA by TUNEL labeling demonstrated that the bursicon expressing neurons successively undergo apoptosis following eclosion. Taken altogether, these data describe A. gambiae as another holometabolous insect in which bursicon ceases to be produced in adults, and in which the bursicon expressing neurons are removed from the ventral nerve cord.
Collapse
Affiliation(s)
- Hans-Willi Honegger
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | | | | |
Collapse
|
48
|
Talmat-Amar Y, Arribat Y, Redt-Clouet C, Feuillette S, Bougé AL, Lecourtois M, Parmentier ML. Important neuronal toxicity of microtubule-bound Tau in vivo in Drosophila. Hum Mol Genet 2011; 20:3738-45. [DOI: 10.1093/hmg/ddr290] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
49
|
Synaptic neuropeptide release induced by octopamine without Ca2+ entry into the nerve terminal. Proc Natl Acad Sci U S A 2011; 108:4477-81. [PMID: 21368121 DOI: 10.1073/pnas.1017837108] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Synaptic release of neurotransmitters is evoked by activity-dependent Ca(2+) entry into the nerve terminal. However, here it is shown that robust synaptic neuropeptide release from Drosophila motoneurons is evoked in the absence of extracellular Ca(2+) by octopamine, the arthropod homolog to norepinephrine. Genetic and pharmacology experiments demonstrate that this surprising peptidergic transmission requires cAMP-dependent protein kinase, with only a minor contribution of exchange protein activated by cAMP (epac). Octopamine-evoked neuropeptide release also requires endoplasmic reticulum Ca(2+) mobilization by the ryanodine receptor and the inositol trisphosphate receptor. Hence, rather than relying exclusively on activity-dependent Ca(2+) entry into the nerve terminal, a behaviorally important neuromodulator uses synergistic cAMP-dependent protein kinase and endoplasmic reticulum Ca(2+) signaling to induce synaptic neuropeptide release.
Collapse
|