1
|
Zhang R, Ng R, Wu ST, Su CY. Targeted deletion of olfactory receptors in D. melanogaster via CRISPR/Cas9-mediated LexA knock-in. J Neurogenet 2024; 38:122-133. [PMID: 39529229 PMCID: PMC11617259 DOI: 10.1080/01677063.2024.2426014] [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: 09/20/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The study of olfaction in Drosophila melanogaster has greatly benefited from genetic reagents such as olfactory receptor mutant lines and GAL4 reporter lines. The CRISPR/Cas9 gene-editing system has been increasingly used to create null receptor mutants or replace coding regions with GAL4 reporters. To further expand this toolkit for manipulating fly olfactory receptor neurons (ORNs), we generated null alleles for 11 different olfactory receptors by using CRISPR/Cas9 to knock in LexA drivers, including multiple lines for receptors which have thus far lacked knock-in mutants. The targeted neuronal types represent a broad range of antennal ORNs from all four morphological sensillum classes. Additionally, we confirmed their loss-of-function phenotypes, assessed receptor haploinsufficiency, and evaluated the specificity of the LexA knock-in drivers. These receptor mutant lines have been deposited at the Bloomington Drosophila Stock Center for use by the broader scientific community.
Collapse
Affiliation(s)
- Runqi Zhang
- Department of Neurobiology, University of California San Diego, La Jolla, USA
| | - Renny Ng
- Department of Neurobiology, University of California San Diego, La Jolla, USA
| | - Shiuan-Tze Wu
- Department of Neurobiology, University of California San Diego, La Jolla, USA
| | - Chih-Ying Su
- Department of Neurobiology, University of California San Diego, La Jolla, USA
| |
Collapse
|
2
|
Tabuloc CA, Carlson CR, Ganjisaffar F, Truong CC, Chen CH, Lewald KM, Hidalgo S, Nicola NL, Jones CE, Sial AA, Zalom FG, Chiu JC. Transcriptome analysis of Drosophila suzukii reveals molecular mechanisms conferring pyrethroid and spinosad resistance. Sci Rep 2024; 14:19867. [PMID: 39191909 PMCID: PMC11349914 DOI: 10.1038/s41598-024-70037-x] [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: 11/20/2023] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Drosophila suzukii lay eggs in soft-skinned, ripening fruits, making this insect a serious threat to berry production. Since its 2008 introduction into North America, growers have used insecticides, such as pyrethroids and spinosads, as the primary approach for D. suzukii management, resulting in development of insecticide resistance in this pest. This study sought to identify the molecular mechanisms conferring insecticide resistance in these populations. We sequenced the transcriptomes of two pyrethroid- and two spinosad-resistant isofemale lines. In both pyrethroid-resistant lines and one spinosad-resistant line, we identified overexpression of metabolic genes that are implicated in resistance in other insect pests. In the other spinosad-resistant line, we observed an overexpression of cuticular genes that have been linked to resistance. Our findings enabled the development of molecular diagnostics that we used to confirm persistence of insecticide resistance in California, U.S.A. To validate these findings, we leveraged D. melanogaster mutants with reduced expression of metabolic or cuticular genes that were found to be upregulated in resistant D. suzukii to demonstrate that these genes are involved in promoting resistance. This study is the first to characterize the molecular mechanisms of insecticide resistance in D. suzukii and provides insights into how current management practices can be optimized.
Collapse
Affiliation(s)
- Christine A Tabuloc
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Curtis R Carlson
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Fatemeh Ganjisaffar
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Cindy C Truong
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Ching-Hsuan Chen
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Kyle M Lewald
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Sergio Hidalgo
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Nicole L Nicola
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Cera E Jones
- Department of Entomology, University of Georgia, Athens, GA, USA
| | - Ashfaq A Sial
- Department of Entomology, University of Georgia, Athens, GA, USA
| | - Frank G Zalom
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA
| | - Joanna C Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, California, USA.
| |
Collapse
|
3
|
Lv P, Yang X, Du J. LKRSDH-dependent histone modifications of insulin-like peptide sites contribute to age-related circadian rhythm changes. Nat Commun 2024; 15:3336. [PMID: 38637528 PMCID: PMC11026460 DOI: 10.1038/s41467-024-47740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
To understand aging impact on the circadian rhythm, we screened for factors influencing circadian changes during aging. Our findings reveal that LKRSDH mutation significantly reduces rhythmicity in aged flies. RNA-seq identifies a significant increase in insulin-like peptides (dilps) in LKRSDH mutants due to the combined effects of H3R17me2 and H3K27me3 on transcription. Genetic evidence suggests that LKRSDH regulates age-related circadian rhythm changes through art4 and dilps. ChIP-seq analyzes whole genome changes in H3R17me2 and H3K27me3 histone modifications in young and old flies with LKRSDH mutation and controls. The results reveal a correlation between H3R17me2 and H3K27me3, underscoring the role of LKRSDH in regulating gene expression and modification levels during aging. Overall, our study demonstrates that LKRSDH-dependent histone modifications at dilps sites contribute to age-related circadian rhythm changes. This data offers insights and a foundational reference for aging research by unveiling the relationship between LKRSDH and H3R17me2/H3K27me3 histone modifications in aging.
Collapse
Affiliation(s)
- Pengfei Lv
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Xingzhuo Yang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China
| | - Juan Du
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
4
|
Nicolson S, Manning JA, Lim Y, Jiang X, Kolze E, Dayan S, Umargamwala R, Xu T, Sandow JJ, Webb AI, Kumar S, Denton D. The Drosophila ZNRF1/2 homologue, detour, interacts with HOPS complex and regulates autophagy. Commun Biol 2024; 7:183. [PMID: 38360932 PMCID: PMC10869362 DOI: 10.1038/s42003-024-05834-1] [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: 12/14/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
Autophagy, the process of elimination of cellular components by lysosomal degradation, is essential for animal development and homeostasis. Using the autophagy-dependent Drosophila larval midgut degradation model we identified an autophagy regulator, the RING domain ubiquitin ligase CG14435 (detour). Depletion of detour resulted in increased early-stage autophagic vesicles, premature tissue contraction, and overexpression of detour or mammalian homologues, ZNRF1 and ZNRF2, increased autophagic vesicle size. The ablation of ZNRF1 or ZNRF2 in mammalian cells increased basal autophagy. We identified detour interacting proteins including HOPS subunits, deep orange (dor/VPS18), Vacuolar protein sorting 16A (VPS16A), and light (lt/VPS41) and found that detour promotes their ubiquitination. The detour mutant accumulated autophagy-related proteins in young adults, displayed premature ageing, impaired motor function, and activation of innate immunity. Collectively, our findings suggest a role for detour in autophagy, likely through regulation of HOPS complex, with implications for healthy aging.
Collapse
Affiliation(s)
- Shannon Nicolson
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Jantina A Manning
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Yoon Lim
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Xin Jiang
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Erica Kolze
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5001, Australia
| | - Sonia Dayan
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Ruchi Umargamwala
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Tianqi Xu
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Jarrod J Sandow
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Andrew I Webb
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia.
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, 5001, Australia.
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia.
| |
Collapse
|
5
|
Eiman MN, Kumar S, Serrano Negron YL, Tansey TR, Harbison ST. Genome-wide association in Drosophila identifies a role for Piezo and Proc-R in sleep latency. Sci Rep 2024; 14:260. [PMID: 38168575 PMCID: PMC10761942 DOI: 10.1038/s41598-023-50552-z] [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: 03/28/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Sleep latency, the amount of time that it takes an individual to fall asleep, is a key indicator of sleep need. Sleep latency varies considerably both among and within species and is heritable, but lacks a comprehensive description of its underlying genetic network. Here we conduct a genome-wide association study of sleep latency. Using previously collected sleep and activity data on a wild-derived population of flies, we calculate sleep latency, confirming significant, heritable genetic variation for this complex trait. We identify 520 polymorphisms in 248 genes contributing to variability in sleep latency. Tests of mutations in 23 candidate genes and additional putative pan-neuronal knockdown of 9 of them implicated CG44153, Piezo, Proc-R and Rbp6 in sleep latency. Two large-effect mutations in the genes Proc-R and Piezo were further confirmed via genetic rescue. This work greatly enhances our understanding of the genetic factors that influence variation in sleep latency.
Collapse
Affiliation(s)
- Matthew N Eiman
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Shailesh Kumar
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Division of Neuroscience and Behavior, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Yazmin L Serrano Negron
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Terry R Tansey
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan T Harbison
- Laboratory of Systems Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Tsuji M, Nishizuka Y, Emoto K. Threat gates visual aversion via theta activity in Tachykinergic neurons. Nat Commun 2023; 14:3987. [PMID: 37443364 PMCID: PMC10345120 DOI: 10.1038/s41467-023-39667-z] [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: 10/19/2022] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Animals must adapt sensory responses to an ever-changing environment for survival. Such sensory modulation is especially critical in a threatening situation, in which animals often promote aversive responses to, among others, visual stimuli. Recently, threatened Drosophila has been shown to exhibit a defensive internal state. Whether and how threatened Drosophila promotes visual aversion, however, remains elusive. Here we report that mechanical threats to Drosophila transiently gate aversion from an otherwise neutral visual object. We further identified the neuropeptide tachykinin, and a single cluster of neurons expressing it ("Tk-GAL42 ∩ Vglut neurons"), that are responsible for gating visual aversion. Calcium imaging analysis revealed that mechanical threats are encoded in Tk-GAL42 ∩ Vglut neurons as elevated activity. Remarkably, we also discovered that a visual object is encoded in Tk-GAL42 ∩ Vglut neurons as θ oscillation, which is causally linked to visual aversion. Our data reveal how a single cluster of neurons adapt organismal sensory response to a threatening situation through a neuropeptide and a combination of rate/temporal coding schemes.
Collapse
Affiliation(s)
- Masato Tsuji
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuto Nishizuka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kazuo Emoto
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
7
|
Neuropeptide diuretic hormone 31 mediates memory and sleep via distinct neural pathways in Drosophila. Neurosci Res 2023:S0168-0102(23)00037-8. [PMID: 36780946 DOI: 10.1016/j.neures.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/29/2023] [Accepted: 02/07/2023] [Indexed: 02/13/2023]
Abstract
Memory formation and sleep regulation are critical for brain functions in animals from invertebrates to humans. Neuropeptides play a pivotal role in regulating physiological behaviors, including memory formation and sleep. However, the detailed mechanisms by which neuropeptides regulate these physiological behaviors remains unclear. Herein, we report that neuropeptide diuretic hormone 31 (DH31) positively regulates memory formation and sleep in Drosophila melanogaster. The expression of DH31 in the dorsal and ventral fan-shaped body (dFB and vFB) neurons of the central complex and ventral lateral clock neurons (LNvs) in the brain was responsive to sleep regulation. In addition, the expression of membrane-tethered DH31 in dFB neurons rescued sleep defects in Dh31 mutants, suggesting that DH31 secreted from dFB, vFB, and LNvs acts on the DH31 receptor in the dFB to regulate sleep partly in an autoregulatory feedback loop. Moreover, the expression of DH31 in octopaminergic neurons, but not in the dFB neurons, is involved in forming intermediate-term memory. Our results suggest that DH31 regulates memory formation and sleep through distinct neural pathways.
Collapse
|
8
|
Trammell CE, Rowe EH, Jones BJ, Char AB, Fawcett S, Ahlers LR, Goodman AG. Insulin-mediated endothelin signaling is antiviral during West Nile virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524426. [PMID: 36712090 PMCID: PMC9882177 DOI: 10.1101/2023.01.17.524426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
West Nile virus (WNV) is the most prevalent mosquito-borne virus in the United States with approximately 2,000 cases each year. There are currently no approved human vaccines and a lack of prophylactic and therapeutic treatments. Understanding host responses to infection may reveal potential intervention targets to reduce virus replication and disease progression. The use of Drosophila melanogaster as a model organism to understand innate immunity and host antiviral responses is well established. Previous studies revealed that insulin-mediated signaling regulates WNV infection in invertebrates by regulating canonical antiviral pathways. Because insulin signaling is well-conserved across insect and mammalian species, we sought to determine if results using D. melanogaster can be extrapolated for the analysis of orthologous pathways in humans. Here, we identify insulin-mediated endothelin signaling using the D. melanogaster model and evaluate an orthologous pathway in human cells during WNV infection. We demonstrate that endothelin signaling reduces WNV replication through the activation of canonical antiviral signaling. Taken together, our findings show that endothelin-mediated antiviral immunity is broadly conserved across species and reduces replication of viruses that can cause severe human disease. IMPORTANCE Arboviruses, particularly those transmitted by mosquitoes, pose a significant threat to humans and are an increasing concern because of climate change, human activity, and expanding vector-competent populations. West Nile virus is of significant concern as the most frequent mosquito-borne disease transmitted annually within the continental United States. Here, we identify a previously uncharacterized signaling pathway that impacts West Nile virus infection, namely endothelin signaling. Additionally, we demonstrate that we can successfully translate results obtained from D. melanogaster into the more relevant human system. Our results add to the growing field of insulin-mediated antiviral immunity and identifies potential biomarkers or intervention targets to better address West Nile virus infection and severe disease.
Collapse
Affiliation(s)
- Chasity E. Trammell
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Evelyn H. Rowe
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Brianne J. Jones
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Aditya B. Char
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Stephen Fawcett
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Laura R.H. Ahlers
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alan G. Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
9
|
Identification of the Telomere elongation Mutation in Drosophila. Cells 2022; 11:cells11213484. [DOI: 10.3390/cells11213484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Telomeres in Drosophila melanogaster, which have inspired a large part of Sergio Pimpinelli work, are similar to those of other eukaryotes in terms of their function. Yet, their length maintenance relies on the transposition of the specialized retrotransposons Het-A, TART, and TAHRE, rather than on the activity of the enzyme telomerase as it occurs in most other eukaryotic organisms. The length of the telomeres in Drosophila thus depends on the number of copies of these transposable elements. Our previous work has led to the isolation of a dominant mutation, Tel1, that caused a several-fold elongation of telomeres. In this study, we molecularly identified the Tel1 mutation by a combination of transposon-induced, site-specific recombination and next-generation sequencing. Recombination located Tel1 to a 15 kb region in 92A. Comparison of the DNA sequence in this region with the Drosophila Genetic Reference Panel of wild-type genomic sequences delimited Tel1 to a 3 bp deletion inside intron 8 of Ino80. Furthermore, CRISPR/Cas9-induced deletions surrounding the same region exhibited the Tel1 telomere phenotype, confirming a strict requirement of this intron 8 gene sequence for a proper regulation of Drosophila telomere length.
Collapse
|
10
|
Kanca O, Zirin J, Hu Y, Tepe B, Dutta D, Lin WW, Ma L, Ge M, Zuo Z, Liu LP, Levis RW, Perrimon N, Bellen HJ. An expanded toolkit for Drosophila gene tagging using synthesized homology donor constructs for CRISPR-mediated homologous recombination. eLife 2022; 11:e76077. [PMID: 35723254 PMCID: PMC9239680 DOI: 10.7554/elife.76077] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 06/19/2022] [Indexed: 11/13/2022] Open
Abstract
Previously, we described a large collection of Drosophila strains that each carry an artificial exon containing a T2AGAL4 cassette inserted in an intron of a target gene based on CRISPR-mediated homologous recombination. These alleles permit numerous applications and have proven to be very useful. Initially, the homologous recombination-based donor constructs had long homology arms (>500 bps) to promote precise integration of large constructs (>5 kb). Recently, we showed that in vivo linearization of the donor constructs enables insertion of large artificial exons in introns using short homology arms (100-200 bps). Shorter homology arms make it feasible to commercially synthesize homology donors and minimize the cloning steps for donor construct generation. Unfortunately, about 58% of Drosophila genes lack a suitable coding intron for integration of artificial exons in all of the annotated isoforms. Here, we report the development of new set of constructs that allow the replacement of the coding region of genes that lack suitable introns with a KozakGAL4 cassette, generating a knock-out/knock-in allele that expresses GAL4 similarly as the targeted gene. We also developed custom vector backbones to further facilitate and improve transgenesis. Synthesis of homology donor constructs in custom plasmid backbones that contain the target gene sgRNA obviates the need to inject a separate sgRNA plasmid and significantly increases the transgenesis efficiency. These upgrades will enable the targeting of nearly every fly gene, regardless of exon-intron structure, with a 70-80% success rate.
Collapse
Affiliation(s)
- Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Duncan Neurological Research Institute, Texas Children HospitalHoustonUnited States
| | - Jonathan Zirin
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Yanhui Hu
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Burak Tepe
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Duncan Neurological Research Institute, Texas Children HospitalHoustonUnited States
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Duncan Neurological Research Institute, Texas Children HospitalHoustonUnited States
| | - Wen-Wen Lin
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Duncan Neurological Research Institute, Texas Children HospitalHoustonUnited States
| | - Liwen Ma
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Duncan Neurological Research Institute, Texas Children HospitalHoustonUnited States
| | - Ming Ge
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Duncan Neurological Research Institute, Texas Children HospitalHoustonUnited States
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Duncan Neurological Research Institute, Texas Children HospitalHoustonUnited States
| | - Lu-Ping Liu
- Department of Genetics, Harvard Medical SchoolBostonUnited States
| | - Robert W Levis
- Department of Embryology, Carnegie Institution for ScienceBaltimoreUnited States
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical SchoolBostonUnited States
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
11
|
Qiu S, Xiao C, Robertson RM. Knockdown of a Cyclic Nucleotide-Gated Ion Channel Impairs Locomotor Activity and Recovery From Hypoxia in Adult Drosophila melanogaster. Front Physiol 2022; 13:852919. [PMID: 35530504 PMCID: PMC9075734 DOI: 10.3389/fphys.2022.852919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclic guanosine monophosphate (cGMP) modulates the speed of recovery from anoxia in adult Drosophila and mediates hypoxia-related behaviors in larvae. Cyclic nucleotide-gated channels (CNG) and cGMP-activated protein kinase (PKG) are two cGMP downstream targets. PKG is involved in behavioral tolerance to hypoxia and anoxia in adults, however little is known about a role for CNG channels. We used a CNGL (CNG-like) mutant with reduced CNGL transcripts to investigate the contribution of CNGL to the hypoxia response. CNGL mutants had reduced locomotor activity under normoxia. A shorter distance travelled in a standard locomotor assay was due to a slower walking speed and more frequent stops. In control flies, hypoxia immediately reduced path length per minute. Flies took 30–40 min in normoxia for >90% recovery of path length per minute from 15 min hypoxia. CNGL mutants had impaired recovery from hypoxia; 40 min for ∼10% recovery of walking speed. The effects of CNGL mutation on locomotor activity and recovery from hypoxia were recapitulated by pan-neuronal CNGL knockdown. Genetic manipulation to increase cGMP in the CNGL mutants increased locomotor activity under normoxia and eliminated the impairment of recovery from hypoxia. We conclude that CNGL channels and cGMP signaling are involved in the control of locomotor activity and the hypoxic response of adult Drosophila.
Collapse
Affiliation(s)
- Shuang Qiu
- Department of Biology, Queen's University, Kingston, ON, Canada
| | - Chengfeng Xiao
- Department of Biology, Queen's University, Kingston, ON, Canada
| | | |
Collapse
|
12
|
Neophytou C, Pitsouli C. Biotin controls intestinal stem cell mitosis and host-microbiome interactions. Cell Rep 2022; 38:110505. [PMID: 35263602 DOI: 10.1016/j.celrep.2022.110505] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 12/11/2021] [Accepted: 02/16/2022] [Indexed: 12/12/2022] Open
Abstract
Diet is a key regulator of metabolism and interacts with the intestinal microbiome. Here, we study the role of the Drosophila intestinal stem cell (ISC)-specific biotin transporter Smvt in midgut homeostasis, infection-induced regeneration, and tumorigenesis. We show that Smvt-transported biotin in ISCs is necessary for ISC mitosis. Smvt deficiency impairs intestinal maintenance, which can be rescued by the human Smvt, encoded by SLC5A6. ISC-specific, Smvt-silenced flies exhibit microbial dysbiosis, whereby the growth of Providencia sneebia, an opportunistic pathogen, is favored. Dysbiosis correlates with increased Nox expression, reactive oxygen species (ROS), and enterocyte apoptosis. Flies acquire biotin from their diet and microbiota. We show that, when dietary biotin is scarce, biotin-producing commensals, e.g., E. coli, can rescue reduced ISC mitosis. Smvt and commensals also control intestinal tumor growth. Our findings suggest that direct modification of the gut microbiome by biotin can serve as an approach for the treatment of dysbiosis-promoted diseases and tumorigenesis control.
Collapse
Affiliation(s)
- Constantina Neophytou
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia 2109, Cyprus.
| |
Collapse
|
13
|
Slo2/K Na Channels in Drosophila Protect against Spontaneous and Induced Seizure-like Behavior Associated with an Increased Persistent Na + Current. J Neurosci 2021; 41:9047-9063. [PMID: 34544836 DOI: 10.1523/jneurosci.0290-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/20/2021] [Accepted: 09/13/2021] [Indexed: 11/21/2022] Open
Abstract
Na+ sensitivity is a unique feature of Na+-activated K+ (KNa) channels, making them naturally suited to counter a sudden influx in Na+ ions. As such, it has long been suggested that KNa channels may serve a protective function against excessive excitation associated with neuronal injury and disease. This hypothesis, however, has remained largely untested. Here, we examine KNa channels encoded by the Drosophila Slo2 (dSlo2) gene in males and females. We show that dSlo2/KNa channels are selectively expressed in cholinergic neurons in the adult brain, as well as in glutamatergic motor neurons, where dampening excitation may function to inhibit global hyperactivity and seizure-like behavior. Indeed, we show that effects of feeding Drosophila a cholinergic agonist are exacerbated by the loss of dSlo2/KNa channels. Similar to mammalian Slo2/KNa channels, we show that dSlo2/KNa channels encode a TTX-sensitive K+ conductance, indicating that dSlo2/KNa channels can be activated by Na+ carried by voltage-dependent Na+ channels. We then tested the role of dSlo2/KNa channels in established genetic seizure models in which the voltage-dependent persistent Na+ current (INap) is elevated. We show that the absence of dSlo2/KNa channels increased susceptibility to mechanically induced seizure-like behavior. Similar results were observed in WT flies treated with veratridine, an enhancer of INap Finally, we show that loss of dSlo2/KNa channels in both genetic and pharmacologically primed seizure models resulted in the appearance of spontaneous seizures. Together, our results support a model in which dSlo2/KNa channels, activated by neuronal overexcitation, contribute to a protective threshold to suppress the induction of seizure-like activity.SIGNIFICANCE STATEMENT Slo2/KNa channels are unique in that they constitute a repolarizing K+ pore that is activated by the depolarizing Na+ ion, making them naturally suited to function as a protective "brake" against overexcitation and Na+ overload. Here, we test this hypothesis in vivo by examining how a null mutation of the Drosophila Slo2 (dSlo2)/KNa gene affects seizure-like behavior in genetic and pharmacological models of epilepsy. We show that indeed the loss of dSlo2/KNa channels results in increased incidence and severity of induced seizure behavior, as well as the appearance of spontaneous seizure activity. Our results advance our understanding of neuronal excitability and protective mechanisms that preserve normal physiology and the suppression of seizure susceptibility.
Collapse
|
14
|
Rhee JE, Choi JH, Park JH, Lee G, Pak B, Kwon SH, Jeon SH. CG11426 gene product negatively regulates glial population size in the Drosophila eye imaginal disc. Dev Neurobiol 2021; 81:805-816. [PMID: 34047015 DOI: 10.1002/dneu.22838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/03/2021] [Accepted: 05/16/2021] [Indexed: 11/09/2022]
Abstract
Glial cells play essential roles in the nervous system. Although glial populations are tightly regulated, the mechanisms regulating the population size remain poorly understood. Since Drosophila glial cells are similar to the human counterparts in their functions and shapes, rendering them an excellent model system to understand the human glia biology. Lipid phosphate phosphatases (LPPs) are important for regulating bioactive lipids. In Drosophila, there are three known LPP-encoding genes: wunen, wunen-2, and lazaro. The wunens are important for germ cell migration and survival and septate junction formation during tracheal development. Lazaro is involved in phototransduction. In the present study, we characterized a novel Drosophila LPP-encoding gene, CG11426. Suppression of CG11426 increased glial cell number in the eye imaginal disc during larval development, while ectopic CG11426 expression decreased it. Both types of mutation also caused defects in axon projection to the optic lobe in larval eye-brain complexes. Moreover, CG11426 promoted apoptosis via inhibiting ERK signaling in the eye imaginal disc. Taken together, these findings demonstrated that CG11426 gene product negatively regulates ERK signaling to promote apoptosis for proper maintenance of the glial population in the developing eye disc.
Collapse
Affiliation(s)
- Jong-Eun Rhee
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| | - Jin-Hyeon Choi
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| | - Jae H Park
- Department of Biochemistry & Cellular and Molecular Biology, and Neuronet Research Center, University of Tennessee, Knoxville, Tennessee, USA
| | - Gyunghee Lee
- Department of Biochemistry & Cellular and Molecular Biology, and Neuronet Research Center, University of Tennessee, Knoxville, Tennessee, USA
| | - Banya Pak
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| | - Seung-Hae Kwon
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Sang-Hak Jeon
- Department of Biology Education, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Physical and Chemical Barriers in the Larval Midgut Confer Developmental Resistance to Virus Infection in Drosophila. Viruses 2021; 13:v13050894. [PMID: 34065985 PMCID: PMC8151258 DOI: 10.3390/v13050894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/23/2023] Open
Abstract
Insects can become lethally infected by the oral intake of a number of insect-specific viruses. Virus infection commonly occurs in larvae, given their active feeding behaviour; however, older larvae often become resistant to oral viral infections. To investigate mechanisms that contribute to resistance throughout the larval development, we orally challenged Drosophila larvae at different stages of their development with Drosophila C virus (DCV, Dicistroviridae). Here, we showed that DCV-induced mortality is highest when infection initiates early in larval development and decreases the later in development the infection occurs. We then evaluated the peritrophic matrix as an antiviral barrier within the gut using a Crystallin-deficient fly line (Crys-/-), whose PM is weakened and becomes more permeable to DCV-sized particles as the larva ages. This phenotype correlated with increasing mortality the later in development oral challenge occurred. Lastly, we tested in vitro the infectivity of DCV after incubation at pH conditions that may occur in the midgut. DCV virions were stable in a pH range between 3.0 and 10.5, but their infectivity decreased at least 100-fold below (1.0) and above (12.0) this range. We did not observe such acidic conditions in recently hatched larvae. We hypothesise that, in Drosophila larvae, the PM is essential for containing ingested virions separated from the gut epithelium, while highly acidic conditions inactivate the majority of the virions as they transit.
Collapse
|
16
|
Spierer AN, Mossman JA, Smith SP, Crawford L, Ramachandran S, Rand DM. Natural variation in the regulation of neurodevelopmental genes modifies flight performance in Drosophila. PLoS Genet 2021; 17:e1008887. [PMID: 33735180 PMCID: PMC7971549 DOI: 10.1371/journal.pgen.1008887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 01/26/2021] [Indexed: 12/28/2022] Open
Abstract
The winged insects of the order Diptera are colloquially named for their most recognizable phenotype: flight. These insects rely on flight for a number of important life history traits, such as dispersal, foraging, and courtship. Despite the importance of flight, relatively little is known about the genetic architecture of flight performance. Accordingly, we sought to uncover the genetic modifiers of flight using a measure of flies’ reaction and response to an abrupt drop in a vertical flight column. We conducted a genome wide association study (GWAS) using 197 of the Drosophila Genetic Reference Panel (DGRP) lines, and identified a combination of additive and marginal variants, epistatic interactions, whole genes, and enrichment across interaction networks. Egfr, a highly pleiotropic developmental gene, was among the most significant additive variants identified. We functionally validated 13 of the additive candidate genes’ (Adgf-A/Adgf-A2/CG32181, bru1, CadN, flapper (CG11073), CG15236, flippy (CG9766), CREG, Dscam4, form3, fry, Lasp/CG9692, Pde6, Snoo), and introduce a novel approach to whole gene significance screens: PEGASUS_flies. Additionally, we identified ppk23, an Acid Sensing Ion Channel (ASIC) homolog, as an important hub for epistatic interactions. We propose a model that suggests genetic modifiers of wing and muscle morphology, nervous system development and function, BMP signaling, sexually dimorphic neural wiring, and gene regulation are all important for the observed differences flight performance in a natural population. Additionally, these results represent a snapshot of the genetic modifiers affecting drop-response flight performance in Drosophila, with implications for other insects. Insect flight is a widely recognizable phenotype of many winged insects, hence the name: flies. While fruit flies, or Drosophila melanogaster, are a genetically tractable model, flight performance is a highly integrative phenotype, and therefore challenging to identify comprehensively which genetic modifiers contribute to its genetic architecture. Accordingly, we screened 197 Drosophila Genetic Reference Panel lines for their ability to react and respond to an abrupt drop. Using several computational approaches, we identified additive, marginal, and epistatic variants, as well as whole genes and altered sub-networks of gene-gene and protein-protein interaction networks that contribute to variation in flight performance. More generally, we demonstrate the benefits of employing multiple methodologies to elucidate the genetic architecture of complex traits. Many variants and genes mapped to regions of the genome that affect neurodevelopment, wing and muscle development, and regulation of gene expression. We also introduce PEGASUS_flies, a Drosophila-adapted version of the PEGASUS platform first used in human studies, to infer gene-level significance of association based on the gene’s distribution of individual variant P-values. Our results contribute to the debate over the relative importance of individual, additive factors and epistatic, or higher order, interactions, in the mapping of genotype to phenotype.
Collapse
Affiliation(s)
- Adam N Spierer
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
| | - Jim A Mossman
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Samuel Pattillo Smith
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Lorin Crawford
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
- Microsoft Research New England, Cambridge, Massachusetts, United States of America
| | - Sohini Ramachandran
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - David M Rand
- Department of Ecology and Evolutionary Biology, Brown University, Providence, Rhode Island, United States of America
- Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
17
|
Li W, Liang J, Outeda P, Turner S, Wakimoto BT, Watnick T. A genetic screen in Drosophila reveals an unexpected role for the KIP1 ubiquitination-promoting complex in male fertility. PLoS Genet 2020; 16:e1009217. [PMID: 33378371 PMCID: PMC7802972 DOI: 10.1371/journal.pgen.1009217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 01/12/2021] [Accepted: 10/19/2020] [Indexed: 12/22/2022] Open
Abstract
A unifying feature of polycystin-2 channels is their localization to both primary and motile cilia/flagella. In Drosophila melanogaster, the fly polycystin-2 homologue, Amo, is an ER protein early in sperm development but the protein must ultimately cluster at the flagellar tip in mature sperm to be fully functional. Male flies lacking appropriate Amo localization are sterile due to abnormal sperm motility and failure of sperm storage. We performed a forward genetic screen to identify additional proteins that mediate ciliary trafficking of Amo. Here we report that Drosophila homologues of KPC1 and KPC2, which comprise the mammalian KIP1 ubiquitination-promoting complex (KPC), form a conserved unit that is required for the sperm tail tip localization of Amo. Male flies lacking either KPC1 or KPC2 phenocopy amo mutants and are sterile due to a failure of sperm storage. KPC is a heterodimer composed of KPC1, an E3 ligase, and KPC2 (or UBAC1), an adaptor protein. Like their mammalian counterparts Drosophila KPC1 and KPC2 physically interact and they stabilize one another at the protein level. In flies, KPC2 is monoubiquitinated and phosphorylated and this modified form of the protein is located in mature sperm. Neither KPC1 nor KPC2 directly interact with Amo but they are detected in proximity to Amo at the tip of the sperm flagellum. In summary we have identified a new complex that is involved in male fertility in Drosophila melanogaster.
Collapse
Affiliation(s)
- Weizhe Li
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Jinqing Liang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Patricia Outeda
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Stacey Turner
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Barbara T. Wakimoto
- Department of Biology, University of Washington Seattle, WA, United States of America
| | - Terry Watnick
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
18
|
Gao B, Zong W, Miskey C, Ullah N, Diaby M, Chen C, Wang X, Ivics Z, Song C. Intruder (DD38E), a recently evolved sibling family of DD34E/Tc1 transposons in animals. Mob DNA 2020; 11:32. [PMID: 33303022 PMCID: PMC7731502 DOI: 10.1186/s13100-020-00227-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A family of Tc1/mariner transposons with a characteristic DD38E triad of catalytic amino acid residues, named Intruder (IT), was previously discovered in sturgeon genomes, but their evolutionary landscapes remain largely unknown. RESULTS Here, we comprehensively investigated the evolutionary profiles of ITs, and evaluated their cut-and-paste activities in cells. ITs exhibited a narrow taxonomic distribution pattern in the animal kingdom, with invasions into two invertebrate phyla (Arthropoda and Cnidaria) and three vertebrate lineages (Actinopterygii, Agnatha, and Anura): very similar to that of the DD36E/IC family. Some animal orders and species seem to be more hospitable to Tc1/mariner transposons, one order of Amphibia and seven Actinopterygian orders are the most common orders with horizontal transfer events and have been invaded by all four families (DD38E/IT, DD35E/TR, DD36E/IC and DD37E/TRT) of Tc1/mariner transposons, and eight Actinopterygii species were identified as the major hosts of these families. Intact ITs have a total length of 1.5-1.7 kb containing a transposase gene flanked by terminal inverted repeats (TIRs). The phylogenetic tree and sequence identity showed that IT transposases were most closely related to DD34E/Tc1. ITs have been involved in multiple events of horizontal transfer in vertebrates and have invaded most lineages recently (< 5 million years ago) based on insertion age analysis. Accordingly, ITs presented high average sequence identity (86-95%) across most vertebrate species, suggesting that some are putatively active. ITs can transpose in human HeLa cells, and the transposition efficiency of consensus TIRs was higher than that of the TIRs of natural isolates. CONCLUSIONS We conclude that DD38E/IT originated from DD34E/Tc1 and can be detected in two invertebrate phyla (Arthropoda and Cnidaria), and in three vertebrate lineages (Actinopterygii, Agnatha and Anura). IT has experienced multiple HT events in animals, dominated by recent amplifications in most species and has high identity among vertebrate taxa. Our reconstructed IT transposon vector designed according to the sequence from the "cat" genome showed high cut-and-paste activity. The data suggest that IT has been acquired recently and is active in many species. This study is meaningful for understanding the evolution of the Tc1/mariner superfamily members and their hosts.
Collapse
Affiliation(s)
- Bo Gao
- College of Animal Science & Technology, Yangzhou University, 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China.,Division of Medical Biotechnology, Paul Ehrlich Institute, 63225, Langen, Germany
| | - Wencheng Zong
- College of Animal Science & Technology, Yangzhou University, 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Csaba Miskey
- Division of Medical Biotechnology, Paul Ehrlich Institute, 63225, Langen, Germany
| | - Numan Ullah
- College of Animal Science & Technology, Yangzhou University, 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Mohamed Diaby
- College of Animal Science & Technology, Yangzhou University, 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Cai Chen
- College of Animal Science & Technology, Yangzhou University, 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Xiaoyan Wang
- College of Animal Science & Technology, Yangzhou University, 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, 63225, Langen, Germany
| | - Chengyi Song
- College of Animal Science & Technology, Yangzhou University, 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
19
|
Kumar S, Tunc I, Tansey TR, Pirooznia M, Harbison ST. Identification of Genes Contributing to a Long Circadian Period in Drosophila Melanogaster. J Biol Rhythms 2020; 36:239-253. [PMID: 33274675 DOI: 10.1177/0748730420975946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The endogenous circadian period of animals and humans is typically very close to 24 h. Individuals with much longer circadian periods have been observed, however, and in the case of humans, these deviations have health implications. Previously, we observed a line of Drosophila with a very long average period of 31.3 h for locomotor activity behavior. Preliminary mapping indicated that the long period did not map to known canonical clock genes but instead mapped to multiple chromosomes. Using RNA-Seq, we surveyed the whole transcriptome of fly heads from this line across time and compared it with a wild-type control. A three-way generalized linear model revealed that approximately two-thirds of the genes were expressed differentially among the two genotypes, while only one quarter of the genes varied across time. Using these results, we applied algorithms to search for genes that oscillated over 24 h, identifying genes not previously known to cycle. We identified 166 differentially expressed genes that overlapped with a previous Genome-wide Association Study (GWAS) of circadian behavior, strongly implicating them in the long-period phenotype. We tested mutations in 45 of these genes for their effect on the circadian period. Mutations in Alk, alph, CG10089, CG42540, CG6034, Kairos (CG6123), CG8768, klg, Lar, sick, and tinc had significant effects on the circadian period, with seven of these mutations increasing the circadian period of locomotor activity behavior. Genetic rescue of mutant Kairos restored the circadian period to wild-type levels, suggesting it has a critical role in determining period length in constant darkness.
Collapse
Affiliation(s)
- Shailesh Kumar
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Ilker Tunc
- Bioinformatics and Computational Biology Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Terry R Tansey
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Core, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Susan T Harbison
- Laboratory of Systems Genetics, Systems Biology Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| |
Collapse
|
20
|
Spana EP, Abrams AB, Ellis KT, Klein JC, Ruderman BT, Shi AH, Zhu D, Stewart A, May S. speck, First Identified in Drosophila melanogaster in 1910, Is Encoded by the Arylalkalamine N-Acetyltransferase (AANAT1) Gene. G3 (BETHESDA, MD.) 2020; 10:3387-3398. [PMID: 32709620 PMCID: PMC7466976 DOI: 10.1534/g3.120.401470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022]
Abstract
The pigmentation mutation speck is a commonly used recombination marker characterized by a darkly pigmented region at the wing hinge. Identified in 1910 by Thomas Hunt Morgan, speck was characterized by Sturtevant as the most "workable" mutant in the rightmost region of the second chromosome and eventually localized to 2-107.0 and 60C1-2. Though the first speck mutation was isolated over 110 years ago, speck is still not associated with any gene. Here, as part of an undergraduate-led research effort, we show that speck is encoded by the Arylalkylamine N-acetyltransferase 1 (AANAT1) gene. Both alleles from the Morgan lab contain a retrotransposon in exon 1 of the RB transcript of the AANAT1 gene. We have also identified a new insertion allele and generated multiple deletion alleles in AANAT1 that all give a strong speck phenotype. In addition, expression of AANAT1 RNAi constructs either ubiquitously or in the dorsal portion of the developing wing generates a similar speck phenotype. We find that speck alleles have additional phenotypes, including ectopic pigmentation in the posterior pupal case, leg joints, cuticular sutures and overall body color. We propose that the acetylated dopamine generated by AANAT1 decreases the dopamine pool available for melanin production. When AANAT1 function is decreased, the excess dopamine enters the melanin pathway to generate the speck phenotype.
Collapse
Affiliation(s)
- Eric P Spana
- Department of Biology, Duke University, Durham, NC 27708
| | | | | | - Jason C Klein
- Department of Biology, Duke University, Durham, NC 27708
| | | | - Alvin H Shi
- Department of Biology, Duke University, Durham, NC 27708
| | - Daniel Zhu
- Department of Biology, Duke University, Durham, NC 27708
| | - Andrea Stewart
- Department of Biology, Duke University, Durham, NC 27708
| | - Susan May
- Department of Biology, Duke University, Durham, NC 27708
| |
Collapse
|
21
|
Ho CH, Treisman JE. Specific Isoforms of the Guanine-Nucleotide Exchange Factor dPix Couple Neuromuscular Synapse Growth to Muscle Growth. Dev Cell 2020; 54:117-131.e5. [PMID: 32516570 DOI: 10.1016/j.devcel.2020.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/09/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022]
Abstract
Developmental growth requires coordination between the growth rates of individual tissues and organs. Here, we examine how Drosophila neuromuscular synapses grow to match the size of their target muscles. We show that changes in muscle growth driven by autonomous modulation of insulin receptor signaling produce corresponding changes in synapse size, with each muscle affecting only its presynaptic motor neuron branches. This scaling growth is mechanistically distinct from synaptic plasticity driven by neuronal activity and requires increased postsynaptic differentiation induced by insulin receptor signaling in muscle. We identify the guanine-nucleotide exchange factor dPix as an effector of insulin receptor signaling. Alternatively spliced dPix isoforms that contain a specific exon are necessary and sufficient for postsynaptic differentiation and scaling growth, and their mRNA levels are regulated by insulin receptor signaling. These findings define a mechanism by which the same signaling pathway promotes both autonomous muscle growth and non-autonomous synapse growth.
Collapse
Affiliation(s)
- Cheuk Hei Ho
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
22
|
Kelleher ES, Barbash DA, Blumenstiel JP. Taming the Turmoil Within: New Insights on the Containment of Transposable Elements. Trends Genet 2020; 36:474-489. [PMID: 32473745 DOI: 10.1016/j.tig.2020.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/28/2022]
Abstract
Transposable elements (TEs) are mobile genetic parasites that can exponentially increase their genomic abundance through self-propagation. Classic theoretical papers highlighted the importance of two potentially escalating forces that oppose TE spread: regulated transposition and purifying selection. Here, we review new insights into mechanisms of TE regulation and purifying selection, which reveal the remarkable foresight of these theoretical models. We further highlight emergent connections between transcriptional control enacted by small RNAs and the contribution of TE insertions to structural mutation and host-gene regulation. Finally, we call for increased comparative analysis of TE dynamics and fitness effects, as well as host control mechanisms, to reveal how interconnected forces shape the differential prevalence and distribution of TEs across the tree of life.
Collapse
|
23
|
Genetic Variation and Potential for Resistance Development to the tTA Overexpression Lethal System in Insects. G3 (BETHESDA, MD.) 2020; 10:1271-1281. [PMID: 32019873 PMCID: PMC7144068 DOI: 10.1534/g3.120.400990] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Release of insect pests carrying the dominant lethal tetracycline transactivator (tTA) overexpression system has been proposed as a means for population suppression. High levels of the tTA transcription factor are thought to be toxic due to either transcriptional squelching or interference with protein ubiquitination. Here we utilized the Drosophila melanogaster Genetic Reference Panel (DGRP) to examine the influence of genetic variation on the efficacy of a female-specific tTA overexpression system. The level of female lethality between DGRP lines varied from 11 to 97% with a broad sense heritability of 0.89. A genome-wide association analysis identified 192 allelic variants associated with high or low lethality (P < 10-5), although none were significant when corrected for multiple testing. 151 of the variants fell within 108 genes that were associated with several biological processes including transcription and protein ubiquitination. In four lines with high female lethality, tTA RNA levels were similar or higher than in the parental tTA overexpression strain. In two lines with low lethality, tTA levels were about two fold lower than in the parental strain. However, in two other lines with low lethality, tTA levels were similar or approximately 30% lower. RNAseq analysis identified genes that were up or downregulated in the four low female lethal lines compared to the four high lethal lines. For example, genes associated with RNA processing and rRNA maturation were significantly upregulated in low lethal lines. Our data suggest that standing genetic variation in an insect population could provide multiple mechanisms for resistance to the tTA overexpression system.
Collapse
|
24
|
Sang Y, Gao B, Diaby M, Zong W, Chen C, Shen D, Wang S, Wang Y, Ivics Z, Song C. Incomer, a DD36E family of Tc1/mariner transposons newly discovered in animals. Mob DNA 2019; 10:45. [PMID: 31788035 PMCID: PMC6875036 DOI: 10.1186/s13100-019-0188-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/11/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The Tc1/mariner superfamily might represent the most diverse and widely distributed group of DNA transposons. Several families have been identified; however, exploring the diversity of this superfamily and updating its classification is still ongoing in the life sciences. RESULTS Here we identified a new family of Tc1/mariner transposons, named Incomer (IC), which is close to, but distinct from the known family DD34E/Tc1. ICs have a total length of about 1.2 kb, and harbor a single open reading frame encoding a ~ 346 amino acid transposase with a DD36E motif and flanked by short terminal inverted repeats (TIRs) (22-32 base pairs, bp). This family is absent from prokaryotes, and is mainly distributed among vertebrates (141 species of four classes), including Agnatha (one species of jawless fish), Actinopterygii (132 species of ray-finned fish), Amphibia (four species of frogs), and Mammalia (four species of bats), but have a restricted distribution in invertebrates (four species in Insecta and nine in Arachnida). All ICs in bats (Myotis lucifugus, Eptesicus fuscus, Myotis davidii, and Myotis brandtii) are present as truncated copies in these genomes, and most of them are flanked by relatively long TIRs (51-126 bp). High copy numbers of miniature inverted-repeat transposable elements (MITEs) derived from ICs were also identified in bat genomes. Phylogenetic analysis revealed that ICs are more closely related to DD34E/Tc1 than to other families of Tc1/mariner (e.g., DD34D/mariner and DD × D/pogo), and can be classified into four distinct clusters. The host and IC phylogenies and pairwise distance comparisons between RAG1 genes and all consensus sequences of ICs support the idea that multiple episodes of horizontal transfer (HT) of ICs have occurred in vertebrates. In addition, the discovery of intact transposases, perfect TIRs and target site duplications of ICs suggests that this family may still be active in Insecta, Arachnida, frogs, and fish. CONCLUSIONS Exploring the diversity of Tc1/mariner transposons and revealing their evolutionary profiles will help provide a better understanding of the evolution of DNA transposons and their impact on genomic evolution. Here, a newly discovered family (DD36E/Incomer) of Tc1/mariner transposons is described in animals. It displays a similar structural organization and close relationship with the known DD34E/Tc1 elements, but has a relatively narrow distribution, indicating that DD36E/IC might have originated from the DD34E/Tc1 family. Our data also support the hypothesis of horizontal transfer of IC in vertebrates, even invading one lineage of mammals (bats). This study expands our understanding of the diversity of Tc1/mariner transposons and updates the classification of this superfamily.
Collapse
Affiliation(s)
- Yatong Sang
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Bo Gao
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
- Division of Medical Biotechnology, Paul Ehrlich Institute, 63225 Langen, Germany
| | - Mohamed Diaby
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Wencheng Zong
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Cai Chen
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Dan Shen
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Saisai Wang
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Yali Wang
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, 63225 Langen, Germany
| | - Chengyi Song
- Institute of Animal Mobilome and Genome, College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009 Jiangsu China
| |
Collapse
|
25
|
Protein trap: a new Swiss army knife for geneticists? Mol Biol Rep 2019; 47:1445-1458. [PMID: 31728729 DOI: 10.1007/s11033-019-05181-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/04/2019] [Indexed: 10/25/2022]
Abstract
The protein trap is a powerful tool for genetic and biochemical studies of gene function in the animal kingdom. Although the original protein trap was developed for flies, it can be easily adapted to other multicellular organisms, both known models and ones with an unsequenced genome. The protein trap has been successfully applied to the fruit fly, crustaceans Parhyale hawaiensis, zebrafish, and insect and animal cell cultures. This approach is based on the integration into genes of an artificial exon that carries DNA encoding a fluorescent marker, standardized immunoepitopes, an integrase docking site, and splice acceptor and donor sites. The protein trap for cell cultures additionally contains an antibiotic resistance gene, which facilitates the selection of trapped clones. Resulting chimeric tagged mRNAs can be interfered by dsRNA against GFP (iGFPi-in vivo GFP interference), or the chimeric proteins can be efficiently knocked down by deGradFP technology. Both RNA and protein knockdowns produce a strong loss of function phenotype in tagged cells. The fluorescent and protein affinity tags can be used for tagged protein localisation within the cell and for identifying their binding partners in their native complexes. Insertion into protein trap integrase docking sites allows the replacement of trap contents by any new constructs, including other markers, cell toxins, stop-codons, and binary expression systems such as GAL4/UAS, LexA/LexAop and QF/QUAS, that reliably reflect endogenous gene expression. A distinctive feature of the protein trap approach is that all manipulations with a gene or its product occur only in the endogenous locus, which cannot be achieved by any other method.
Collapse
|
26
|
Garcia-Marques J, Yang CP, Espinosa-Medina I, Mok K, Koyama M, Lee T. Unlimited Genetic Switches for Cell-Type-Specific Manipulation. Neuron 2019; 104:227-238.e7. [PMID: 31395429 DOI: 10.1016/j.neuron.2019.07.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/11/2019] [Accepted: 07/03/2019] [Indexed: 01/23/2023]
Abstract
Gaining independent genetic access to discrete cell types is critical to interrogate their biological functions as well as to deliver precise gene therapy. Transcriptomics has allowed us to profile cell populations with extraordinary precision, revealing that cell types are typically defined by a unique combination of genetic markers. Given the lack of adequate tools to target cell types based on multiple markers, most cell types remain inaccessible to genetic manipulation. Here we present CaSSA, a platform to create unlimited genetic switches based on CRISPR/Cas9 (Ca) and the DNA repair mechanism known as single-strand annealing (SSA). CaSSA allows engineering of independent genetic switches, each responding to a specific gRNA. Expressing multiple gRNAs in specific patterns enables multiplex cell-type-specific manipulations and combinatorial genetic targeting. CaSSA is a new genetic tool that conceptually works as an unlimited number of recombinases and will facilitate genetic access to cell types in diverse organisms.
Collapse
Affiliation(s)
- Jorge Garcia-Marques
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Ching-Po Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | | | - Kent Mok
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Minoru Koyama
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Tzumin Lee
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| |
Collapse
|
27
|
Wolfstetter G, Dahlitz I, Pfeifer K, Töpfer U, Alt JA, Pfeifer DC, Lakes-Harlan R, Baumgartner S, Palmer RH, Holz A. Characterization of Drosophila Nidogen/ entactin reveals roles in basement membrane stability, barrier function and nervous system patterning. Development 2019; 146:dev.168948. [PMID: 30567930 DOI: 10.1242/dev.168948] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022]
Abstract
Basement membranes (BMs) are specialized layers of extracellular matrix (ECM) mainly composed of Laminin, type IV Collagen, Perlecan and Nidogen/entactin (NDG). Recent in vivo studies challenged the initially proposed role of NDG as a major ECM linker molecule by revealing dispensability for viability and BM formation. Here, we report the characterization of the single Ndg gene in Drosophila. Embryonic Ndg expression was primarily observed in mesodermal tissues and the chordotonal organs, whereas NDG protein localized to all BMs. Although loss of Laminin strongly affected BM localization of NDG, Ndg-null mutants exhibited no overt changes in the distribution of BM components. Although Drosophila Ndg mutants were viable, loss of NDG led to ultrastructural BM defects that compromised barrier function and stability in vivo Moreover, loss of NDG impaired larval crawling behavior and reduced responses to vibrational stimuli. Further morphological analysis revealed accompanying defects in the larval peripheral nervous system, especially in the chordotonal organs and the neuromuscular junction (NMJ). Taken together, our analysis suggests that NDG is not essential for BM assembly but mediates BM stability and ECM-dependent neural plasticity during Drosophila development.
Collapse
Affiliation(s)
- Georg Wolfstetter
- Justus-Liebig-Universitaet Giessen, Institut für Allgemeine und Spezielle Zoologie, Allgemeine Zoologie und Entwicklungsbiologie, Stephanstraße 24, 35390 Gießen, Germany.,The Sahlgrenska Academy at the University of Gothenburg, Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Medicinaregatan 9A, 41390 Gothenburg, Sweden
| | - Ina Dahlitz
- Justus-Liebig-Universitaet Giessen, Institut für Allgemeine und Spezielle Zoologie, Allgemeine Zoologie und Entwicklungsbiologie, Stephanstraße 24, 35390 Gießen, Germany
| | - Kathrin Pfeifer
- The Sahlgrenska Academy at the University of Gothenburg, Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Medicinaregatan 9A, 41390 Gothenburg, Sweden
| | - Uwe Töpfer
- Justus-Liebig-Universitaet Giessen, Institut für Allgemeine und Spezielle Zoologie, Allgemeine Zoologie und Entwicklungsbiologie, Stephanstraße 24, 35390 Gießen, Germany
| | - Joscha Arne Alt
- Justus-Liebig-Universitaet Giessen, Institut für Tierphysiologie, Integrative Sinnesphysiologie, Heinrich-Buff-Ring 26, 35392 Gießen, Germany
| | - Daniel Christoph Pfeifer
- Justus-Liebig-Universitaet Giessen, Institut für Allgemeine und Spezielle Zoologie, Allgemeine Zoologie und Entwicklungsbiologie, Stephanstraße 24, 35390 Gießen, Germany
| | - Reinhard Lakes-Harlan
- Justus-Liebig-Universitaet Giessen, Institut für Tierphysiologie, Integrative Sinnesphysiologie, Heinrich-Buff-Ring 26, 35392 Gießen, Germany
| | - Stefan Baumgartner
- Lund University, Department of Experimental Medical Sciences, BMC D10, 22184 Lund, Sweden
| | - Ruth H Palmer
- The Sahlgrenska Academy at the University of Gothenburg, Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Medicinaregatan 9A, 41390 Gothenburg, Sweden
| | - Anne Holz
- Justus-Liebig-Universitaet Giessen, Institut für Allgemeine und Spezielle Zoologie, Allgemeine Zoologie und Entwicklungsbiologie, Stephanstraße 24, 35390 Gießen, Germany
| |
Collapse
|
28
|
Read RD. Pvr receptor tyrosine kinase signaling promotes post-embryonic morphogenesis, and survival of glia and neural progenitor cells in Drosophila. Development 2018; 145:dev.164285. [PMID: 30327326 DOI: 10.1242/dev.164285] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 10/10/2018] [Indexed: 12/30/2022]
Abstract
Stem cells reside in specialized microenvironments, called niches, that regulate their development and the development of their progeny. However, the development and maintenance of niches are poorly understood. In the Drosophila brain, cortex glial cells provide a niche that promotes self-renewal and proliferation of neural stem cell-like cells (neuroblasts). In the central brain, neuroblasts and their progeny control post-embryonic morphogenesis of cortex glia through PDGF-like ligands, and this PDGFR receptor tyrosine kinase (RTK) signaling in cortex glia is required for expression of DE-cadherin, which sustains neuroblasts. Thus, through an RTK-dependent feed-forward loop, neuroblasts and their glial niche actively maintain each other. When the EGFR RTK is constitutively activated in cortex glia, they overexpress PDGF orthologs to stimulate autocrine PDGFR signaling, which uncouples their growth and survival from neuroblasts, and drives neoplastic glial transformation and elimination of neuroblasts. These results provide fundamental insights into glial development and niche regulation, and show that niche-neural stem cell feed-forward signaling becomes hijacked to drive neural tumorigenesis.
Collapse
Affiliation(s)
- Renee D Read
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
29
|
Astigarraga S, Douthit J, Tarnogorska D, Creamer MS, Mano O, Clark DA, Meinertzhagen IA, Treisman JE. Drosophila Sidekick is required in developing photoreceptors to enable visual motion detection. Development 2018; 145:dev.158246. [PMID: 29361567 DOI: 10.1242/dev.158246] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/09/2018] [Indexed: 12/15/2022]
Abstract
The assembly of functional neuronal circuits requires growth cones to extend in defined directions and recognize the correct synaptic partners. Homophilic adhesion between vertebrate Sidekick proteins promotes synapse formation between retinal neurons involved in visual motion detection. We show here that Drosophila Sidekick accumulates in specific synaptic layers of the developing motion detection circuit and is necessary for normal optomotor behavior. Sidekick is required in photoreceptors, but not in their target lamina neurons, to promote the alignment of lamina neurons into columns and subsequent sorting of photoreceptor axons into synaptic modules based on their precise spatial orientation. Sidekick is also localized to the dendrites of the direction-selective T4 and T5 cells, and is expressed in some of their presynaptic partners. In contrast to its vertebrate homologs, Sidekick is not essential for T4 and T5 to direct their dendrites to the appropriate layers or to receive synaptic contacts. These results illustrate a conserved requirement for Sidekick proteins in establishing visual motion detection circuits that is achieved through distinct cellular mechanisms in Drosophila and vertebrates.
Collapse
Affiliation(s)
- Sergio Astigarraga
- Skirball Institute for Biomolecular Medicine and Department of Cell Biology, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jessica Douthit
- Skirball Institute for Biomolecular Medicine and Department of Cell Biology, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Dorota Tarnogorska
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, 1355 Oxford Street, Halifax, NS B3H 4R2, Canada
| | - Matthew S Creamer
- Interdepartmental Neuroscience Program, Yale University, Kline Biology Tower Room 224, 219 Whitney Avenue, New Haven, CT 06511, USA
| | - Omer Mano
- Department of Molecular, Cellular and Developmental Biology, Yale University, Kline Biology Tower Room 224, 219 Whitney Avenue, New Haven, CT 06511, USA
| | - Damon A Clark
- Interdepartmental Neuroscience Program, Yale University, Kline Biology Tower Room 224, 219 Whitney Avenue, New Haven, CT 06511, USA
| | - Ian A Meinertzhagen
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, 1355 Oxford Street, Halifax, NS B3H 4R2, Canada
| | - Jessica E Treisman
- Skirball Institute for Biomolecular Medicine and Department of Cell Biology, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
30
|
Genetic and Neurobiological Analyses of the Noradrenergic-like System in Vulnerability to Sugar Overconsumption Using a Drosophila Model. Sci Rep 2017; 7:17642. [PMID: 29247240 PMCID: PMC5732301 DOI: 10.1038/s41598-017-17760-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/30/2017] [Indexed: 12/22/2022] Open
Abstract
Regular overconsumption of sugar is associated with obesity and type-2 diabetes, but how genetic factors contribute to variable sugar preferences and intake levels remains mostly unclear. Here we provide evidence for the usefulness of a Drosophila larva model to investigate genetic influence on vulnerability to sugar overconsumption. Using genetic and RNA interference approaches, we show that the activity of the Oamb gene, which encodes a receptor for octopamine (OA, the invertebrate homologue of norepinephrine), plays a major role in controlled sugar consumption. Furthermore, Oamb appears to suppress sugar food intake in fed larvae in an acute manner, and neurons expressing this Oamb receptor do not overlap with neurons expressing Octβ3R, another OA receptor previously implicated in hunger-driven exuberant sugar intake. Together, these results suggest that two separate sub-circuits, defined by Oamb and Octβ3R respectively, co-regulate sugar consumption according to changes in energy needs. We propose that the noradrenergic-like system defines an ancient regulatory mechanism for prevention of sugar overload.
Collapse
|
31
|
Rojas-Ríos P, Chartier A, Pierson S, Simonelig M. Aubergine and piRNAs promote germline stem cell self-renewal by repressing the proto-oncogene Cbl. EMBO J 2017; 36:3194-3211. [PMID: 29030484 PMCID: PMC5666619 DOI: 10.15252/embj.201797259] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/30/2017] [Accepted: 09/04/2017] [Indexed: 12/19/2022] Open
Abstract
PIWI proteins play essential roles in germ cells and stem cell lineages. In Drosophila, Piwi is required in somatic niche cells and germline stem cells (GSCs) to support GSC self‐renewal and differentiation. Whether and how other PIWI proteins are involved in GSC biology remains unknown. Here, we show that Aubergine (Aub), another PIWI protein, is intrinsically required in GSCs for their self‐renewal and differentiation. Aub needs to be loaded with piRNAs to control GSC self‐renewal and acts through direct mRNA regulation. We identify the Cbl proto‐oncogene, a regulator of mammalian hematopoietic stem cells, as a novel GSC differentiation factor. Aub stimulates GSC self‐renewal by repressing Cbl mRNA translation and does so in part through recruitment of the CCR4‐NOT complex. This study reveals the role of piRNAs and PIWI proteins in controlling stem cell homeostasis via translational repression and highlights piRNAs as major post‐transcriptional regulators in key developmental decisions.
Collapse
Affiliation(s)
- Patricia Rojas-Ríos
- mRNA Regulation and Development, Institute of Human Genetics, UMR9002 CNRS-Université de Montpellier, Montpellier Cedex 5, France
| | - Aymeric Chartier
- mRNA Regulation and Development, Institute of Human Genetics, UMR9002 CNRS-Université de Montpellier, Montpellier Cedex 5, France
| | - Stéphanie Pierson
- mRNA Regulation and Development, Institute of Human Genetics, UMR9002 CNRS-Université de Montpellier, Montpellier Cedex 5, France
| | - Martine Simonelig
- mRNA Regulation and Development, Institute of Human Genetics, UMR9002 CNRS-Université de Montpellier, Montpellier Cedex 5, France
| |
Collapse
|
32
|
Khandelwal R, Sipani R, Govinda Rajan S, Kumar R, Joshi R. Combinatorial action of Grainyhead, Extradenticle and Notch in regulating Hox mediated apoptosis in Drosophila larval CNS. PLoS Genet 2017; 13:e1007043. [PMID: 29023471 PMCID: PMC5667929 DOI: 10.1371/journal.pgen.1007043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/02/2017] [Accepted: 09/26/2017] [Indexed: 01/09/2023] Open
Abstract
Hox mediated neuroblast apoptosis is a prevalent way to pattern larval central nervous system (CNS) by different Hox genes, but the mechanism of this apoptosis is not understood. Our studies with Abdominal-A (Abd-A) mediated larval neuroblast (pNB) apoptosis suggests that AbdA, its cofactor Extradenticle (Exd), a helix-loop-helix transcription factor Grainyhead (Grh), and Notch signaling transcriptionally contribute to expression of RHG family of apoptotic genes. We find that Grh, AbdA, and Exd function together at multiple motifs on the apoptotic enhancer. In vivo mutagenesis of these motifs suggest that they are important for the maintenance of the activity of the enhancer rather than its initiation. We also find that Exd function is independent of its known partner homothorax in this apoptosis. We extend some of our findings to Deformed expressing region of sub-esophageal ganglia where pNBs undergo a similar Hox dependent apoptosis. We propose a mechanism where common players like Exd-Grh-Notch work with different Hox genes through region specific enhancers to pattern respective segments of larval central nervous system. Specification of the head to tail axis of the developing central nervous system is carried out by Hox genes. Hox mediated programmed cell death of the neural progenitor cells plays an important role in specification of this axis, but the molecular mechanism of this phenomenon is not well understood. We have studied this phenomenon in abdominal and subesophageal regions of larval central nervous system of Drosophila. We find that different Hox genes use a combination of common players (Extradenticle, Grainyhead and Notch) but employ region specific enhancers to cause progenitor cell death in different segments of developing central nervous system.
Collapse
Affiliation(s)
- Risha Khandelwal
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- Graduate Studies, Manipal University, Manipal, India
| | - Rashmi Sipani
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- Graduate Studies, Manipal University, Manipal, India
| | - Sriivatsan Govinda Rajan
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
| | - Raviranjan Kumar
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- Graduate Studies, Manipal University, Manipal, India
| | - Rohit Joshi
- Laboratory of Drosophila Neural Development, Centre for DNA Fingerprinting and Diagnostics (CDFD), Tuljaguda Complex, Nampally, Hyderabad, India
- * E-mail: ,
| |
Collapse
|
33
|
Kanca O, Bellen HJ, Schnorrer F. Gene Tagging Strategies To Assess Protein Expression, Localization, and Function in Drosophila. Genetics 2017; 207:389-412. [PMID: 28978772 PMCID: PMC5629313 DOI: 10.1534/genetics.117.199968] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/13/2017] [Indexed: 01/15/2023] Open
Abstract
Analysis of gene function in complex organisms relies extensively on tools to detect the cellular and subcellular localization of gene products, especially proteins. Typically, immunostaining with antibodies provides these data. However, due to cost, time, and labor limitations, generating specific antibodies against all proteins of a complex organism is not feasible. Furthermore, antibodies do not enable live imaging studies of protein dynamics. Hence, tagging genes with standardized immunoepitopes or fluorescent tags that permit live imaging has become popular. Importantly, tagging genes present in large genomic clones or at their endogenous locus often reports proper expression, subcellular localization, and dynamics of the encoded protein. Moreover, these tagging approaches allow the generation of elegant protein removal strategies, standardization of visualization protocols, and permit protein interaction studies using mass spectrometry. Here, we summarize available genomic resources and techniques to tag genes and discuss relevant applications that are rarely, if at all, possible with antibodies.
Collapse
Affiliation(s)
- Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Howard Hughes Medical Institute, Houston, Texas 77030
| | - Frank Schnorrer
- Developmental Biology Institute of Marseille (IBDM), UMR 7288, CNRS, Aix-Marseille Université, 13288, France
| |
Collapse
|
34
|
Highfill CA, Tran JH, Nguyen SKT, Moldenhauer TR, Wang X, Macdonald SJ. Naturally Segregating Variation at Ugt86Dd Contributes to Nicotine Resistance in Drosophila melanogaster. Genetics 2017; 207:311-325. [PMID: 28743761 PMCID: PMC5586381 DOI: 10.1534/genetics.117.300058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/24/2017] [Indexed: 12/16/2022] Open
Abstract
Identifying the sequence polymorphisms underlying complex trait variation is a key goal of genetics research, since knowing the precise causative molecular events allows insight into the pathways governing trait variation. Genetic analysis of complex traits in model systems regularly starts by constructing QTL maps, but generally fails to identify causative sequence polymorphisms. Previously we mapped a series of QTL contributing to resistance to nicotine in a Drosophila melanogaster multiparental mapping resource and here use a battery of functional tests to resolve QTL to the molecular level. One large-effect QTL resided over a cluster of UDP-glucuronosyltransferases, and quantitative complementation tests using deficiencies eliminating subsets of these detoxification genes revealed allelic variation impacting resistance. RNAseq showed that Ugt86Dd had significantly higher expression in genotypes that are more resistant to nicotine, and anterior midgut-specific RNA interference (RNAi) of this gene reduced resistance. We discovered a segregating 22-bp frameshift deletion in Ugt86Dd, and accounting for the InDel during mapping largely eliminates the QTL, implying the event explains the bulk of the effect of the mapped locus. CRISPR/Cas9 editing of a relatively resistant genotype to generate lesions in Ugt86Dd that recapitulate the naturally occurring putative loss-of-function allele, leads to a large reduction in resistance. Despite this major effect of the deletion, the allele appears to be very rare in wild-caught populations and likely explains only a small fraction of the natural variation for the trait. Nonetheless, this putatively causative coding InDel can be a launchpad for future mechanistic exploration of xenobiotic detoxification.
Collapse
Affiliation(s)
- Chad A Highfill
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Jonathan H Tran
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Samantha K T Nguyen
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Taylor R Moldenhauer
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Xiaofei Wang
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Stuart J Macdonald
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
- Center for Computational Biology, University of Kansas, Lawrence, Kansas 66047
| |
Collapse
|
35
|
Neukomm LJ, Burdett TC, Seeds AM, Hampel S, Coutinho-Budd JC, Farley JE, Wong J, Karadeniz YB, Osterloh JM, Sheehan AE, Freeman MR. Axon Death Pathways Converge on Axundead to Promote Functional and Structural Axon Disassembly. Neuron 2017; 95:78-91.e5. [PMID: 28683272 DOI: 10.1016/j.neuron.2017.06.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/25/2017] [Accepted: 06/19/2017] [Indexed: 01/04/2023]
Abstract
Axon degeneration is a hallmark of neurodegenerative disease and neural injury. Axotomy activates an intrinsic pro-degenerative axon death signaling cascade involving loss of the NAD+ biosynthetic enzyme Nmnat/Nmnat2 in axons, activation of dSarm/Sarm1, and subsequent Sarm-dependent depletion of NAD+. Here we identify Axundead (Axed) as a mediator of axon death. axed mutants suppress axon death in several types of axons for the lifespan of the fly and block the pro-degenerative effects of activated dSarm in vivo. Neurodegeneration induced by loss of the sole fly Nmnat ortholog is also fully blocked by axed, but not dsarm, mutants. Thus, pro-degenerative pathways activated by dSarm signaling or Nmnat elimination ultimately converge on Axed. Remarkably, severed axons morphologically preserved by axon death pathway mutations remain integrated in circuits and able to elicit complex behaviors after stimulation, indicating that blockade of axon death signaling results in long-term functional preservation of axons.
Collapse
Affiliation(s)
- Lukas J Neukomm
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA.
| | - Thomas C Burdett
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Andrew M Seeds
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Stefanie Hampel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jaeda C Coutinho-Budd
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jonathan E Farley
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jack Wong
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yonca B Karadeniz
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jeannette M Osterloh
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Amy E Sheehan
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
36
|
Shank Modulates Postsynaptic Wnt Signaling to Regulate Synaptic Development. J Neurosci 2017; 36:5820-32. [PMID: 27225771 DOI: 10.1523/jneurosci.4279-15.2016] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/25/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Prosap/Shank scaffolding proteins regulate the formation, organization, and plasticity of excitatory synapses. Mutations in SHANK family genes are implicated in autism spectrum disorder and other neuropsychiatric conditions. However, the molecular mechanisms underlying Shank function are not fully understood, and no study to date has examined the consequences of complete loss of all Shank proteins in vivo Here we characterize the single Drosophila Prosap/Shank family homolog. Shank is enriched at the postsynaptic membrane of glutamatergic neuromuscular junctions and controls multiple parameters of synapse biology in a dose-dependent manner. Both loss and overexpression of Shank result in defects in synaptic bouton number and maturation. We find that Shank regulates a noncanonical Wnt signaling pathway in the postsynaptic cell by modulating the internalization of the Wnt receptor Fz2. This study identifies Shank as a key component of synaptic Wnt signaling, defining a novel mechanism for how Shank contributes to synapse maturation during neuronal development. SIGNIFICANCE STATEMENT Haploinsufficiency for SHANK3 is one of the most prevalent monogenic causes of autism spectrum disorder, making it imperative to understand how the Shank family regulates neurodevelopment and synapse function. We created the first animal model lacking all Shank proteins and used the Drosophila neuromuscular junction, a model glutamatergic synapse, to characterize the role of Shank at synapses. We identified a novel function of Shank in synapse maturation via regulation of Wnt signaling in the postsynaptic cell.
Collapse
|
37
|
The Drosophila DAXX-Like Protein (DLP) Cooperates with ASF1 for H3.3 Deposition and Heterochromatin Formation. Mol Cell Biol 2017; 37:MCB.00597-16. [PMID: 28320872 DOI: 10.1128/mcb.00597-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/09/2017] [Indexed: 01/22/2023] Open
Abstract
Histone variants are nonallelic isoforms of canonical histones, and they are deposited, in contrast to canonical histones, in a replication-independent (RI) manner. RI deposition of H3.3, a histone variant from the H3.3 family, is mediated in mammals by distinct pathways involving either the histone regulator A (HIRA) complex or the death-associated protein (DAXX)/α-thalassemia X-linked mental retardation protein (ATRX) complex. Here, we investigated the function of the Drosophila DAXX-like protein (DLP) by using both fly genetic approaches and protein biochemistry. DLP specifically interacts with H3.3 and shows a prominent localization on the base of the X chromosome, where it appears to act in concert with XNP, the Drosophila homolog of ATRX, in heterochromatin assembly and maintenance. The functional association between DLP and XNP is further supported by a series of experiments that illustrate genetic interactions and the DLP-XNP-dependent localization of specific chromosomal proteins. In addition, DLP both participates in the RI deposition of H3.3 and associates with anti-silencing factor 1 (ASF1). We suggest, in agreement with a recently proposed model, that DLP and ASF1 are part of a predeposition complex, which is recruited by XNP and is necessary to prevent DNA exposure in the nucleus.
Collapse
|
38
|
Ren X, Holsteens K, Li H, Sun J, Zhang Y, Liu LP, Liu Q, Ni JQ. Genome editing in Drosophila melanogaster: from basic genome engineering to the multipurpose CRISPR-Cas9 system. SCIENCE CHINA-LIFE SCIENCES 2017; 60:476-489. [PMID: 28527116 DOI: 10.1007/s11427-017-9029-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/05/2017] [Indexed: 12/16/2022]
Abstract
Nowadays, genome editing tools are indispensable for studying gene function in order to increase our knowledge of biochemical processes and disease mechanisms. The extensive availability of mutagenesis and transgenesis tools make Drosophila melanogaster an excellent model organism for geneticists. Early mutagenesis tools relied on chemical or physical methods, ethyl methane sulfonate (EMS) and X-rays respectively, to randomly alter DNA at a nucleotide or chromosomal level. Since the discovery of transposable elements and the availability of the complete fly genome, specific genome editing tools, such as P-elements, zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), have undergone rapid development. Currently, one of the leading and most effective contemporary tools is the CRISPR-cas9 system made popular because of its low cost, effectiveness, specificity and simplicity of use. This review briefly addresses the most commonly used mutagenesis and transgenesis tools in Drosophila, followed by an in-depth review of the multipurpose CRISPR-Cas9 system and its current applications.
Collapse
Affiliation(s)
- Xingjie Ren
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Kristof Holsteens
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Haiyi Li
- French International School of Hong Kong, Hong Kong SAR, 999000, China
| | - Jin Sun
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yifan Zhang
- Department of Biology, University of California, San Diego, 92093, USA
| | - Lu-Ping Liu
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Qingfei Liu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Jian-Quan Ni
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
39
|
Tan FHP, Azzam G. Drosophila melanogaster: Deciphering Alzheimer's Disease. Malays J Med Sci 2017; 24:6-20. [PMID: 28894399 PMCID: PMC5566057 DOI: 10.21315/mjms2017.24.2.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 02/21/2017] [Indexed: 09/29/2022] Open
Abstract
Alzheimer's disease (AD) is the most widespread neurodegenerative disorder worldwide. Its pathogenesis involves two hallmarks: aggregation of amyloid beta (Aβ) and occurrence of neurofibrillary tangles (NFTs). The mechanism behind the disease is still unknown. This has prompted the use of animal models to mirror the disease. The fruit fly, Drosophila melanogaster has garnered considerable attention as an organism to recapitulate human disorders. With the ability to monopolise a multitude of traditional and novel genetic tools, Drosophila is ideal for studying not only cellular aspects but also physiological and behavioural traits of human neurodegenerative diseases. Here, we discuss the use of the Drosophila model in understanding AD pathology and the insights gained in discovering drug therapies for AD.
Collapse
Affiliation(s)
- Florence Hui Ping Tan
- School of Biological Sciences, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
| | - Ghows Azzam
- School of Biological Sciences, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
| |
Collapse
|
40
|
Myachina F, Bosshardt F, Bischof J, Kirschmann M, Lehner CF. Drosophila beta-tubulin 97EF is upregulated at low temperature and stabilizes microtubules. Development 2017; 144:4573-4587. [DOI: 10.1242/dev.156109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/20/2017] [Indexed: 12/22/2022]
Abstract
Cells in ectotherms function normally within an often wide temperature range. As temperature dependence is not uniform across all the distinct biological processes, acclimation presumably requires complex regulation. The molecular mechanisms coping with the disruptive effects of temperature variation are still poorly understood. Interestingly, one of five different beta-tubulin paralogs, betaTub97EF, was among the genes up-regulated at low temperature in cultured Drosophila cells. As microtubules are known to be cold-sensitive, we analyzed whether betaTub97EF protects microtubules at low temperatures. During development at the optimal temperature (25°C), betaTub97EF was expressed in a tissue-specific pattern primarily in the gut. There, as well as in hemocytes, expression was increased at low temperature (14°C). While betaTub97EF mutants were viable and fertile at 25°C, their sensitivity within the well-tolerated range was slightly enhanced during embryogenesis specifically at low temperatures. Changing beta-tubulin isoform ratios in hemocytes demonstrated that beta-Tubulin 97EF has a pronounced microtubule stabilizing effect. Moreover, betaTub97EF is required for normal microtubule stability in the gut. These results suggest that betaTub97EF up-regulation at low temperature contributes to acclimation by stabilizing microtubules.
Collapse
Affiliation(s)
- Faina Myachina
- Institute of Molecular Life Sciences (IMLS), University of Zurich, 8057 Zurich, Switzerland
| | - Fritz Bosshardt
- Institute of Molecular Life Sciences (IMLS), University of Zurich, 8057 Zurich, Switzerland
| | - Johannes Bischof
- Institute of Molecular Life Sciences (IMLS), University of Zurich, 8057 Zurich, Switzerland
| | - Moritz Kirschmann
- Center for Microscopy and Image Analysis, University of Zurich, 8057 Zurich, Switzerland
| | - Christian F. Lehner
- Institute of Molecular Life Sciences (IMLS), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
41
|
Fogle KJ, Hertzler JI, Shon JH, Palladino MJ. The ATP-sensitive K channel is seizure protective and required for effective dietary therapy in a model of mitochondrial encephalomyopathy. J Neurogenet 2016; 30:247-258. [PMID: 27868454 DOI: 10.1080/01677063.2016.1252765] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Effective therapies are lacking for mitochondrial encephalomyopathies (MEs). MEs are devastating diseases that predominantly affect the energy-demanding tissues of the nervous system and muscle, causing symptoms such as seizures, cardiomyopathy, and neuro- and muscular degeneration. Even common anti-epileptic drugs which are frequently successful in ameliorating seizures in other diseases tend to have a lower success rate in ME, highlighting the need for novel drug targets, especially those that may couple metabolic sensitivity to neuronal excitability. Furthermore, alternative epilepsy therapies such as dietary modification are gaining in clinical popularity but have not been thoroughly studied in ME. Using the Drosophila ATP61 model of ME, we have studied dietary therapy throughout disease progression and found that it is highly effective against the seizures of ME, especially a high fat/ketogenic diet, and that the benefits are dependent upon a functional KATP channel complex. Further experiments with KATP show that it is seizure-protective in this model, and that pharmacological promotion of its open state also ameliorates seizures. These studies represent important steps forward in the development of novel therapies for a class of diseases that is notoriously difficult to treat, and lay the foundation for mechanistic studies of currently existing therapies in the context of metabolic disease.
Collapse
Affiliation(s)
- Keri J Fogle
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - J Ian Hertzler
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Joy H Shon
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| | - Michael J Palladino
- a Department of Pharmacology & Chemical Biology , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Pittsburgh Institute for Neurodegenerative Diseases (PIND), University of Pittsburgh School of Medicine , Pittsburgh , PA , USA
| |
Collapse
|
42
|
Vedelek V, Laurinyecz B, Kovács AL, Juhász G, Sinka R. Testis-Specific Bb8 Is Essential in the Development of Spermatid Mitochondria. PLoS One 2016; 11:e0161289. [PMID: 27529784 PMCID: PMC4986964 DOI: 10.1371/journal.pone.0161289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/02/2016] [Indexed: 11/18/2022] Open
Abstract
Mitochondria are essential organelles of developing spermatids in Drosophila, which undergo dramatic changes in size and shape after meiotic division, where mitochondria localized in the cytoplasm, migrate near the nucleus, aggregate, fuse and create the Nebenkern. During spermatid elongation the two similar mitochondrial derivatives of the Nebenkern start to elongate parallel to the axoneme. One of the elongated mitochondrial derivatives starts to lose volume and becomes the minor mitochondrial derivative, while the other one accumulates paracrystalline and becomes the major mitochondrial derivative. Proteins and intracellular environment that are responsible for cyst elongation and paracrystalline formation in the major mitochondrial derivative need to be identified. In this work we investigate the function of the testis specific big bubble 8 (bb8) gene during spermatogenesis. We show that a Minos element insertion in bb8 gene, a predicted glutamate dehydrogenase, causes recessive male sterility. We demonstrate bb8 mRNA enrichment in spermatids and the mitochondrial localisation of Bb8 protein during spermatogenesis. We report that megamitochondria develop in the homozygous mutant testes, in elongating spermatids. Ultrastructural analysis of the cross section of elongated spermatids shows enlarged mitochondria and the production of paracrystalline in both major and minor mitochondrial derivatives. Our results suggest that the Bb8 protein and presumably glutamate metabolism has a crucial role in the normal development and establishment of the identity of the mitochondrial derivatives during spermatid elongation.
Collapse
Affiliation(s)
- Viktor Vedelek
- Department of Genetics, University of Szeged, Szeged, Hungary
| | | | - Attila L Kovács
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Gábor Juhász
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Rita Sinka
- Department of Genetics, University of Szeged, Szeged, Hungary
| |
Collapse
|
43
|
Lin CC, Potter CJ. Editing Transgenic DNA Components by Inducible Gene Replacement in Drosophila melanogaster. Genetics 2016; 203:1613-28. [PMID: 27334272 PMCID: PMC4981265 DOI: 10.1534/genetics.116.191783] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/07/2016] [Indexed: 11/18/2022] Open
Abstract
Gene conversions occur when genomic double-strand DNA breaks (DSBs) trigger unidirectional transfer of genetic material from a homologous template sequence. Exogenous or mutated sequence can be introduced through this homology-directed repair (HDR). We leveraged gene conversion to develop a method for genomic editing of existing transgenic insertions in Drosophila melanogaster The clustered regularly-interspaced palindromic repeats (CRISPR)/Cas9 system is used in the H: omology A: ssisted C: RISPR K: nock-in (HACK) method to induce DSBs in a GAL4 transgene, which is repaired by a single-genomic transgenic construct containing GAL4 homologous sequences flanking a T2A-QF2 cassette. With two crosses, this technique converts existing GAL4 lines, including enhancer traps, into functional QF2 expressing lines. We used HACK to convert the most commonly-used GAL4 lines (labeling tissues such as neurons, fat, glia, muscle, and hemocytes) to QF2 lines. We also identified regions of the genome that exhibited differential efficiencies of HDR. The HACK technique is robust and readily adaptable for targeting and replacement of other genomic sequences, and could be a useful approach to repurpose existing transgenes as new genetic reagents become available.
Collapse
Affiliation(s)
- Chun-Chieh Lin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Christopher J Potter
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| |
Collapse
|
44
|
Manivannan SN, Simcox A. Targeted genetics in Drosophila cell lines: Inserting single transgenes in vitro. Fly (Austin) 2016; 10:134-41. [PMID: 27261098 PMCID: PMC4970541 DOI: 10.1080/19336934.2016.1191716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/14/2016] [Indexed: 12/31/2022] Open
Abstract
A long-standing problem with analyzing transgene expression in tissue-culture cells is the variation caused by random integration of different copy numbers of transfected transgenes. In mammalian cells, single transgenes can be inserted by homologous recombination but this process is inefficient in Drosophila cells. To tackle this problem, our group, and the Cherbas group, used recombination-mediated cassette exchange (RMCE) to introduce single-copy transgenes into specific locations in the Drosophila genome. In both cases, ϕC31 was used to catalyze recombination between its target sequences attP in the genome, and attB flanking the donor sequence. We generated cell lines de novo with a single attP-flanked cassette for recombination, whereas, Cherbas et al. introduced a single attP-flanked cassette into existing cell lines. In both approaches, a 2-drug selection scheme was used to select for cells with a single copy of the donor sequence inserted by RMCE and against cells with random integration of multiple copies. Here we describe the general advantages of using RMCE to introduce genes into fly cells, the different attributes of the 2 methods, and how future work could make use of other recombinases and CRISPR/Cas9 genome editing to further enable genetic manipulation of Drosophila cells in vitro.
Collapse
Affiliation(s)
| | - Amanda Simcox
- Department of Molecular Genetics, The Ohio State University, Columbus, OH
| |
Collapse
|
45
|
Kim M, Semple I, Kim B, Kiers A, Nam S, Park HW, Park H, Ro SH, Kim JS, Juhász G, Lee JH. Drosophila Gyf/GRB10 interacting GYF protein is an autophagy regulator that controls neuron and muscle homeostasis. Autophagy 2016; 11:1358-72. [PMID: 26086452 DOI: 10.1080/15548627.2015.1063766] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Autophagy is an essential process for eliminating ubiquitinated protein aggregates and dysfunctional organelles. Defective autophagy is associated with various degenerative diseases such as Parkinson disease. Through a genetic screening in Drosophila, we identified CG11148, whose product is orthologous to GIGYF1 (GRB10-interacting GYF protein 1) and GIGYF2 in mammals, as a new autophagy regulator; we hereafter refer to this gene as Gyf. Silencing of Gyf completely suppressed the effect of Atg1-Atg13 activation in stimulating autophagic flux and inducing autophagic eye degeneration. Although Gyf silencing did not affect Atg1-induced Atg13 phosphorylation or Atg6-Pi3K59F (class III PtdIns3K)-dependent Fyve puncta formation, it inhibited formation of Atg13 puncta, suggesting that Gyf controls autophagy through regulating subcellular localization of the Atg1-Atg13 complex. Gyf silencing also inhibited Atg1-Atg13-induced formation of Atg9 puncta, which is accumulated upon active membrane trafficking into autophagosomes. Gyf-null mutants also exhibited substantial defects in developmental or starvation-induced accumulation of autophagosomes and autolysosomes in the larval fat body. Furthermore, heads and thoraxes from Gyf-null adults exhibited strongly reduced expression of autophagosome-associated Atg8a-II compared to wild-type (WT) tissues. The decrease in Atg8a-II was directly correlated with an increased accumulation of ubiquitinated proteins and dysfunctional mitochondria in neuron and muscle, which together led to severe locomotor defects and early mortality. These results suggest that Gyf-mediated autophagy regulation is important for maintaining neuromuscular homeostasis and preventing degenerative pathologies of the tissues. Since human mutations in the GIGYF2 locus were reported to be associated with a type of familial Parkinson disease, the homeostatic role of Gyf-family proteins is likely to be evolutionarily conserved.
Collapse
Affiliation(s)
- Myungjin Kim
- a Department of Molecular and Integrative Physiology ; University of Michigan ; Ann Arbor , MI USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Rohde PD, Madsen LS, Neumann Arvidson SM, Loeschcke V, Demontis D, Kristensen TN. Testing candidate genes for attention-deficit/hyperactivity disorder in fruit flies using a high throughput assay for complex behavior. Fly (Austin) 2016; 10:25-34. [PMID: 26954609 DOI: 10.1080/19336934.2016.1158365] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Fruit flies are important model organisms for functional testing of candidate genes in multiple disciplines, including the study of human diseases. Here we use a high-throughput locomotor activity assay to test the response on activity behavior of gene disruption in Drosophila melanogaster. The aim was to investigate the impact of disruption of 14 candidate genes for human attention-deficit/hyperactivity disorder (ADHD) on fly behavior. By obtaining a range of correlated measures describing the space of variables for behavioral activity we show, that some mutants display similar phenotypic responses, and furthermore, that the genes disrupted in those mutants had common molecular functions; namely processes related to cGMP activity, cation channels and serotonin receptors. All but one of the candidate genes resulted in aberrant behavioral activity, suggesting involvement of these genes in behavioral activity in fruit flies. Results provide additional support for the investigated genes being risk candidate genes for ADHD in humans.
Collapse
Affiliation(s)
- Palle Duun Rohde
- a Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University , Tjele , Denmark.,b The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH , Aarhus , Denmark.,c Center for Integrative Sequencing, iSEQ, Aarhus University , Aarhus , Denmark
| | - Lisbeth Strøm Madsen
- d Section of Biology and Environmental Science, Department of Chemistry and Bioscience, Aalborg University , Aalborg , Denmark
| | - Sandra Marie Neumann Arvidson
- d Section of Biology and Environmental Science, Department of Chemistry and Bioscience, Aalborg University , Aalborg , Denmark
| | - Volker Loeschcke
- e Section for Genetics, Ecology and Evolution, Department of Bioscience, Aarhus University , Aarhus , Denmark
| | - Ditte Demontis
- b The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH , Aarhus , Denmark.,c Center for Integrative Sequencing, iSEQ, Aarhus University , Aarhus , Denmark.,f Department of Biomedicine , Aarhus University , Aarhus , Denmark
| | - Torsten Nygaard Kristensen
- d Section of Biology and Environmental Science, Department of Chemistry and Bioscience, Aalborg University , Aalborg , Denmark
| |
Collapse
|
47
|
Frost B, Bardai FH, Feany MB. Lamin Dysfunction Mediates Neurodegeneration in Tauopathies. Curr Biol 2015; 26:129-36. [PMID: 26725200 DOI: 10.1016/j.cub.2015.11.039] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/19/2015] [Accepted: 11/11/2015] [Indexed: 10/22/2022]
Abstract
The filamentous meshwork formed by the lamin nucleoskeleton provides a scaffold for the anchoring of highly condensed heterochromatic DNA to the nuclear envelope, thereby establishing the three-dimensional architecture of the genome [1]. Insight into the importance of lamins to cellular viability can be gleaned from laminopathies, severe disorders caused by mutations in genes encoding lamins. A cellular consequence of lamin dysfunction in laminopathies is relaxation of heterochromatic DNA [1]. Similarly, we have recently reported the widespread relaxation of heterochromatin in tauopathies [1]: age-related progressive neurodegenerative disorders, including Alzheimer's disease, that are pathologically characterized by aggregates of phosphorylated tau protein in the brain [2, 3]. Here we demonstrate that acquired lamin misregulation though aberrant cytoskeletal-nucleoskeletal coupling promotes relaxation of heterochromatin and neuronal death in an in vivo model of neurodegenerative tauopathy. Genetic manipulation of lamin function significantly modifies neurodegeneration in vivo, demonstrating that lamin pathology plays a causal role in tau-mediated neurotoxicity. We show that lamin dysfunction is conserved in human tauopathy, as super-resolution microscopy reveals a significantly disrupted nuclear lamina in postmortem tissue from human Alzheimer's disease brain. Our study provides strong evidence that tauopathies are neurodegenerative laminopathies and identifies a new pathway mediating neuronal death in currently untreatable human neurodegenerative disorders, including Alzheimer's disease.
Collapse
Affiliation(s)
- Bess Frost
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Farah H Bardai
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Bharadwaj R, Cunningham KM, Zhang K, Lloyd TE. FIG4 regulates lysosome membrane homeostasis independent of phosphatase function. Hum Mol Genet 2015; 25:681-92. [PMID: 26662798 DOI: 10.1093/hmg/ddv505] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 12/07/2015] [Indexed: 12/31/2022] Open
Abstract
FIG4 is a phosphoinositide phosphatase that is mutated in several diseases including Charcot-Marie-Tooth Disease 4J (CMT4J) and Yunis-Varon syndrome (YVS). To investigate the mechanism of disease pathogenesis, we generated Drosophila models of FIG4-related diseases. Fig4 null mutant animals are viable but exhibit marked enlargement of the lysosomal compartment in muscle cells and neurons, accompanied by an age-related decline in flight ability. Transgenic animals expressing Drosophila Fig4 missense mutations corresponding to human pathogenic mutations can partially rescue lysosomal expansion phenotypes, consistent with these mutations causing decreased FIG4 function. Interestingly, Fig4 mutations predicted to inactivate FIG4 phosphatase activity rescue lysosome expansion phenotypes, and mutations in the phosphoinositide (3) phosphate kinase Fab1 that performs the reverse enzymatic reaction also causes a lysosome expansion phenotype. Since FIG4 and FAB1 are present together in the same biochemical complex, these data are consistent with a model in which FIG4 serves a phosphatase-independent biosynthetic function that is essential for lysosomal membrane homeostasis. Lysosomal phenotypes are suppressed by genetic inhibition of Rab7 or the HOPS complex, demonstrating that FIG4 functions after endosome-to-lysosome fusion. Furthermore, disruption of the retromer complex, implicated in recycling from the lysosome to Golgi, does not lead to similar phenotypes as Fig4, suggesting that the lysosomal defects are not due to compromised retromer-mediated recycling of endolysosomal membranes. These data show that FIG4 plays a critical noncatalytic function in maintaining lysosomal membrane homeostasis, and that this function is disrupted by mutations that cause CMT4J and YVS.
Collapse
Affiliation(s)
| | | | | | - Thomas E Lloyd
- Department of Neurology, and The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
49
|
Garelli A, Heredia F, Casimiro AP, Macedo A, Nunes C, Garcez M, Dias ARM, Volonte YA, Uhlmann T, Caparros E, Koyama T, Gontijo AM. Dilp8 requires the neuronal relaxin receptor Lgr3 to couple growth to developmental timing. Nat Commun 2015; 6:8732. [PMID: 26510564 PMCID: PMC4640092 DOI: 10.1038/ncomms9732] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/25/2015] [Indexed: 11/18/2022] Open
Abstract
How different organs in the body sense growth perturbations in distant tissues to coordinate their size during development is poorly understood. Here we mutate an invertebrate orphan relaxin receptor gene, the Drosophila Leucine-rich repeat-containing G protein-coupled receptor 3 (Lgr3), and find body asymmetries similar to those found in insulin-like peptide 8 (dilp8) mutants, which fail to coordinate growth with developmental timing. Indeed, mutation or RNA intereference (RNAi) against Lgr3 suppresses the delay in pupariation induced by imaginal disc growth perturbation or ectopic Dilp8 expression. By tagging endogenous Lgr3 and performing cell type-specific RNAi, we map this Lgr3 activity to a new subset of CNS neurons, four of which are a pair of bilateral pars intercerebralis Lgr3-positive (PIL) neurons that respond specifically to ectopic Dilp8 by increasing cAMP-dependent signalling. Our work sheds new light on the function and evolution of relaxin receptors and reveals a novel neuroendocrine circuit responsive to growth aberrations. The orphan ligand Dilp8 has been shown to coordinate growth and developmental timing in Drosophila. Here, using Gal4 drivers and CRISPR/Cas9 approaches, Garelli et al. identify a role for relaxin-like receptor Lgr3 in regulating the Dilp8 developmental delay pathway.
Collapse
Affiliation(s)
- Andres Garelli
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal.,Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), CONICET and Universidad Nacional del Sur, Camino La Carrindanga km7, Bahía Blanca B8000 FWB, Argentina
| | - Fabiana Heredia
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Andreia P Casimiro
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Andre Macedo
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Catarina Nunes
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Marcia Garcez
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Angela R Mantas Dias
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Yanel A Volonte
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), CONICET and Universidad Nacional del Sur, Camino La Carrindanga km7, Bahía Blanca B8000 FWB, Argentina
| | - Thomas Uhlmann
- Dualsystems Biotech AG, Grabenstrasse 11a, Schlieren CH-8952, Switzerland
| | - Esther Caparros
- Departamento de Medicina Clínica, Facultad de Medicina, Universidad Miguel Hernández, Ctra. Alicante-Valencia, km 87, San Juan, Alicante 03550, Spain
| | - Takashi Koyama
- Development, Evolution and the Environment Laboratory, Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| | - Alisson M Gontijo
- Integrative Biomedicine Laboratory, CEDOC-Chronic Diseases Research Center, NOVA Medical School
- Faculdade de Ciencias Medicas, NOVA University of Lisbon, Campus do IGC, Rua da Quinta Grande, 6, Oeiras 2780-156, Portugal
| |
Collapse
|
50
|
Hagel KR, Beriont J, Tessier CR. Drosophila Cbp53E Regulates Axon Growth at the Neuromuscular Junction. PLoS One 2015; 10:e0132636. [PMID: 26167908 PMCID: PMC4500412 DOI: 10.1371/journal.pone.0132636] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 06/16/2015] [Indexed: 11/19/2022] Open
Abstract
Calcium is a primary second messenger in all cells that functions in processes ranging from cellular proliferation to synaptic transmission. Proper regulation of calcium is achieved through numerous mechanisms involving channels, sensors, and buffers notably containing one or more EF-hand calcium binding domains. The Drosophila genome encodes only a single 6 EF-hand domain containing protein, Cbp53E, which is likely the prototypic member of a small family of related mammalian proteins that act as calcium buffers and calcium sensors. Like the mammalian homologs, Cbp53E is broadly though discretely expressed throughout the nervous system. Despite the importance of calcium in neuronal function and growth, nothing is known about Cbp53E's function in neuronal development. To address this deficiency, we generated novel null alleles of Drosophila Cbp53E and examined neuronal development at the well-characterized larval neuromuscular junction. Loss of Cbp53E resulted in increases in axonal branching at both peptidergic and glutamatergic neuronal terminals. This overgrowth could be completely rescued by expression of exogenous Cbp53E. Overexpression of Cbp53E, however, only affected the growth of peptidergic neuronal processes. These findings indicate that Cbp53E plays a significant role in neuronal growth and suggest that it may function in both local synaptic and global cellular mechanisms.
Collapse
Affiliation(s)
- Kimberly R. Hagel
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Jane Beriont
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Charles R. Tessier
- Department of Medical and Molecular Genetics, Indiana University School of Medicine-South Bend, South Bend, Indiana, United States of America
- * E-mail:
| |
Collapse
|