1
|
Li XC, Srinivasan V, Laiker I, Misunou N, Frankel N, Pallares LF, Crocker J. TF-High-Evolutionary: In Vivo Mutagenesis of Gene Regulatory Networks for the Study of the Genetics and Evolution of the Drosophila Regulatory Genome. Mol Biol Evol 2024; 41:msae167. [PMID: 39117360 PMCID: PMC11342961 DOI: 10.1093/molbev/msae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/10/2024] Open
Abstract
Understanding the evolutionary potential of mutations in gene regulatory networks is essential to furthering the study of evolution and development. However, in multicellular systems, genetic manipulation of regulatory networks in a targeted and high-throughput way remains challenging. In this study, we designed TF-High-Evolutionary (HighEvo), a transcription factor (TF) fused with a base editor (activation-induced deaminase), to continuously induce germline mutations at TF-binding sites across regulatory networks in Drosophila. Populations of flies expressing TF-HighEvo in their germlines accumulated mutations at rates an order of magnitude higher than natural populations. Importantly, these mutations accumulated around the targeted TF-binding sites across the genome, leading to distinct morphological phenotypes consistent with the developmental roles of the tagged TFs. As such, this TF-HighEvo method allows the interrogation of the mutational space of gene regulatory networks at scale and can serve as a powerful reagent for experimental evolution and genetic screens focused on the regulatory genome.
Collapse
Affiliation(s)
- Xueying C Li
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Ian Laiker
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Universidad de Buenos Aires (UBA), Buenos Aires 1428, Argentina
| | | | - Nicolás Frankel
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Universidad de Buenos Aires (UBA), Buenos Aires 1428, Argentina
| | - Luisa F Pallares
- Friedrich Miescher Laboratory, Max Planck Society, Tübingen, Germany
| | - Justin Crocker
- European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
2
|
Genome-wide analysis identifies Homothorax and Extradenticle as regulators of insulin in Drosophila Insulin-Producing cells. PLoS Genet 2022; 18:e1010380. [PMID: 36095003 PMCID: PMC9499297 DOI: 10.1371/journal.pgen.1010380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 09/22/2022] [Accepted: 08/15/2022] [Indexed: 11/19/2022] Open
Abstract
Drosophila Insulin-Producing Cells (IPCs) are the main production site of the Drosophila Insulin-like peptides or dilps which have key roles in regulating growth, development, reproduction, lifespan and metabolism. To better understand the signalling pathways and transcriptional networks that are active in the IPCs we queried publicly available transcriptome data of over 180 highly inbred fly lines for dilp expression and used dilp expression as the input for a Genome-wide association study (GWAS). This resulted in the identification of variants in 125 genes that were associated with variation in dilp expression. The function of 57 of these genes in the IPCs was tested using an RNAi-based approach. We found that IPC-specific depletion of most genes resulted in differences in expression of one or more of the dilps. We then elaborated further on one of the candidate genes with the strongest effect on dilp expression, Homothorax, a transcription factor known for its role in eye development. We found that Homothorax and its binding partner Extradenticle are involved in regulating dilp2, -3 and -5 expression and that genetic depletion of both TFs shows phenotypes associated with reduced insulin signalling. Furthermore, we provide evidence that other transcription factors involved in eye development are also functional in the IPCs. In conclusion, we showed that this expression level-based GWAS approach identified genetic regulators implicated in IPC function and dilp expression. Insulin signalling has a central and evolutionarily conserved role in many processes including growth, development, reproduction, lifespan, stress resistance and metabolic homeostasis. In the fruitfly Drosophila melanogaster insulin-producing cells in the brain are the main source of three insulin-like peptides, Dilp2, -3 and -5. How the production and secretion of these three insulin-like peptides are regulated remains incompletely understood. In the current study, genome-wide association studies were used to identify 50 novel regulators of Dilp2, -3 and -5. We show that one of the top candidate regulators, Homothorax, is an important regulator of dilp2, -3 and –5 expression in the IPCs and is necessary for normal systemic insulin signalling and regulates adult size and developmental timing. We also show that the Hth interactor Extradenticle (Exd) is equally required in the adult but not in the larval IPCs. Finally, we show that most genes of the so-called retinal determination gene network are expressed in the IPCs and regulate normal dilp2 and -5 expression. Together, these results identify further regulatory levels active in the IPCs and implicate a reshuffled version of a previously identified gene regulatory network therein.
Collapse
|
3
|
Li Q, Fu Y, Liu X, Sun J, Hou M, Zhang Y, Chen J. Activation of Wheat Defense Response by Buchnera aphidicola-Derived Small Chaperone Protein GroES in Wheat Aphid Saliva. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1058-1067. [PMID: 35076234 DOI: 10.1021/acs.jafc.1c07046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Salivary proteins secreted by aphids during feeding play an important role in regulating the plant defense response. We used mass spectrometry to identify 155 proteins from the wheat aphid, Sitobion miscanthi, among which 44 proteins were derived from the primary symbiont, Buchnera aphidicola. GroES, which is a highly abundant molecular chaperone that binds to GroEL, was detected in saliva. In vitro injection of purified GroES protein and overexpression of GroES in wheat leaves verified that GroES induced hydrogen peroxide accumulation and callose deposition in wheat and further activated the plant salic acid and jasmonic acid defense pathways. Our findings indicate that plants may have evolved new strategies to detect aphid attack and trigger defense responses by recognizing proteins derived from B. aphidicola, which is present in almost all aphid species.
Collapse
Affiliation(s)
- Qian Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Yu Fu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Xiaobei Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Jingxuan Sun
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Maolin Hou
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Yong Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Julian Chen
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
- MARA-CABI Joint Laboratory for Bio-Safety, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| |
Collapse
|
4
|
Huygens C, Ribeiro Lopes M, Gaget K, Duport G, Peignier S, De Groef S, Parisot N, Calevro F, Callaerts P. Evolutionary diversification of insulin-related peptides (IRPs) in aphids and spatiotemporal distribution in Acyrthosiphon pisum. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 141:103670. [PMID: 34666188 DOI: 10.1016/j.ibmb.2021.103670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 06/13/2023]
Abstract
Members of the insulin superfamily activate the evolutionarily highly conserved insulin/insulin-like growth factor signaling pathway, involved in regulation of growth, energy homeostasis, and longevity. In the current study we focus on aphids to gain more insight into the evolution of the IRPs and how they may contribute to regulation of the insulin-signaling pathway. Using the latest annotation of the pea aphid (Acyrthosiphon pisum) genome, and combining sequence alignments and phylogenetic analyses, we identified seven putative IRP encoding-genes, with IRP1-IRP4 resembling the classical insulin and insulin-like protein structures, and IRP5 and IRP6 bearing insulin-like growth factor (IGF) features. We also identified IRP11 as a new and structurally divergent IRP present in at least eight aphid genomes. Globally the ten aphid genomes analyzed in this work contain four to 15 IRPs, while only three IRPs were found in the genome of the grape phylloxera, a hemipteran insect representing an earlier evolutionary branch of the aphid group. Expression analyses revealed spatial and temporal variation in the expression patterns of the different A. pisum IRPs. IRP1 and IRP4 are expressed throughout all developmental stages and morphs in neuroendocrine cells of the brain, while IRP5 and IRP6 are expressed in the fat body. IRP2 is expressed in specific cells of the gut in aphids in non-crowded conditions and in the head of aphids under crowded conditions, IRP3 in salivary glands, and both IRP2 and IRP3 in the male morph. IRP11 expression is enriched in the carcass. This complex spatiotemporal expression pattern suggests functional diversification of the IRPs.
Collapse
Affiliation(s)
- C Huygens
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KULeuven, University of Leuven, B-3000, Leuven, Belgium; Univ Lyon, INSA Lyon, INRAE, BF2I, UMR 203, 69621, Villeurbanne, France
| | - M Ribeiro Lopes
- Univ Lyon, INSA Lyon, INRAE, BF2I, UMR 203, 69621, Villeurbanne, France
| | - K Gaget
- Univ Lyon, INSA Lyon, INRAE, BF2I, UMR 203, 69621, Villeurbanne, France
| | - G Duport
- Univ Lyon, INSA Lyon, INRAE, BF2I, UMR 203, 69621, Villeurbanne, France
| | - S Peignier
- Univ Lyon, INSA Lyon, INRAE, BF2I, UMR 203, 69621, Villeurbanne, France
| | - S De Groef
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KULeuven, University of Leuven, B-3000, Leuven, Belgium
| | - N Parisot
- Univ Lyon, INSA Lyon, INRAE, BF2I, UMR 203, 69621, Villeurbanne, France
| | - F Calevro
- Univ Lyon, INSA Lyon, INRAE, BF2I, UMR 203, 69621, Villeurbanne, France.
| | - P Callaerts
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KULeuven, University of Leuven, B-3000, Leuven, Belgium.
| |
Collapse
|
5
|
Kim SK, Tsao DD, Suh GSB, Miguel-Aliaga I. Discovering signaling mechanisms governing metabolism and metabolic diseases with Drosophila. Cell Metab 2021; 33:1279-1292. [PMID: 34139200 PMCID: PMC8612010 DOI: 10.1016/j.cmet.2021.05.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/30/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022]
Abstract
There has been rapid growth in the use of Drosophila and other invertebrate systems to dissect mechanisms governing metabolism. New assays and approaches to physiology have aligned with superlative genetic tools in fruit flies to provide a powerful platform for posing new questions, or dissecting classical problems in metabolism and disease genetics. In multiple examples, these discoveries exploit experimental advantages as-yet unavailable in mammalian systems. Here, we illustrate how fly studies have addressed long-standing questions in three broad areas-inter-organ signaling through hormonal or neural mechanisms governing metabolism, intestinal interoception and feeding, and the cellular and signaling basis of sexually dimorphic metabolism and physiology-and how these findings relate to human (patho)physiology. The imaginative application of integrative physiology and related approaches in flies to questions in metabolism is expanding, and will be an engine of discovery, revealing paradigmatic features of metabolism underlying human diseases and physiological equipoise in health.
Collapse
Affiliation(s)
- Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine (Endocrinology), Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Deborah D Tsao
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Greg S B Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea.
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
6
|
Evidence from oyster suggests an ancient role for Pdx in regulating insulin gene expression in animals. Nat Commun 2021; 12:3117. [PMID: 34035261 PMCID: PMC8149454 DOI: 10.1038/s41467-021-23216-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 04/19/2021] [Indexed: 11/17/2022] Open
Abstract
Hox and ParaHox genes encode transcription factors with similar expression patterns in divergent animals. The Pdx (Xlox) homeobox gene, for example, is expressed in a sharp spatial domain in the endodermal cell layer of the gut in chordates, echinoderms, annelids and molluscs. The significance of comparable gene expression patterns is unclear because it is not known if downstream transcriptional targets are also conserved. Here, we report evidence indicating that a classic transcriptional target of Pdx1 in vertebrates, the insulin gene, is a likely direct target of Pdx in Pacific oyster adults. We show that one insulin-related gene, cgILP, is co-expressed with cgPdx in oyster digestive tissue. Transcriptomic comparison suggests that this tissue plays a similar role to the vertebrate pancreas. Using ATAC-seq and ChIP, we identify an upstream regulatory element of the cgILP gene which shows binding interaction with cgPdx protein in oyster hepatopancreas and demonstrate, using a cell culture assay, that the oyster Pdx can act as a transcriptional activator through this site, possibly in synergy with NeuroD. These data argue that a classic homeodomain-target gene interaction dates back to the origin of Bilateria. In vertebrates insulin is a direct transcriptional target of Pdx: the same is true in Pacific oysters and the authors show insulin-related gene, cgILP, is co-expressed with cgPdx in oyster digestive tissue, showing this gene interaction dates back to the origin of Bilateria.
Collapse
|
7
|
Yang L, Webb SE, Jin N, Lee HM, Chan TF, Xu G, Chan JC, Miller AL, Ma RC. Investigating the role of dachshund b in the development of the pancreatic islet in zebrafish. J Diabetes Investig 2021; 12:710-727. [PMID: 33449448 PMCID: PMC8089008 DOI: 10.1111/jdi.13503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/28/2020] [Accepted: 01/01/2021] [Indexed: 12/29/2022] Open
Abstract
Aims/Introduction β‐Cell dysfunction is a hallmark of type 2 diabetes. In a previous pilot study, we identified an association between genetic variants within the human DACH1 gene and young‐onset type 2 diabetes. Here, we characterized the function of dachb, the only dach homologue to be expressed in the pancreas, in developing zebrafish embryos. Materials and Methods We injected one‐cell stage embryos with a dachb‐morpholino (MO) or with the dachb‐MO and dachb messenger ribonucleic acid, and determined the effect on the development of the pancreatic islet. We also carried out quantitative polymerase chain reaction and ribonucleic acid sequencing on the dachb‐MO group to determine the effect of dachb knockdown on gene expression. Results MO‐mediated dachb knockdown resulted in impaired islet cell development, with a significant decrease in both the β‐cell and islet cell numbers. This islet developmental defect was rescued when embryos were co‐injected with dachb‐MO and dachb messenger ribonucleic acid. Knockdown of dachb was associated with a significant downregulation of the β‐cell specific marker gene, insa, and the somatostatin cell marker, sst2, as well as regulators of pancreas development, ptf1a, neuroD, pax6a and nkx6.1, and the cell cycle gene, insm1a. Furthermore, ribonucleic sequencing analysis showed an upregulation of genes enriched in the forkhead box O and mitogen‐activated protein kinase signaling pathways in the dachb‐MO group, when compared with the control groups. Conclusions Together, our results suggest the possible role of dachb in islet development in zebrafish.
Collapse
Affiliation(s)
- Lingling Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Sarah E Webb
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Nana Jin
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Heung Man Lee
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong
| | - Ting Fung Chan
- School of Life Sciences and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Gang Xu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Teaching and Research Division, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Juliana Cn Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Andrew L Miller
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Ronald Cw Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
8
|
Clements J, Buhler K, Winant M, Vulsteke V, Callaerts P. Glial and Neuronal Neuroglian, Semaphorin-1a and Plexin A Regulate Morphological and Functional Differentiation of Drosophila Insulin-Producing Cells. Front Endocrinol (Lausanne) 2021; 12:600251. [PMID: 34276554 PMCID: PMC8281472 DOI: 10.3389/fendo.2021.600251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 06/11/2021] [Indexed: 11/21/2022] Open
Abstract
The insulin-producing cells (IPCs), a group of 14 neurons in the Drosophila brain, regulate numerous processes, including energy homeostasis, lifespan, stress response, fecundity, and various behaviors, such as foraging and sleep. Despite their importance, little is known about the development and the factors that regulate morphological and functional differentiation of IPCs. In this study, we describe the use of a new transgenic reporter to characterize the role of the Drosophila L1-CAM homolog Neuroglian (Nrg), and the transmembrane Semaphorin-1a (Sema-1a) and its receptor Plexin A (PlexA) in the differentiation of the insulin-producing neurons. Loss of Nrg results in defasciculation and abnormal neurite branching, including ectopic neurites in the IPC neurons. Cell-type specific RNAi knockdown experiments reveal that Nrg, Sema-1a and PlexA are required in IPCs and glia to control normal morphological differentiation of IPCs albeit with a stronger contribution of Nrg and Sema-1a in glia and of PlexA in the IPCs. These observations provide new insights into the development of the IPC neurons and identify a novel role for Sema-1a in glia. In addition, we show that Nrg, Sema-1a and PlexA in glia and IPCs not only regulate morphological but also functional differentiation of the IPCs and that the functional deficits are likely independent of the morphological phenotypes. The requirements of nrg, Sema-1a, and PlexA in IPC development and the expression of their vertebrate counterparts in the hypothalamic-pituitary axis, suggest that these functions may be evolutionarily conserved in the establishment of vertebrate endocrine systems.
Collapse
|
9
|
Nässel DR, Zandawala M. Hormonal axes in Drosophila: regulation of hormone release and multiplicity of actions. Cell Tissue Res 2020; 382:233-266. [PMID: 32827072 PMCID: PMC7584566 DOI: 10.1007/s00441-020-03264-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Hormones regulate development, as well as many vital processes in the daily life of an animal. Many of these hormones are peptides that act at a higher hierarchical level in the animal with roles as organizers that globally orchestrate metabolism, physiology and behavior. Peptide hormones can act on multiple peripheral targets and simultaneously convey basal states, such as metabolic status and sleep-awake or arousal across many central neuronal circuits. Thereby, they coordinate responses to changing internal and external environments. The activity of neurosecretory cells is controlled either by (1) cell autonomous sensors, or (2) by other neurons that relay signals from sensors in peripheral tissues and (3) by feedback from target cells. Thus, a hormonal signaling axis commonly comprises several components. In mammals and other vertebrates, several hormonal axes are known, such as the hypothalamic-pituitary-gonad axis or the hypothalamic-pituitary-thyroid axis that regulate reproduction and metabolism, respectively. It has been proposed that the basic organization of such hormonal axes is evolutionarily old and that cellular homologs of the hypothalamic-pituitary system can be found for instance in insects. To obtain an appreciation of the similarities between insect and vertebrate neurosecretory axes, we review the organization of neurosecretory cell systems in Drosophila. Our review outlines the major peptidergic hormonal pathways known in Drosophila and presents a set of schemes of hormonal axes and orchestrating peptidergic systems. The detailed organization of the larval and adult Drosophila neurosecretory systems displays only very basic similarities to those in other arthropods and vertebrates.
Collapse
Affiliation(s)
- Dick R. Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Meet Zandawala
- Department of Neuroscience, Brown University, Providence, RI USA
| |
Collapse
|
10
|
Manière G, Alves G, Berthelot-Grosjean M, Grosjean Y. Growth regulation by amino acid transporters in Drosophila larvae. Cell Mol Life Sci 2020; 77:4289-4297. [PMID: 32358623 PMCID: PMC7588360 DOI: 10.1007/s00018-020-03535-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 03/27/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022]
Abstract
Drosophila larvae need to adapt their metabolism to reach a critical body size to pupate. This process needs food resources and has to be tightly adjusted to control metamorphosis timing and adult size. Nutrients such as amino acids either directly present in the food or obtained via protein digestion play key regulatory roles in controlling metabolism and growth. Amino acids act especially on two organs, the fat body and the brain, to control larval growth, body size developmental timing and pupariation. The expression of specific amino acid transporters in fat body cells, and in the brain through specific neurons and glial cells is essential to activate downstream molecular signaling pathways in response to amino acid levels. In this review, we highlight some of these specific networks dependent on amino acid diet to control DILP levels, and by consequence larval metabolism and growth.
Collapse
Affiliation(s)
- Gérard Manière
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000, Dijon, France.
| | - Georges Alves
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Martine Berthelot-Grosjean
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000, Dijon, France
| | - Yael Grosjean
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Université Bourgogne Franche-Comté, 21000, Dijon, France.
| |
Collapse
|
11
|
Koyama T, Texada MJ, Halberg KA, Rewitz K. Metabolism and growth adaptation to environmental conditions in Drosophila. Cell Mol Life Sci 2020; 77:4523-4551. [PMID: 32448994 PMCID: PMC7599194 DOI: 10.1007/s00018-020-03547-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/19/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Organisms adapt to changing environments by adjusting their development, metabolism, and behavior to improve their chances of survival and reproduction. To achieve such flexibility, organisms must be able to sense and respond to changes in external environmental conditions and their internal state. Metabolic adaptation in response to altered nutrient availability is key to maintaining energy homeostasis and sustaining developmental growth. Furthermore, environmental variables exert major influences on growth and final adult body size in animals. This developmental plasticity depends on adaptive responses to internal state and external cues that are essential for developmental processes. Genetic studies have shown that the fruit fly Drosophila, similarly to mammals, regulates its metabolism, growth, and behavior in response to the environment through several key hormones including insulin, peptides with glucagon-like function, and steroid hormones. Here we review emerging evidence showing that various environmental cues and internal conditions are sensed in different organs that, via inter-organ communication, relay information to neuroendocrine centers that control insulin and steroid signaling. This review focuses on endocrine regulation of development, metabolism, and behavior in Drosophila, highlighting recent advances in the role of the neuroendocrine system as a signaling hub that integrates environmental inputs and drives adaptive responses.
Collapse
Affiliation(s)
- Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth A Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Texada MJ, Koyama T, Rewitz K. Regulation of Body Size and Growth Control. Genetics 2020; 216:269-313. [PMID: 33023929 PMCID: PMC7536854 DOI: 10.1534/genetics.120.303095] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
The control of body and organ growth is essential for the development of adults with proper size and proportions, which is important for survival and reproduction. In animals, adult body size is determined by the rate and duration of juvenile growth, which are influenced by the environment. In nutrient-scarce environments in which more time is needed for growth, the juvenile growth period can be extended by delaying maturation, whereas juvenile development is rapidly completed in nutrient-rich conditions. This flexibility requires the integration of environmental cues with developmental signals that govern internal checkpoints to ensure that maturation does not begin until sufficient tissue growth has occurred to reach a proper adult size. The Target of Rapamycin (TOR) pathway is the primary cell-autonomous nutrient sensor, while circulating hormones such as steroids and insulin-like growth factors are the main systemic regulators of growth and maturation in animals. We discuss recent findings in Drosophila melanogaster showing that cell-autonomous environment and growth-sensing mechanisms, involving TOR and other growth-regulatory pathways, that converge on insulin and steroid relay centers are responsible for adjusting systemic growth, and development, in response to external and internal conditions. In addition to this, proper organ growth is also monitored and coordinated with whole-body growth and the timing of maturation through modulation of steroid signaling. This coordination involves interorgan communication mediated by Drosophila insulin-like peptide 8 in response to tissue growth status. Together, these multiple nutritional and developmental cues feed into neuroendocrine hubs controlling insulin and steroid signaling, serving as checkpoints at which developmental progression toward maturation can be delayed. This review focuses on these mechanisms by which external and internal conditions can modulate developmental growth and ensure proper adult body size, and highlights the conserved architecture of this system, which has made Drosophila a prime model for understanding the coordination of growth and maturation in animals.
Collapse
Affiliation(s)
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, 2100, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100, Denmark
| |
Collapse
|
13
|
Agbu P, Cassidy JJ, Braverman J, Jacobson A, Carthew RW. MicroRNA miR-7 Regulates Secretion of Insulin-Like Peptides. Endocrinology 2020; 161:5686887. [PMID: 31875904 PMCID: PMC7029775 DOI: 10.1210/endocr/bqz040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/20/2019] [Indexed: 01/01/2023]
Abstract
The insulin/insulin-like growth factor (IGF) pathway is essential for linking nutritional status to growth and metabolism. MicroRNAs (miRNAs) are short RNAs that are players in the regulation of this process. The miRNA miR-7 shows highly conserved expression in insulin-producing cells across the animal kingdom. However, its conserved functions in regulation of insulin-like peptides (ILPs) remain unknown. Using Drosophila as a model, we demonstrate that miR-7 limits ILP availability by inhibiting its production and secretion. Increasing miR-7 alters body growth and metabolism in an ILP-dependent manner, elevating circulating sugars and total body triglycerides, while decreasing animal growth. These effects are not due to direct targeting of ILP mRNA, but instead arise through alternate targets that affect the function of ILP-producing cells. The Drosophila F-actin capping protein alpha (CPA) is a direct target of miR-7, and knockdown of CPA in insulin-producing cells phenocopies the effects of miR-7 on ILP secretion. This regulation of CPA is conserved in mammals, with the mouse ortholog Capza1 also targeted by miR-7 in β-islet cells. Taken together, these results support a role for miR-7 regulation of an actin capping protein in insulin regulation, and highlight a conserved mechanism of action for an evolutionarily ancient microRNA.
Collapse
Affiliation(s)
- Pamela Agbu
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois
| | - Justin J Cassidy
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois
| | - Jonathan Braverman
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois
| | - Alec Jacobson
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois
| | - Richard W Carthew
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, Illinois
- Correspondence: Richard W. Carthew, PhD, Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208. E-mail:
| |
Collapse
|
14
|
Maier D, Nagel AC, Preiss A. Genetic interactions between Protein Kinase D and Lobe mutants during eye development of Drosophila melanogaster. Hereditas 2019; 156:37. [PMID: 31889943 PMCID: PMC6924039 DOI: 10.1186/s41065-019-0113-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Background In Drosophila, the development of the fly eye involves the activity of several, interconnected pathways that first define the presumptive eye field within the eye anlagen, followed by establishment of the dorso-ventral boundary, and the regulation of growth and apoptosis. In Lobe (L) mutant flies, parts of the eye or even the complete eye are absent because the eye field has not been properly defined. Manifold genetic interactions indicate that L influences the activity of several signalling pathways, resulting in a conversion of eye tissue into epidermis, and in the induction of apoptosis. As information on the molecular nature of the L mutation is lacking, the underlying molecular mechanisms are still an enigma. Results We have identified Protein Kinase D (PKD) as a strong modifier of the L mutant phenotype. PKD belongs to the PKC/CAMK class of Ser/Thr kinases that have been involved in diverse cellular processes including stress resistance and growth. Despite the many roles of PKD, Drosophila PKD null mutants are without apparent phenotype apart from sensitivity to oxidative stress. Here we report an involvement of PKD in eye development in the sensitized genetic background of Lobe. Absence of PKD strongly enhanced the dominant eye defects of heterozygous L2 flies, and decreased their viability. Moreover, eye-specific overexpression of an activated isoform of PKD considerably ameliorated the dominant L2 phenotype. This genetic interaction was not allele specific but similarly seen with three additional, weaker L alleles (L1, L5, LG), demonstrating its specificity. Conclusions We propose that PKD-mediated phosphorylation is involved in underlying processes causing the L phenotype, i.e. in the regulation of growth, the epidermal transformation of eye tissue and apoptosis, respectively.
Collapse
Affiliation(s)
- Dieter Maier
- Universität Hohenheim, Institut für Genetik (240A), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anja C Nagel
- Universität Hohenheim, Institut für Genetik (240A), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anette Preiss
- Universität Hohenheim, Institut für Genetik (240A), Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
15
|
Abstract
Beneficial symbiotic associations, ubiquitously found in nature, have led to the emergence of eukaryotic cells, the bacteriocytes, specialized in harboring microbial partners. One of the most fundamental questions concerning these enigmatic cells is how organismal homeostasis controls their elimination. Here we report that aphid bacteriocytes have evolved a form of cell death distinct from the conserved cell-death mechanisms hitherto characterized. This cell-death mechanism is a nonapoptotic multistep process that starts with the hypervacuolation of the endoplasmic reticulum, followed by a cascade of cellular stress responses. Our findings provide a framework to study biological functioning of bacteriocytes and the cellular mechanisms associated with symbiosis and contribute to the understanding of eukaryotic cell-death diversity. Symbiotic associations play a pivotal role in multicellular life by facilitating acquisition of new traits and expanding the ecological capabilities of organisms. In insects that are obligatorily dependent on intracellular bacterial symbionts, novel host cells (bacteriocytes) or organs (bacteriomes) have evolved for harboring beneficial microbial partners. The processes regulating the cellular life cycle of these endosymbiont-bearing cells, such as the cell-death mechanisms controlling their fate and elimination in response to host physiology, are fundamental questions in the biology of symbiosis. Here we report the discovery of a cell-death process involved in the degeneration of bacteriocytes in the hemipteran insect Acyrthosiphon pisum. This process is activated progressively throughout aphid adulthood and exhibits morphological features distinct from known cell-death pathways. By combining electron microscopy, immunohistochemistry, and molecular analyses, we demonstrated that the initial event of bacteriocyte cell death is the cytoplasmic accumulation of nonautophagic vacuoles, followed by a sequence of cellular stress responses including the formation of autophagosomes in intervacuolar spaces, activation of reactive oxygen species, and Buchnera endosymbiont degradation by the lysosomal system. We showed that this multistep cell-death process originates from the endoplasmic reticulum, an organelle exhibiting a unique reticular network organization spread throughout the entire cytoplasm and surrounding Buchnera aphidicola endosymbionts. Our findings provide insights into the cellular and molecular processes that coordinate eukaryotic host and endosymbiont homeostasis and death in a symbiotic system and shed light on previously unknown aspects of bacteriocyte biological functioning.
Collapse
|
16
|
Friedrich M. Ancient genetic redundancy of eyeless and twin of eyeless in the arthropod ocular segment. Dev Biol 2017; 432:192-200. [PMID: 28993201 DOI: 10.1016/j.ydbio.2017.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 01/28/2023]
Abstract
Pax6 transcription factors are essential upstream regulators in the developing anterior brain and peripheral visual system of most bilaterian animals. While a single homolog is in charge of these functions in vertebrates, two Pax6 genes are in Drosophila: eyeless (ey) and twin of eyeless (toy). At first glance, their co-existence seems sufficiently explained by their differential involvement in the specification of two types of insect visual organs: the lateral compound eyes (ey) and the dorsal ocelli (toy). Less straightforward to understand, however, is their genetic redundancy in promoting defined early and late growth phases of the precursor tissue to these organs: the eye-antennal imaginal disc. Drawing on comparative sequence, expression, and gene function evidence, I here conclude that this gene regulatory network module dates back to the dawn of arthropod evolution, securing the embryonic development of the ocular head segment. Thus, ey and toy constitute a paradigm to explore the organization and functional significance of longterm conserved genetic redundancy of duplicated genes. Indeed, as first steps in this direction, recent studies uncovered the shared use of binding sites in shared enhancers of target genes that are under redundant (string) and, strikingly, even subfunctionalized control by ey and toy (atonal). Equally significant, the evolutionarily recent and paralog-specific function of ey to repress the transcription of the antenna fate regulator Distal-less offers a functionally and phylogenetically well-defined opportunity to study the reconciliation of shared, partitioned, and newly acquired functions in a duplicated developmental gene pair.
Collapse
Affiliation(s)
- Markus Friedrich
- Department of Biological Sciences, Wayne State University, 5047 Gullen Mall, Detroit, MI 48202, USA; Department of Anatomy and Cell Biology, Wayne State University, School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201,USA.
| |
Collapse
|
17
|
Zwarts L, Vulsteke V, Buhl E, Hodge JJL, Callaerts P. SlgA, encoded by the homolog of the human schizophrenia-associated gene PRODH, acts in clock neurons to regulate Drosophila aggression. Dis Model Mech 2017; 10:705-716. [PMID: 28331058 PMCID: PMC5483002 DOI: 10.1242/dmm.027151] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 03/09/2017] [Indexed: 12/11/2022] Open
Abstract
Mutations in the proline dehydrogenase gene PRODH are linked to behavioral alterations in schizophrenia and as part of DiGeorge and velo-cardio-facial syndromes, but the role of PRODH in their etiology remains unclear. Here, we establish a Drosophila model to study the role of PRODH in behavioral disorders. We determine the distribution of the Drosophila PRODH homolog slgA in the brain and show that knockdown and overexpression of human PRODH and slgA in the lateral neurons ventral (LNv) lead to altered aggressive behavior. SlgA acts in an isoform-specific manner and is regulated by casein kinase II (CkII). Our data suggest that these effects are, at least partially, due to effects on mitochondrial function. We thus show that precise regulation of proline metabolism is essential to drive normal behavior and we identify Drosophila aggression as a model behavior relevant for the study of the mechanisms that are impaired in neuropsychiatric disorders. Editors' choice: A Drosophila model to study the role of PRODH, a schizophrenia-associated gene, in behavioral disorders.
Collapse
Affiliation(s)
- Liesbeth Zwarts
- KU Leuven - University of Leuven, Department of Human Genetics, Laboratory of Behavioral and Developmental Genetics, Leuven B-3000, Belgium.,VIB Center for the Biology of Disease, Laboratory of Behavioral and Developmental Genetics, Leuven B-3000, Belgium
| | - Veerle Vulsteke
- KU Leuven - University of Leuven, Department of Human Genetics, Laboratory of Behavioral and Developmental Genetics, Leuven B-3000, Belgium.,VIB Center for the Biology of Disease, Laboratory of Behavioral and Developmental Genetics, Leuven B-3000, Belgium
| | - Edgar Buhl
- University of Bristol, School of Physiology, Pharmacology and Neuroscience, Bristol BS8 1TD, UK
| | - James J L Hodge
- University of Bristol, School of Physiology, Pharmacology and Neuroscience, Bristol BS8 1TD, UK
| | - Patrick Callaerts
- KU Leuven - University of Leuven, Department of Human Genetics, Laboratory of Behavioral and Developmental Genetics, Leuven B-3000, Belgium .,VIB Center for the Biology of Disease, Laboratory of Behavioral and Developmental Genetics, Leuven B-3000, Belgium
| |
Collapse
|
18
|
Cattenoz PB, Giangrande A. Revisiting the role of the Gcm transcription factor, from master regulator to Swiss army knife. Fly (Austin) 2016; 10:210-8. [PMID: 27434165 DOI: 10.1080/19336934.2016.1212793] [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: 12/23/2022] Open
Abstract
Master genes are known to induce the differentiation of a multipotent cell into a specific cell type. These molecules are often transcription factors that switch on the regulatory cascade that triggers cell specification. Gcm was first described as the master gene of the glial fate in Drosophila as it induces the differentiation of neuroblasts into glia in the developing nervous system. Later on, Gcm was also shown to regulate the differentiation of blood, tendon and peritracheal cells as well as that of neuronal subsets. Thus, the glial master gene is used in at least 4 additional systems to promote differentiation. To understand the numerous roles of Gcm, we recently reported a genome-wide screen of Gcm direct targets in the Drosophila embryo. This screen provided new insight into the role and mode of action of this powerful transcription factor, notably on the interactions between Gcm and major differentiation pathways such as the Hedgehog, Notch and JAK/STAT. Here, we discuss the mode of action of Gcm in the different systems, we present new tissues that require Gcm and we revise the concept of 'master gene'.
Collapse
Affiliation(s)
- Pierre B Cattenoz
- a Department of Functional Genomics and Cancer , Institut de Génétique et de Biologie Moléculaire et Cellulaire , Illkirch , France
| | - Angela Giangrande
- a Department of Functional Genomics and Cancer , Institut de Génétique et de Biologie Moléculaire et Cellulaire , Illkirch , France
| |
Collapse
|
19
|
Nässel DR, Vanden Broeck J. Insulin/IGF signaling in Drosophila and other insects: factors that regulate production, release and post-release action of the insulin-like peptides. Cell Mol Life Sci 2016; 73:271-90. [PMID: 26472340 PMCID: PMC11108470 DOI: 10.1007/s00018-015-2063-3] [Citation(s) in RCA: 226] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 01/02/2023]
Abstract
Insulin, insulin-like growth factors (IGFs) and insulin-like peptides (ILPs) are important regulators of metabolism, growth, reproduction and lifespan, and mechanisms of insulin/IGF signaling (IIS) have been well conserved over evolution. In insects, between one and 38 ILPs have been identified in each species. Relatively few insect species have been investigated in depth with respect to ILP functions, and therefore we focus mainly on the well-studied fruitfly Drosophila melanogaster. In Drosophila eight ILPs (DILP1-8), but only two receptors (dInR and Lgr3) are known. DILP2, 3 and 5 are produced by a set of neurosecretory cells (IPCs) in the brain and their biosynthesis and release are controlled by a number of mechanisms differing between larvae and adults. Adult IPCs display cell-autonomous sensing of circulating glucose, coupled to evolutionarily conserved mechanisms for DILP release. The glucose-mediated DILP secretion is modulated by neurotransmitters and neuropeptides, as well as by factors released from the intestine and adipocytes. Larval IPCs, however, are indirectly regulated by glucose-sensing endocrine cells producing adipokinetic hormone, or by circulating factors from the intestine and fat body. Furthermore, IIS is situated within a complex physiological regulatory network that also encompasses the lipophilic hormones, 20-hydroxyecdysone and juvenile hormone. After release from IPCs, the ILP action can be modulated by circulating proteins that act either as protective carriers (binding proteins), or competitive inhibitors. Some of these proteins appear to have additional functions that are independent of ILPs. Taken together, the signaling with multiple ILPs is under complex control, ensuring tightly regulated IIS in the organism.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, 10691, Stockholm, Sweden.
| | - Jozef Vanden Broeck
- Molecular Developmental Physiology and Signal Transduction, Department of Animal Physiology and Neurobiology, Zoological Institute, K.U. Leuven, Louvain, Belgium
| |
Collapse
|
20
|
Okamoto N, Yamanaka N. Nutrition-dependent control of insect development by insulin-like peptides. CURRENT OPINION IN INSECT SCIENCE 2015; 11:21-30. [PMID: 26664828 PMCID: PMC4671074 DOI: 10.1016/j.cois.2015.08.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
In metazoans, members of the insulin-like peptide (ILP) family play a role in multiple physiological functions in response to the nutritional status. ILPs have been identified and characterized in a wide variety of insect species. Insect ILPs that are mainly produced by several pairs of medial neurosecretory cells in the brain circulate in the hemolymph and act systemically on target tissues. Physiological and biochemical studies in Lepidoptera and genetic studies in the fruit fly have greatly expanded our knowledge of the physiological functions of ILPs. Here, we outline the recent progress of the structural classification of insect ILPs and overview recent studies that have elucidated the physiological functions of insect ILPs involved in nutrient-dependent growth during development.
Collapse
Affiliation(s)
- Naoki Okamoto
- Department of Entomology, Institute for Integrative Genome Biology, Center for Disease Vector Research, University of California, Riverside, Riverside, CA 92521, USA
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, Center for Disease Vector Research, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
21
|
Nässel DR, Liu Y, Luo J. Insulin/IGF signaling and its regulation in Drosophila. Gen Comp Endocrinol 2015; 221:255-66. [PMID: 25616197 DOI: 10.1016/j.ygcen.2014.11.021] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 11/25/2014] [Accepted: 11/28/2014] [Indexed: 12/17/2022]
Abstract
Taking advantage of Drosophila as a genetically tractable experimental animal much progress has been made in our understanding of how the insulin/IGF signaling (IIS) pathway regulates development, growth, metabolism, stress responses and lifespan. The role of IIS in regulation of neuronal activity and behavior has also become apparent from experiments in Drosophila. This review briefly summarizes these functional roles of IIS, and also how the insulin producing cells (IPCs) are regulated in the fly. Furthermore, we discuss functional aspects of the spatio-temporal production of eight different insulin-like peptides (DILP1-8) that are thought to act on one known receptor (dInR) in Drosophila.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, S-10691 Stockholm, Sweden.
| | - Yiting Liu
- Department of Zoology, Stockholm University, S-10691 Stockholm, Sweden
| | - Jiangnan Luo
- Department of Zoology, Stockholm University, S-10691 Stockholm, Sweden
| |
Collapse
|
22
|
Sun J, Rockowitz S, Chauss D, Wang P, Kantorow M, Zheng D, Cvekl A. Chromatin features, RNA polymerase II and the comparative expression of lens genes encoding crystallins, transcription factors, and autophagy mediators. Mol Vis 2015; 21:955-73. [PMID: 26330747 PMCID: PMC4551281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/26/2015] [Indexed: 10/26/2022] Open
Abstract
PURPOSE Gene expression correlates with local chromatin structure. Our studies have mapped histone post-translational modifications, RNA polymerase II (pol II), and transcription factor Pax6 in lens chromatin. These data represent the first genome-wide insights into the relationship between lens chromatin structure and lens transcriptomes and serve as an excellent source for additional data analysis and refinement. The principal lens proteins, the crystallins, are encoded by predominantly expressed mRNAs; however, the regulatory mechanisms underlying their high expression in the lens remain poorly understood. METHODS The formaldehyde-assisted identification of regulatory regions (FAIRE-Seq) was employed to analyze newborn lens chromatin. ChIP-seq and RNA-seq data published earlier (GSE66961) have been used to assist in FAIRE-seq data interpretation. RNA transcriptomes from murine lens epithelium, lens fibers, erythrocytes, forebrain, liver, neurons, and pancreas were compared to establish the gene expression levels of the most abundant mRNAs versus median gene expression across other differentiated cells. RESULTS Normalized RNA expression data from multiple tissues show that crystallins rank among the most highly expressed genes in mammalian cells. These findings correlate with the extremely high abundance of pol II all across the crystallin loci, including crystallin genes clustered on chromosomes 1 and 5, as well as within regions of "open" chromatin, as identified by FAIRE-seq. The expression levels of mRNAs encoding DNA-binding transcription factors (e.g., Foxe3, Hsf4, Maf, Pax6, Prox1, Sox1, and Tfap2a) revealed that their transcripts form "clusters" of abundant mRNAs in either lens fibers or lens epithelium. The expression of three autophagy regulatory mRNAs, encoding Tfeb, FoxO1, and Hif1α, was found within a group of lens preferentially expressed transcription factors compared to the E12.5 forebrain. CONCLUSIONS This study reveals novel features of lens chromatin, including the remarkably high abundance of pol II at the crystallin loci that exhibit features of "open" chromatin. Hsf4 ranks among the most abundant fiber cell-preferred DNA-binding transcription factors. Notable transcripts, including Atf4, Ctcf, E2F4, Hey1, Hmgb1, Mycn, RXRβ, Smad4, Sp1, and Taf1 (transcription factors) and Ctsd, Gabarapl1, and Park7 (autophagy regulators) have been identified with high levels of expression in lens fibers, which suggests specific roles in lens fiber cell terminal differentiation.
Collapse
Affiliation(s)
- Jian Sun
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY,Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| | - Shira Rockowitz
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| | - Daniel Chauss
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL
| | - Ping Wang
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY
| | - Marc Kantorow
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY,Department of Neurology, Albert Einstein College of Medicine, Bronx, NY,Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY
| | - Ales Cvekl
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY,Department of Genetics, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
23
|
Kawasaki K, Yamada S, Ogata K, Saito Y, Takahama A, Yamada T, Matsumoto K, Kose H. Use of Drosophila as an evaluation method reveals imp as a candidate gene for type 2 diabetes in rat locus Niddm22. J Diabetes Res 2015; 2015:758564. [PMID: 25821834 PMCID: PMC4363715 DOI: 10.1155/2015/758564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 01/03/2015] [Accepted: 01/03/2015] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes (T2D) is one of the most common human diseases. QTL analysis of the diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats has identified numerous hyperglycemic loci. However, molecular characterization and/or gene identification largely remains to be elucidated due mostly to the weak genetic variances contributed by each locus. Here we utilized Drosophila melanogaster as a secondary model organism for functional evaluation of the candidate gene. We demonstrate that the tissue specific knockdown of a homologue of igf2bp2 RNA binding protein leads to increased sugar levels similar to that found in the OLETF rat. In the mutant, the expression of two of the insulin-like peptides encoded in the fly genome, dilp2 and dilp3, were found to be downregulated. Consistent with previous reports of dilp mutants, the imp mutant flies exhibited an extension of life span; in contrast, starvation tolerance was reduced. These results further reinforce the possibility that imp is involved in sugar metabolism by modulating insulin expression.
Collapse
Affiliation(s)
- Kurenai Kawasaki
- Division of Natural Sciences, Department of Life Science, International Christian University, Mitaka, Tokyo 181-8585, Japan
| | - Sawaka Yamada
- Division of Natural Sciences, Department of Life Science, International Christian University, Mitaka, Tokyo 181-8585, Japan
| | - Koki Ogata
- Division of Natural Sciences, Department of Life Science, International Christian University, Mitaka, Tokyo 181-8585, Japan
| | - Yumiko Saito
- Division of Natural Sciences, Department of Life Science, International Christian University, Mitaka, Tokyo 181-8585, Japan
| | - Aiko Takahama
- Division of Natural Sciences, Department of Life Science, International Christian University, Mitaka, Tokyo 181-8585, Japan
| | - Takahisa Yamada
- Laboratory of Animal Genetics, Graduate School of Science and Technology, Niigata University, Niigata 950-2181, Japan
| | - Kozo Matsumoto
- Department of Animal Medical Sciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Hiroyuki Kose
- Division of Natural Sciences, Department of Life Science, International Christian University, Mitaka, Tokyo 181-8585, Japan
- *Hiroyuki Kose:
| |
Collapse
|
24
|
Luan Q, Chen Q, Friedrich M. The Pax6 genes eyeless and twin of eyeless are required for global patterning of the ocular segment in the Tribolium embryo. Dev Biol 2014; 394:367-81. [PMID: 25149513 DOI: 10.1016/j.ydbio.2014.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 06/23/2014] [Accepted: 08/06/2014] [Indexed: 01/08/2023]
Abstract
The transcription factor gene Pax6 is widely considered a master regulator of eye development in bilaterian animals. However, the existence of visual organs that develop without Pax6 input and the considerable pleiotropy of Pax6 outside the visual system dictate further studies into defining ancestral functions of this important regulator. Previous work has shown that the combinatorial knockdown of the insect Pax6 orthologs eyeless (ey) and twin of eyeless (toy) perturbs the development of the visual system but also other areas of the larval head in the red flour beetle Tribolium castaneum. To elucidate the role of Pax6 during Tribolium head development in more detail, we studied head cuticle morphology, brain anatomy, embryonic head morphogenesis, and developmental marker gene expression in combinatorial ey and toy knockdown animals. Our experiments reveal that Pax6 is broadly required for patterning the anterior embryonic head. One of the earliest detectable roles is the formation of the embryonic head lobes, which originate from within the ocular segment and give rise to large parts of the supraesophageal brain including the mushroom body, a part of the posterior head capsule cuticle, and the visual system. We present further evidence that toy continues to be required for the development of the larval eyes after formation of the embryonic head lobes in cooperation with the eye developmental transcription factor dachshund (dac). The sum of our findings suggests that Pax6 functions as a competence factor throughout the development of the insect ocular segment. Comparative evidence identifies this function as an ancestral aspect of bilaterian head development.
Collapse
Affiliation(s)
- Qing Luan
- Department of Biological Sciences, Wayne State University, 5047 Gullen Mall, Detroit, MI 48202, USA; Institute of Molecular Biology and Department of Chemistry and Biochemistry, University of Oregon, Eugene, OR 97403, USA
| | - Qing Chen
- Department of Biological Sciences, Wayne State University, 5047 Gullen Mall, Detroit, MI 48202, USA
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, 5047 Gullen Mall, Detroit, MI 48202, USA; Department of Anatomy and Cell Biology, Wayne State University, School of Medicine, 540 East Canfield Avenue, Detroit, MI 48201, USA.
| |
Collapse
|
25
|
Neprilysins: an evolutionarily conserved family of metalloproteases that play important roles in reproduction in Drosophila. Genetics 2014; 196:781-97. [PMID: 24395329 DOI: 10.1534/genetics.113.160945] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Members of the M13 class of metalloproteases have been implicated in diseases and in reproductive fitness. Nevertheless, their physiological role remains poorly understood. To obtain a tractable model with which to analyze this protein family's function, we characterized the gene family in Drosophila melanogaster and focused on reproductive phenotypes. The D. melanogaster genome contains 24 M13 class protease homologs, some of which are orthologs of human proteases, including neprilysin. Many are expressed in the reproductive tracts of either sex. Using RNAi we individually targeted the five Nep genes most closely related to vertebrate neprilysin, Nep1-5, to investigate their roles in reproduction. A reduction in Nep1, Nep2, or Nep4 expression in females reduced egg laying. Nep1 and Nep2 are required in the CNS and the spermathecae for wild-type fecundity. Females that are null for Nep2 also show defects as hosts of sperm competition as well as an increased rate of depletion for stored sperm. Furthermore, eggs laid by Nep2 mutant females are fertilized normally, but arrest early in embryonic development. In the male, only Nep1 was required to induce normal patterns of female egg laying. Reduction in the expression of Nep2-5 in the male did not cause any dramatic effects on reproductive fitness, which suggests that these genes are either nonessential for male fertility or perform redundant functions. Our results suggest that, consistent with the functions of neprilysins in mammals, these proteins are also required for reproduction in Drosophila, opening up this model system for further functional analysis of this protein class and their substrates.
Collapse
|
26
|
Berger-Müller S, Sugie A, Takahashi F, Tavosanis G, Hakeda-Suzuki S, Suzuki T. Assessing the role of cell-surface molecules in central synaptogenesis in the Drosophila visual system. PLoS One 2013; 8:e83732. [PMID: 24386266 PMCID: PMC3873376 DOI: 10.1371/journal.pone.0083732] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 11/15/2013] [Indexed: 12/12/2022] Open
Abstract
A hallmark of the central nervous system is its spatial and functional organization in synaptic layers. During neuronal development, axons form transient contacts with potential post-synaptic elements and establish synapses with appropriate partners at specific layers. These processes are regulated by synaptic cell-adhesion molecules. In the Drosophila visual system, R7 and R8 photoreceptor subtypes target distinct layers and form en passant pre-synaptic terminals at stereotypic loci of the axonal shaft. A leucine-rich repeat transmembrane protein, Capricious (Caps), is known to be selectively expressed in R8 axons and their recipient layer, which led to the attractive hypothesis that Caps mediates R8 synaptic specificity by homophilic adhesion. Contradicting this assumption, our results indicate that Caps does not have a prominent role in synaptic-layer targeting and synapse formation in Drosophila photoreceptors, and that the specific recognition of the R8 target layer does not involve Caps homophilic axon-target interactions. We generated flies that express a tagged synaptic marker to evaluate the presence and localization of synapses in R7 and R8 photoreceptors. These genetic tools were used to assess how the synaptic profile is affected when axons are forced to target abnormal layers by expressing axon guidance molecules. When R7 axons were mistargeted to the R8-recipient layer, R7s either maintained an R7-like synaptic profile or acquired a similar profile to r8s depending on the overexpressed protein. When R7 axons were redirected to a more superficial medulla layer, the number of presynaptic terminals was reduced. These results indicate that cell-surface molecules are able to dictate synapse loci by changing the axon terminal identity in a partially cell-autonomous manner, but that presynapse formation at specific sites also requires complex interactions between pre- and post-synaptic elements.
Collapse
Affiliation(s)
- Sandra Berger-Müller
- Research Group Axon Guidance and Neuronal Connectivity, Max Planck Institute of Neurobiology, Martinsried, Germany
- CNRS 5273, Unité mixte de recherche STROMALab, Toulouse, France
| | - Atsushi Sugie
- Research Group Axon Guidance and Neuronal Connectivity, Max Planck Institute of Neurobiology, Martinsried, Germany
- Dendrite Differentiation Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Fumio Takahashi
- Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Gaia Tavosanis
- Dendrite Differentiation Group, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Satoko Hakeda-Suzuki
- Research Group Axon Guidance and Neuronal Connectivity, Max Planck Institute of Neurobiology, Martinsried, Germany
- Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Takashi Suzuki
- Research Group Axon Guidance and Neuronal Connectivity, Max Planck Institute of Neurobiology, Martinsried, Germany
- Graduate School of Bioscience & Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
- * E-mail:
| |
Collapse
|
27
|
Kannan K, Fridell YWC. Functional implications of Drosophila insulin-like peptides in metabolism, aging, and dietary restriction. Front Physiol 2013; 4:288. [PMID: 24137131 PMCID: PMC3797364 DOI: 10.3389/fphys.2013.00288] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/22/2013] [Indexed: 11/13/2022] Open
Abstract
The neuroendocrine architecture and insulin/insulin-like signaling (IIS) events in Drosophila are remarkably conserved. As IIS pathway governs growth and development, metabolism, reproduction, stress response, and longevity; temporal, spatial, and nutrient regulation of dilps encoding Drosophila insulin-like peptides (DILPs) provides potential mechanisms in modulating IIS. Of eight DILPs (DILP1–8) identified, recent studies have furthered our understanding of physiological roles of DILP2, DILP3, DILP5, and DILP6 in metabolism, aging, and responses to dietary restriction (DR), which will be the focus of this review. While the DILP producing IPCs of the brain secrete DILP2, 3, and 5, fat body produces DILP6. Identification of factors that influence dilp expression and DILP secretion has provided insight into the intricate regulatory mechanisms underlying transcriptional regulation of those genes and the activity of each peptide. Studies involving loss-of-function dilp mutations have defined the roles of DILP2 and DILP6 in carbohydrate and lipid metabolism, respectively. While DILP3 has been implicated to modulate lipid metabolism, a metabolic role for DILP5 is yet to be determined. Loss of dilp2 or adult fat body specific expression of dilp6 has been shown to extend lifespan, establishing their roles in longevity regulation. The exact role of DILP3 in aging awaits further clarification. While DILP5 has been shown associated with DR-mediated lifespan extension, contradictory evidence that precludes a direct involvement of DILP5 in DR exists. This review highlights recent findings on the importance of conserved DILPs in metabolic homeostasis, DR, and aging, providing strong evidence for the use of DILPs in modeling metabolic disorders such as diabetes and hyperinsulinemia in the fly that could further our understanding of the underlying processes and identify therapeutic strategies to treat them.
Collapse
Affiliation(s)
- Kavitha Kannan
- Department of Molecular and Cell Biology, University of Connecticut-Storrs, Storrs , CT, USA
| | | |
Collapse
|
28
|
Turkel N, Sahota VK, Bolden JE, Goulding KR, Doggett K, Willoughby LF, Blanco E, Martin-Blanco E, Corominas M, Ellul J, Aigaki T, Richardson HE, Brumby AM. The BTB-zinc finger transcription factor abrupt acts as an epithelial oncogene in Drosophila melanogaster through maintaining a progenitor-like cell state. PLoS Genet 2013; 9:e1003627. [PMID: 23874226 PMCID: PMC3715428 DOI: 10.1371/journal.pgen.1003627] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 05/30/2013] [Indexed: 01/07/2023] Open
Abstract
The capacity of tumour cells to maintain continual overgrowth potential has been linked to the commandeering of normal self-renewal pathways. Using an epithelial cancer model in Drosophila melanogaster, we carried out an overexpression screen for oncogenes capable of cooperating with the loss of the epithelial apico-basal cell polarity regulator, scribbled (scrib), and identified the cell fate regulator, Abrupt, a BTB-zinc finger protein. Abrupt overexpression alone is insufficient to transform cells, but in cooperation with scrib loss of function, Abrupt promotes the formation of massive tumours in the eye/antennal disc. The steroid hormone receptor coactivator, Taiman (a homologue of SRC3/AIB1), is known to associate with Abrupt, and Taiman overexpression also drives tumour formation in cooperation with the loss of Scrib. Expression arrays and ChIP-Seq indicates that Abrupt overexpression represses a large number of genes, including steroid hormone-response genes and multiple cell fate regulators, thereby maintaining cells within an epithelial progenitor-like state. The progenitor-like state is characterised by the failure to express the conserved Eyes absent/Dachshund regulatory complex in the eye disc, and in the antennal disc by the failure to express cell fate regulators that define the temporal elaboration of the appendage along the proximo-distal axis downstream of Distalless. Loss of scrib promotes cooperation with Abrupt through impaired Hippo signalling, which is required and sufficient for cooperative overgrowth with Abrupt, and JNK (Jun kinase) signalling, which is required for tumour cell migration/invasion but not overgrowth. These results thus identify a novel cooperating oncogene, identify mammalian family members of which are also known oncogenes, and demonstrate that epithelial tumours in Drosophila can be characterised by the maintenance of a progenitor-like state. Cancer is a multigenic process, involving cooperative interactions between oncogenes or tumour suppressors. In this study, in a genetic screen in the vinegar fly, Drosophila melanogaster, for genes that cooperate with a mutation in the cell polarity (shape) regulator, scribbled (scrib), we identify a novel cooperative oncogene, abrupt. Expression of abrupt in scrib mutant tissue in the developing eye/antennal epithelium results in overgrown invasive tumours. abrupt encodes a BTB-zinc finger transcription factor, which has homology to several cancer-causing proteins in humans, such as BCL6. Analysis of the Abrupt targets and misexpressed genes in abrupt expressing-tissue and abrupt-expressing scrib mutant tumours, revealed cell fate regulators as a major class of targets. Thus, our results reveal that deregulation of multiple cell fate factors by Abrupt expression in the context of polarity disruption is associated with a progenitor-like cell state and the formation of overgrown invasive tumours. Our findings suggest that defective polarity may also be a critical factor in BTB-zinc finger-driven human cancers, and warrants further investigation into this issue.
Collapse
Affiliation(s)
- Nezaket Turkel
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Department of Anatomy and Cell Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Virender K. Sahota
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Jessica E. Bolden
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Karen R. Goulding
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Karen Doggett
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Lee F. Willoughby
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Enrique Blanco
- Departament de Genètica i Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Enrique Martin-Blanco
- Instituto de Biología Molecular de Barcelona (CSIC), Parc Cientific de Barcelona, Barcelona, Spain
| | - Montserrat Corominas
- Departament de Genètica i Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Jason Ellul
- Bioinformatics Core Facility, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Toshiro Aigaki
- Department of Biological Sciences, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Helena E. Richardson
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Department of Anatomy and Cell Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| | - Anthony M. Brumby
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
- Department of Anatomy and Cell Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Genetics, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Eriksson BJ, Samadi L, Schmid A. The expression pattern of the genes engrailed, pax6, otd and six3 with special respect to head and eye development in Euperipatoides kanangrensis Reid 1996 (Onychophora: Peripatopsidae). Dev Genes Evol 2013; 223:237-46. [PMID: 23625086 PMCID: PMC3781328 DOI: 10.1007/s00427-013-0442-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 03/15/2013] [Indexed: 11/29/2022]
Abstract
The genes otd/otx, six3, pax6 and engrailed are involved in eye patterning in many animals. Here, we describe the expression pattern of the homologs to otd/otx, six3, pax6 and engrailed in the developing Euperipatoides kanangrensis embryos. Special reference is given to the expression in the protocerebral/ocular region. E. kanangrensis otd is expressed in the posterior part of the protocerebral/ocular segment before, during and after eye invagination. E. kanangrensis otd is also expressed segmentally in the developing ventral nerve cord. The E. kanangrensis six3 is located at the extreme anterior part of the protocerebral/ocular segment and not at the location of the developing eyes. Pax6 is expressed in a broad zone at the posterior part of the protocerebral/ocular segment but only weak expression can be seen at the early onset of eye invagination. In late stages of development, the expression in the eye is upregulated. Pax6 is also expressed in the invaginating hypocerebral organs, thus supporting earlier suggestions that the hypocerebral organs in onychophorans are glands. Pax6 transcripts are also present in the developing ventral nerve cord. The segment polarity gene engrailed is expressed at the dorsal side of the developing eye including only a subset of the cells of the invaginating eye vesicle. We show that engrailed is not expressed in the neuroectoderm of the protocerebral/ocular segment as in the other segments. In addition, we discuss other aspect of otd, six3 and pax6 expression that are relevant to our understanding of evolutionary changes in morphology and function in arthropods.
Collapse
Affiliation(s)
- Bo Joakim Eriksson
- Department of Neurobiology, University of Vienna, Althanstrasse 14, 1090, Wien, Austria.
| | | | | |
Collapse
|
30
|
Vesala L, Salminen TS, Laiho A, Hoikkala A, Kankare M. Cold tolerance and cold-induced modulation of gene expression in two Drosophila virilis group species with different distributions. INSECT MOLECULAR BIOLOGY 2012; 21:107-118. [PMID: 22122733 DOI: 10.1111/j.1365-2583.2011.01119.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The importance of high and low temperature tolerance in adaptation to changing environmental conditions has evoked new interest in modulations in gene expression and metabolism linked with stress tolerance. We investigated the effects of rapid cold hardening and cold acclimatization on the chill coma recovery times of two Drosophila virilis group species, Drosophila montana and D. virilis, with different distributions and utilized a candidate gene approach to trace changes in their gene expression during and after the cold treatments. The study showed that cold acclimatization clearly decreases chill coma recovery times in both species, whereas rapid cold hardening did not have a significant effect. Microarray analysis revealed several genes showing expression changes during different stages of cold response. Amongst the 219 genes studied, two genes showed rather consistent expression changes: hsr-omega, which was up-regulated in both study species during cold acclimatization, and Eip71CD, which was down-regulated in nearly all of the cold treatments. In addition, 29 genes showed expression changes that were more treatment- and/or species specific. Overall, different stages of cold response elicited changes mainly in genes involved in heat shock response, circadian rhythm and metabolism.
Collapse
Affiliation(s)
- Laura Vesala
- Department of Biological and Environmental Science, Centre of Excellence in Evolutionary Research, University of Jyväskylä, Finland.
| | | | | | | | | |
Collapse
|
31
|
Conserved role for the Dachshund protein with Drosophila Pax6 homolog Eyeless in insulin expression. Proc Natl Acad Sci U S A 2012; 109:2406-11. [PMID: 22308399 DOI: 10.1073/pnas.1116050109] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Members of the insulin family peptides have conserved roles in the regulation of growth and metabolism in a wide variety of metazoans. The Drosophila genome encodes seven insulin-like peptide genes, dilp1-7, and the most prominent dilps (dilp2, dilp3, and dilp5) are expressed in brain neurosecretory cells known as "insulin-producing cells" (IPCs). Although these dilps are expressed in the same cells, the expression of each dilp is regulated independently. However, the molecular mechanisms that regulate the expression of individual dilps in the IPCs remain largely unknown. Here, we show that Dachshund (Dac), which is a highly conserved nuclear protein, is a critical transcription factor that specifically regulates dilp5 expression. Dac was strongly expressed in IPCs throughout development. dac loss-of-function analyses revealed a severely reduced dilp5 expression level in young larvae. Dac interacted physically with the Drosophila Pax6 homolog Eyeless (Ey), and these proteins synergistically promoted dilp5 expression. In addition, the mammalian homolog of Dac, Dach1/2, facilitated the promoting action of Pax6 on the expression of islet hormone genes in cultured mammalian cells. These observations indicate the conserved role of Dac/Dach in controlling insulin expression in conjunction with Ey/Pax6.
Collapse
|
32
|
Altered modes of stem cell division drive adaptive intestinal growth. Cell 2011; 147:603-14. [PMID: 22036568 DOI: 10.1016/j.cell.2011.08.048] [Citation(s) in RCA: 329] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 07/08/2011] [Accepted: 08/23/2011] [Indexed: 01/03/2023]
Abstract
Throughout life, adult organs continually adapt to variable environmental factors. Adaptive mechanisms must fundamentally differ from homeostatic maintenance, but little is known about how physiological factors elicit tissue remodeling. Here, we show that specialized stem cell responses underlie the adaptive resizing of a mature organ. In the adult Drosophila midgut, intestinal stem cells interpret a nutrient cue to "break homeostasis" and drive growth when food is abundant. Activated in part by niche production of insulin, stem cells direct a growth program through two altered modes of behavior: accelerated division rates and predominance of symmetric division fates. Together, these altered modes produce a net increase in total intestinal cells, which is reversed upon withdrawal of food. Thus, tissue renewal programs are not committed to maintain cellular equilibrium; stem cells can remodel organs in response to physiological triggers.
Collapse
|
33
|
Goossens T, Kang YY, Wuytens G, Zimmermann P, Callaerts-Végh Z, Pollarolo G, Islam R, Hortsch M, Callaerts P. The Drosophila L1CAM homolog Neuroglian signals through distinct pathways to control different aspects of mushroom body axon development. Development 2011; 138:1595-605. [PMID: 21389050 DOI: 10.1242/dev.052787] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The spatiotemporal integration of adhesion and signaling during neuritogenesis is an important prerequisite for the establishment of neuronal networks in the developing brain. In this study, we describe the role of the L1-type CAM Neuroglian protein (NRG) in different steps of Drosophila mushroom body (MB) neuron axonogenesis. Selective axon bundling in the peduncle requires both the extracellular and the intracellular domain of NRG. We uncover a novel role for the ZO-1 homolog Polychaetoid (PYD) in axon branching and in sister branch outgrowth and guidance downstream of the neuron-specific isoform NRG-180. Furthermore, genetic analyses show that the role of NRG in different aspects of MB axonal development not only involves PYD, but also TRIO, SEMA-1A and RAC1.
Collapse
Affiliation(s)
- Tim Goossens
- Laboratory of Developmental Genetics, Department of Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Varghese J, Lim SF, Cohen SM. Drosophila miR-14 regulates insulin production and metabolism through its target, sugarbabe. Genes Dev 2011; 24:2748-53. [PMID: 21159815 DOI: 10.1101/gad.1995910] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Energy homeostasis depends on insulin signaling in metazoans. Insulin levels reflect the nutritional status of the animal to control levels of circulating sugar and regulate storage of resources in the form of glycogen and fat. Over the past several years, evidence has begun to accumulate that insulin production and secretion, as well as cellular responsiveness to insulin, are subject to regulation by microRNAs. Here we present evidence that miR-14 acts in the insulin-producing neurosecretory cells in the adult Drosophila brain to control metabolism. miR-14 acts in these cells through its direct target, sugarbabe. sugarbabe encodes a predicted zinc finger protein that regulates insulin gene expression in the neurosecretory cells. Regulation of sugarbabe levels by nutrients and by miR-14 combines to allow the fly to manage resource mobilization in a nutritionally variable environment.
Collapse
Affiliation(s)
- Jishy Varghese
- Institute of Molecular and Cell Biology, Singapore 138673
| | | | | |
Collapse
|
35
|
Morante J, Erclik T, Desplan C. Cell migration in Drosophila optic lobe neurons is controlled by eyeless/Pax6. Development 2011; 138:687-93. [PMID: 21208993 PMCID: PMC3026414 DOI: 10.1242/dev.056069] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2010] [Indexed: 12/13/2022]
Abstract
In the developing Drosophila optic lobe, eyeless, apterous and distal-less, three genes that encode transcription factors with important functions during development, are expressed in broad subsets of medulla neurons. Medulla cortex cells follow two patterns of cell movements to acquire their final position: first, neurons are arranged in columns below each neuroblast. Then, during pupation, they migrate laterally, intermingling with each other to reach their retinotopic position in the adult optic lobe. eyeless, which encodes a Pax6 transcription factor, is expressed early in progenitors and controls aspects of this cell migration. Its loss in medulla neurons leads to overgrowth and a failure of lateral migration during pupation. These defects in cell migration among medulla cortex cells can be rescued by removing DE-Cadherin. Thus, eyeless links neurogenesis and neuronal migration.
Collapse
Affiliation(s)
- Javier Morante
- Center for Developmental Genetics, Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Avenida Santiago Ramón y Cajal s/n, 03550 San Juan de Alicante, Spain
| | - Ted Erclik
- Center for Developmental Genetics, Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Claude Desplan
- Center for Developmental Genetics, Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| |
Collapse
|
36
|
|
37
|
Li L, Yang Y, Xue L. [Regulatory functions of Pax gene family in Drosophila development]. YI CHUAN = HEREDITAS 2010; 32:115-21. [PMID: 20176554 DOI: 10.3724/sp.j.1005.2010.00115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The Pax gene family encodes a group of important transcription factors that have been evolutionary conserved from Drosophila to human. Pax genes play pivotal roles in regulating diverse signal transduction pathways and organogenesis during embryonic development through modulating cell proliferation and self-renewal, embryonic precursor cell migration, and the coordination of specific differentiation programs. Ten members of the Pax gene family, which perform crucial regulatory functions during embryonic and postembryonic development, have been identified in Drosophila. In this report, we described the protein structures, expression patterns, and main functions of Drosophila Pax genes.
Collapse
Affiliation(s)
- Li Li
- School of Life Science and Technology, Tongji University, Shanghai 200092, China.
| | | | | |
Collapse
|
38
|
Lopes CS, Casares F. hth maintains the pool of eye progenitors and its downregulation by Dpp and Hh couples retinal fate acquisition with cell cycle exit. Dev Biol 2010; 339:78-88. [DOI: 10.1016/j.ydbio.2009.12.020] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 12/11/2009] [Accepted: 12/12/2009] [Indexed: 01/11/2023]
|
39
|
Abstract
Nutrition is a key regulator of tissue growth. In animals, nutritional status is monitored and signaled at both the cellular and systemic levels. The main mediator of cellular nutrient sensing is the protein kinase TOR (target of rapamycin). TOR receives information from levels of cellular amino acids and energy, and it regulates the activity of processes involved in cell growth, such as protein synthesis and autophagy. Insulin-like signaling is the main mechanism of systemic nutrient sensing and mediates its growth-regulatory functions largely through the phosphatidylinositol 3-kinase (PI3K)/AKT protein kinase pathway. Other nutrition-regulated hormonal mechanisms contribute to growth control by modulating the activity of insulin-like signaling. The pathways mediating signals from systemic and cellular levels converge, allowing cells to combine information from both sources. Here we give an overview of the mechanisms that adjust animal tissue growth in response to nutrition and highlight some general features of the signaling pathways involved.
Collapse
|
40
|
Clements J, Hens K, Merugu S, Dichtl B, de Couet HG, Callaerts P. Mutational analysis of the eyeless gene and phenotypic rescue reveal that an intact Eyeless protein is necessary for normal eye and brain development in Drosophila. Dev Biol 2009; 334:503-12. [PMID: 19666017 PMCID: PMC2792711 DOI: 10.1016/j.ydbio.2009.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 07/28/2009] [Accepted: 08/03/2009] [Indexed: 11/20/2022]
Abstract
Pax6 genes encode evolutionarily highly conserved transcription factors that are required for eye and brain development. Despite the characterization of mutations in Pax6 homologs in a range of organisms, and despite functional studies, it remains unclear what the relative importance is of the various parts of the Pax6 protein. To address this, we have studied the Drosophila Pax6 homolog eyeless. Specifically, we have generated new eyeless alleles, each with single missense mutations in one of the four domains of the protein. We show that these alleles result in abnormal eye and brain development while maintaining the OK107 eyeless GAL4 activity from which they were derived. We performed in vivo functional rescue experiments by expressing in an eyeless-specific pattern Eyeless proteins in which either the paired domain, the homeodomain, or the C-terminal domain was deleted. Rescue of the eye and brain phenotypes was only observed when full-length Eyeless was expressed, while all deletion constructs failed to rescue. These data, along with the phenotypes observed in the four newly characterized eyeless alleles, demonstrate the requirement for an intact Eyeless protein for normal Drosophila eye and brain development. They also suggest that some endogenous functions may be obscured in ectopic expression experiments.
Collapse
Affiliation(s)
- Jason Clements
- Laboratory of Developmental Genetics, VIB, and Center of Human Genetics, Katholieke Universiteit Leuven, Herestraat 49, Box 602, B-3000, Leuven, Belgium
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001, USA
| | - Korneel Hens
- Laboratory of Developmental Genetics, VIB, and Center of Human Genetics, Katholieke Universiteit Leuven, Herestraat 49, Box 602, B-3000, Leuven, Belgium
| | - Srinivas Merugu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001, USA
| | - Beatriz Dichtl
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001, USA
| | - H. Gert de Couet
- Department of Zoology, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Patrick Callaerts
- Laboratory of Developmental Genetics, VIB, and Center of Human Genetics, Katholieke Universiteit Leuven, Herestraat 49, Box 602, B-3000, Leuven, Belgium
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5001, USA
| |
Collapse
|
41
|
Yang X, Weber M, ZarinKamar N, Posnien N, Friedrich F, Wigand B, Beutel R, Damen WG, Bucher G, Klingler M, Friedrich M. Probing the Drosophila retinal determination gene network in Tribolium (II): The Pax6 genes eyeless and twin of eyeless. Dev Biol 2009; 333:215-27. [DOI: 10.1016/j.ydbio.2009.06.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 05/18/2009] [Accepted: 06/07/2009] [Indexed: 11/15/2022]
|