101
|
Vakaloglou KM, Mouratidou M, Keramidioti A, Zervas CG. Differential Expression of Drosophila Transgelins Throughout Development. Front Cell Dev Biol 2021; 9:648568. [PMID: 34322481 PMCID: PMC8311604 DOI: 10.3389/fcell.2021.648568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/01/2021] [Indexed: 11/26/2022] Open
Abstract
Transgelins are a conserved family of actin-binding proteins involved in cytoskeletal remodeling, cell contractility, and cell shape. In both mammals and Drosophila, three genes encode transgelin proteins. Transgelins exhibit a broad and overlapping expression pattern, which has obscured the precise identification of their role in development. Here, we report the first systematic developmental analysis of all Drosophila transgelin proteins, namely, Mp20, CG5023, and Chd64 in the living organism. Drosophila transgelins display overall higher sequence identity with mammalian TAGLN-3 and TAGLN-2 than with TAGLN. Detailed examination in different developmental stages revealed that Mp20 and CG5023 are predominantly expressed in mesodermal tissues with the onset of myogenesis and accumulate in the cytoplasm of all somatic muscles and heart in the late embryo. Notably, at postembryonic developmental stages, Mp20 and CG5023 are detected in the gut's circumferential muscles with distinct subcellular localization: Z-lines for Mp20 and sarcomere and nucleus for CG5023. Only CG5023 is strongly detected in the adult fly in the abdominal, leg, and synchronous thoracic muscles. Chd64 protein is primarily expressed in endodermal and ectodermal tissues and has a dual subcellular localization in the cytoplasm and the nucleus. During the larval-pupae transition, Chd64 is expressed in the brain, eye, legs, halteres, and wings. In contrast, in the adult fly, Chd64 is expressed in epithelia, including the alimentary tract and genitalia. Based on the non-overlapping tissue expression, we predict that Mp20 and CG5023 mostly cooperate to modulate muscle function, whereas Chd64 has distinct roles in epithelial, neuronal, and endodermal tissues.
Collapse
Affiliation(s)
- Katerina M. Vakaloglou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Maria Mouratidou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Athina Keramidioti
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece
| | - Christos G. Zervas
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
102
|
Ohhara Y, Hoshino G, Imahori K, Matsuyuki T, Yamakawa-Kobayashi K. The Nutrient-Responsive Molecular Chaperone Hsp90 Supports Growth and Development in Drosophila. Front Physiol 2021; 12:690564. [PMID: 34239451 PMCID: PMC8258382 DOI: 10.3389/fphys.2021.690564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Animals can sense internal nutrients, such as amino acids/proteins, and are able to modify their developmental programs in accordance with their nutrient status. In the fruit fly, Drosophila melanogaster, amino acid/protein is sensed by the fat body, an insect adipose tissue, through a nutrient sensor, target of rapamycin (TOR) complex 1 (TORC1). TORC1 promotes the secretion of various peptide hormones from the fat body in an amino acid/protein-dependent manner. Fat-body-derived peptide hormones stimulate the release of insulin-like peptides, which are essential growth-promoting anabolic hormones, from neuroendocrine cells called insulin-producing cells (IPCs). Although the importance of TORC1 and the fat body-IPC axis has been elucidated, the mechanism by which TORC1 regulates the expression of insulinotropic signal peptides remains unclear. Here, we show that an evolutionarily conserved molecular chaperone, heat shock protein 90 (Hsp90), promotes the expression of insulinotropic signal peptides. Fat-body-selective Hsp90 knockdown caused the transcriptional downregulation of insulinotropic signal peptides. IPC activity and systemic growth were also impaired in fat-body-selective Hsp90 knockdown animals. Furthermore, Hsp90 expression depended on protein/amino acid availability and TORC1 signaling. These results strongly suggest that Hsp90 serves as a nutrient-responsive gene that upregulates the fat body-IPC axis and systemic growth. We propose that Hsp90 is induced in a nutrient-dependent manner to support anabolic metabolism during the juvenile growth period.
Collapse
Affiliation(s)
- Yuya Ohhara
- School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Genki Hoshino
- School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kyosuke Imahori
- School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tomoya Matsuyuki
- School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kimiko Yamakawa-Kobayashi
- School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| |
Collapse
|
103
|
Li F, Lo TY, Miles L, Wang Q, Noristani HN, Li D, Niu J, Trombley S, Goldshteyn JI, Wang C, Wang S, Qiu J, Pogoda K, Mandal K, Brewster M, Rompolas P, He Y, Janmey PA, Thomas GM, Li S, Song Y. The Atr-Chek1 pathway inhibits axon regeneration in response to Piezo-dependent mechanosensation. Nat Commun 2021; 12:3845. [PMID: 34158506 PMCID: PMC8219705 DOI: 10.1038/s41467-021-24131-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
Atr is a serine/threonine kinase, known to sense single-stranded DNA breaks and activate the DNA damage checkpoint by phosphorylating Chek1, which inhibits Cdc25, causing cell cycle arrest. This pathway has not been implicated in neuroregeneration. We show that in Drosophila sensory neurons removing Atr or Chek1, or overexpressing Cdc25 promotes regeneration, whereas Atr or Chek1 overexpression, or Cdc25 knockdown impedes regeneration. Inhibiting the Atr-associated checkpoint complex in neurons promotes regeneration and improves synapse/behavioral recovery after CNS injury. Independent of DNA damage, Atr responds to the mechanical stimulus elicited during regeneration, via the mechanosensitive ion channel Piezo and its downstream NO signaling. Sensory neuron-specific knockout of Atr in adult mice, or pharmacological inhibition of Atr-Chek1 in mammalian neurons in vitro and in flies in vivo enhances regeneration. Our findings reveal the Piezo-Atr-Chek1-Cdc25 axis as an evolutionarily conserved inhibitory mechanism for regeneration, and identify potential therapeutic targets for treating nervous system trauma.
Collapse
Affiliation(s)
- Feng Li
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tsz Y Lo
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Leann Miles
- The Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, PA, USA
| | - Qin Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Harun N Noristani
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Temple University School of Medicine, Philadelphia, PA, USA
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Dan Li
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jingwen Niu
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Temple University School of Medicine, Philadelphia, PA, USA
| | - Shannon Trombley
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jessica I Goldshteyn
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chuxi Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shuchao Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jingyun Qiu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katarzyna Pogoda
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Kalpana Mandal
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan Brewster
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ye He
- The City University of New York, Graduate Center - Advanced Science Research Center, Neuroscience Initiative, New York, NY, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Gareth M Thomas
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Temple University School of Medicine, Philadelphia, PA, USA
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Temple University School of Medicine, Philadelphia, PA, USA
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Yuanquan Song
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
104
|
Cell-type-specific, multicolor labeling of endogenous proteins with split fluorescent protein tags in Drosophila. Proc Natl Acad Sci U S A 2021; 118:2024690118. [PMID: 34074768 DOI: 10.1073/pnas.2024690118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The impact of the Drosophila experimental system on studies of modern biology cannot be understated. The ability to tag endogenously expressed proteins is essential to maximize the use of this model organism. Here, we describe a method for labeling endogenous proteins with self-complementing split fluorescent proteins (split FPs) in a cell-type-specific manner in Drosophila A short fragment of an FP coding sequence is inserted into a specific genomic locus while the remainder of the FP is expressed using an available GAL4 driver line. In consequence, complementation fluorescence allows examination of protein localization in particular cells. Besides, when inserting tandem repeats of the short FP fragment at the same genomic locus, we can substantially enhance the fluorescence signal. The enhanced signal is of great value in live-cell imaging at the subcellular level. We can also accomplish a multicolor labeling system with orthogonal split FPs. However, other orthogonal split FPs do not function for in vivo imaging besides split GFP. Through protein engineering and in vivo functional studies, we report a red split FP that we can use for duplexed visualization of endogenous proteins in intricate Drosophila tissues. Using the two orthogonal split FP systems, we have simultaneously imaged proteins that reside in distinct subsynaptic compartments. Our approach allows us to study the proximity between and localization of multiple proteins endogenously expressed in essentially any cell type in Drosophila.
Collapse
|
105
|
Yamagata N, Ezaki T, Takahashi T, Wu H, Tanimoto H. Presynaptic inhibition of dopamine neurons controls optimistic bias. eLife 2021; 10:64907. [PMID: 34061730 PMCID: PMC8169112 DOI: 10.7554/elife.64907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 05/16/2021] [Indexed: 01/04/2023] Open
Abstract
Regulation of reward signaling in the brain is critical for appropriate judgement of the environment and self. In Drosophila, the protocerebral anterior medial (PAM) cluster dopamine neurons mediate reward signals. Here, we show that localized inhibitory input to the presynaptic terminals of the PAM neurons titrates olfactory reward memory and controls memory specificity. The inhibitory regulation was mediated by metabotropic gamma-aminobutyric acid (GABA) receptors clustered in presynaptic microdomain of the PAM boutons. Cell type-specific silencing the GABA receptors enhanced memory by augmenting internal reward signals. Strikingly, the disruption of GABA signaling reduced memory specificity to the rewarded odor by changing local odor representations in the presynaptic terminals of the PAM neurons. The inhibitory microcircuit of the dopamine neurons is thus crucial for both reward values and memory specificity. Maladaptive presynaptic regulation causes optimistic cognitive bias.
Collapse
Affiliation(s)
| | - Takahiro Ezaki
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | | | - Hongyang Wu
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
106
|
Emrich-Mills TZ, Yates G, Barrett J, Girr P, Grouneva I, Lau CS, Walker CE, Kwok TK, Davey JW, Johnson MP, Mackinder LCM. A recombineering pipeline to clone large and complex genes in Chlamydomonas. THE PLANT CELL 2021; 33:1161-1181. [PMID: 33723601 PMCID: PMC8633747 DOI: 10.1093/plcell/koab024] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 01/18/2021] [Indexed: 05/10/2023]
Abstract
The ability to clone genes has greatly advanced cell and molecular biology research, enabling researchers to generate fluorescent protein fusions for localization and confirm genetic causation by mutant complementation. Most gene cloning is polymerase chain reaction (PCR)�or DNA synthesis-dependent, which can become costly and technically challenging as genes increase in size, particularly if they contain complex regions. This has been a long-standing challenge for the Chlamydomonas reinhardtii research community, as this alga has a high percentage of genes containing complex sequence structures. Here we overcame these challenges by developing a recombineering pipeline for the rapid parallel cloning of genes from a Chlamydomonas bacterial artificial chromosome collection. To generate fluorescent protein fusions for localization, we applied the pipeline at both batch and high-throughput scales to 203 genes related to the Chlamydomonas CO2 concentrating mechanism (CCM), with an overall cloning success rate of 77%. Cloning success was independent of gene size and complexity, with cloned genes as large as 23 kb. Localization of a subset of CCM targets confirmed previous mass spectrometry data, identified new pyrenoid components, and enabled complementation of mutants. We provide vectors and detailed protocols to facilitate easy adoption of this technology, which we envision will open up new possibilities in algal and plant research.
Collapse
Affiliation(s)
- Tom Z Emrich-Mills
- Department of Biology, University of York, York YO10 5DD, UK
- Department Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK
| | - Gary Yates
- Department of Biology, University of York, York YO10 5DD, UK
| | - James Barrett
- Department of Biology, University of York, York YO10 5DD, UK
| | - Philipp Girr
- Department of Biology, University of York, York YO10 5DD, UK
| | - Irina Grouneva
- Department of Biology, University of York, York YO10 5DD, UK
| | - Chun Sing Lau
- Department of Biology, University of York, York YO10 5DD, UK
| | | | - Tsz Kam Kwok
- Department of Biology, University of York, York YO10 5DD, UK
| | - John W Davey
- Department of Biology, University of York, York YO10 5DD, UK
| | - Matthew P Johnson
- Department Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, UK
| | - Luke C M Mackinder
- Department of Biology, University of York, York YO10 5DD, UK
- Author for correspondence: (L.C.M.M.)
| |
Collapse
|
107
|
Mukherjee S, Paricio N, Sokol NS. A stress-responsive miRNA regulates BMP signaling to maintain tissue homeostasis. Proc Natl Acad Sci U S A 2021; 118:e2022583118. [PMID: 34016750 PMCID: PMC8166057 DOI: 10.1073/pnas.2022583118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adult organisms must sense and adapt to environmental fluctuations. In high-turnover tissues such as the intestine, these adaptive responses require rapid changes in gene expression that, in turn, likely involve posttranscriptional gene control. However, intestinal-tissue-specific microRNA (miRNA)-mediated regulatory pathways remain unexplored. Here, we report the role of an intestinal-specific miRNA, miR-958, that non-cell autonomously regulates stem cell numbers during tissue homeostasis and regeneration in the Drosophila adult midgut. We identify its downstream target cabut, the Drosophila ortholog of mammalian KLF10/11 transcription factors, which mediates this miR-958 function by promoting paracrine enterocyte-to-stem-cell bone morphogenetic protein (BMP) signaling. We also show that mature miR-958 levels transiently decrease in response to stress and that this decrease is required for proper stem cell expansion during tissue regeneration. In summary, we have identified a posttranscriptional mechanism that modulates BMP signaling activity within Drosophila adult intestinal tissue during both normal homeostasis and tissue regeneration to regulate intestinal stem cell numbers.
Collapse
Affiliation(s)
| | - Nuria Paricio
- Departamento de Genética, Facultad de Ciencies Biológicas and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina, University of Valencia, 46100 Burjasot, Spain
| | - Nicholas S Sokol
- Department of Biology, Indiana University, Bloomington, IN 47405;
| |
Collapse
|
108
|
Avellaneda J, Rodier C, Daian F, Brouilly N, Rival T, Luis NM, Schnorrer F. Myofibril and mitochondria morphogenesis are coordinated by a mechanical feedback mechanism in muscle. Nat Commun 2021; 12:2091. [PMID: 33828099 PMCID: PMC8027795 DOI: 10.1038/s41467-021-22058-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/23/2021] [Indexed: 02/01/2023] Open
Abstract
Complex animals build specialised muscles to match specific biomechanical and energetic needs. Hence, composition and architecture of sarcomeres and mitochondria are muscle type specific. However, mechanisms coordinating mitochondria with sarcomere morphogenesis are elusive. Here we use Drosophila muscles to demonstrate that myofibril and mitochondria morphogenesis are intimately linked. In flight muscles, the muscle selector spalt instructs mitochondria to intercalate between myofibrils, which in turn mechanically constrain mitochondria into elongated shapes. Conversely in cross-striated leg muscles, mitochondria networks surround myofibril bundles, contacting myofibrils only with thin extensions. To investigate the mechanism causing these differences, we manipulated mitochondrial dynamics and found that increased mitochondrial fusion during myofibril assembly prevents mitochondrial intercalation in flight muscles. Strikingly, this causes the expression of cross-striated muscle specific sarcomeric proteins. Consequently, flight muscle myofibrils convert towards a partially cross-striated architecture. Together, these data suggest a biomechanical feedback mechanism downstream of spalt synchronizing mitochondria with myofibril morphogenesis.
Collapse
Affiliation(s)
- Jerome Avellaneda
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Clement Rodier
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Fabrice Daian
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Nicolas Brouilly
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Thomas Rival
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Nuno Miguel Luis
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, Marseille, France.
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Center for Living Systems, Marseille, France.
| |
Collapse
|
109
|
Böhme MA, McCarthy AW, Blaum N, Berezeckaja M, Ponimaskine K, Schwefel D, Walter AM. Glial Synaptobrevin mediates peripheral nerve insulation, neural metabolic supply, and is required for motor function. Glia 2021; 69:1897-1915. [PMID: 33811396 DOI: 10.1002/glia.24000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 01/10/2023]
Abstract
Peripheral nerves contain sensory and motor neuron axons coated by glial cells whose interplay ensures function, but molecular details are lacking. SNARE-proteins mediate the exchange and secretion of cargo by fusing vesicles with target organelles, but how glial SNAREs contribute to peripheral nerve function is largely unknown. We, here, identify non-neuronal Synaptobrevin (Syb) as the essential vesicular SNARE in Drosophila peripheral glia to insulate and metabolically supply neurons. We show that tetanus neurotoxin light chain (TeNT-LC), which potently inhibits SNARE-mediated exocytosis from neurons, also impairs peripheral nerve function when selectively expressed in glia, causing nerve disintegration, defective axonal transport, tetanic muscle hyperactivity, impaired locomotion, and lethality. While TeNT-LC disrupts neural function by cleaving neuronal Synaptobrevin (nSyb), it targets non-neuronal Synaptobrevin (Syb) in glia, which it cleaves at low rates: Glial knockdown of Syb (but not nSyb) phenocopied glial TeNT-LC expression whose effects were reverted by a TeNT-LC-insensitive Syb mutant. We link Syb-necessity to two distinct glial subtypes: Impairing Syb function in subperineurial glia disrupted nerve morphology, axonal transport, and locomotion, likely, because nerve-isolating septate junctions (SJs) could not form as essential SJ components (like the cell adhesion protein Neurexin-IV) were mistargeted. Interference with Syb in axon-encircling wrapping glia left nerve morphology and locomotion intact but impaired axonal transport, likely because neural metabolic supply was disrupted due to the mistargeting of metabolite shuffling monocarboxylate transporters. Our study identifies crucial roles of Syb in various glial subtypes to ensure glial-glial and glial-neural interplay needed for proper nerve function, animal motility, and survival.
Collapse
Affiliation(s)
- Mathias A Böhme
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), FMP im CharitéCrossOver, Berlin, Germany.,Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anthony W McCarthy
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), FMP im CharitéCrossOver, Berlin, Germany
| | - Natalie Blaum
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), FMP im CharitéCrossOver, Berlin, Germany
| | - Monika Berezeckaja
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), FMP im CharitéCrossOver, Berlin, Germany
| | - Kristina Ponimaskine
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), FMP im CharitéCrossOver, Berlin, Germany
| | - David Schwefel
- Institute of Medical Physics and Biophysics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alexander M Walter
- Molecular and Theoretical Neuroscience, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), FMP im CharitéCrossOver, Berlin, Germany.,Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
110
|
Casas-Tintó S, Ferrús A. The haplolethality paradox of the wupA gene in Drosophila. PLoS Genet 2021; 17:e1009108. [PMID: 33739971 PMCID: PMC8011728 DOI: 10.1371/journal.pgen.1009108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/31/2021] [Accepted: 02/15/2021] [Indexed: 11/18/2022] Open
Abstract
Haplolethals (HL) are regions of diploid genomes that in one dose are fatal for the organism. Their biological meaning is obscure because heterozygous loss-of-function mutations result in dominant lethality (DL) and, consequently, should be under strong negative selection. We report an in depth study of the HL associated to the gene wings up A (wupA). It encodes 13 transcripts (A-M) that yield 11 protein isoforms (A-K) of Troponin I (TnI). They are functionally diverse in their control of muscle contraction, cell polarity and cell proliferation. Isoform K transfers to the nucleus where it increases transcription of the cell proliferation related genes CDK2, CDK4, Rap and Rab5. The nuclear translocation of isoform K is prevented by the co-expression of A or B isoforms, which illustrates isoform interactions. The corresponding DL mutations are, either DNA rearrangements clustered towards the gene 3’ end, thus affecting the genomic organization of all transcripts, or CRISPR-induced mutations in one of the two ATG sites which eliminate a subset of wupA products. The joint elimination of isoforms C, F, G and H, however, do not cause DL phenotypes. Genetically driven expression of single isoforms rescue neither DL nor any of the mutants known in the gene, suggesting that normal function requires properly regulated expression of specific combinations, rather than single, TnI isoforms. We conclude that the wupA associated HL results from the combined haploinsufficiency of a large set of TnI isoforms. The qualitative and quantitative normal expression of which, requires the chromosomal integrity of the wupA genomic region. Since all fly TnI isoforms are encoded in the same gene, its HL condition becomes unavoidable. These wupA features are comparable to those of dpp, the only other HL studied to some extent, and reveal a scenario of strict dosage dependence with implications for gene expression regulation and splitting. Most species contain two copies of their genetic endowment, each received from each progenitor. If one of the duplicated genes is non-functional, the remaining copy may supply enough product as to cover the requirements for normal function or, alternatively, may reflect the insufficiency through a visible phenotype. In rare occasions, however, having one copy is so deleterious that causes lethality. These so called “haplolethal regions”, exist across species and represent an evolutionary paradox since they should have been subject to intense negative selection. The inherent difficulties to study haplolethals have precluded their study so far. Here, we analyzed the case of one of the five haplolethal regions of Drosophila, the one associated to the Troponin I encoding gene wupA, by measuring the transcriptional effects of mutations and chromosomal rearrangements affecting this gene. The data show that this haplolethality results from the combined insufficiency of a large number of Troponin I isoforms, which are functionally specialized, show interference and require the integrity of the native chromatin structure for their quantitatively regulated expression. These features unveil novel aspects of gene expression and, possibly, on evolutionary gene splitting. Also, haplolethals underscore the biological significance of protein dosage, in particular for functionally related products.
Collapse
Affiliation(s)
- Sergio Casas-Tintó
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Alberto Ferrús
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
- * E-mail:
| |
Collapse
|
111
|
Marcotti S, Sánchez-Sánchez BJ, Serna-Morales E, Dragu A, Díaz-de-la-Loza MDC, Matsubayashi Y, Stramer BM. Protocol for intervention-free quantification of protein turnover rate by steady-state modeling. STAR Protoc 2021; 2:100377. [PMID: 33786460 PMCID: PMC7988239 DOI: 10.1016/j.xpro.2021.100377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Protein turnover rate is difficult to obtain experimentally. This protocol shows how to mathematically model turnover rates in an intervention-free manner given the ability to quantify mRNA and protein expression from initiation to homeostasis. This approach can be used to calculate production and degradation rates and to infer protein half-life. This model was successfully employed to quantify turnover during Drosophila embryogenesis, and we hypothesize that it will be applicable to diverse in vivo or in vitro systems. For complete details on the use and execution of this protocol, please refer to Matsubayashi et al. (2020).
Collapse
Affiliation(s)
- Stefania Marcotti
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
| | | | - Eduardo Serna-Morales
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
| | - Anca Dragu
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
| | | | - Yutaka Matsubayashi
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
| | - Brian Marc Stramer
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
| |
Collapse
|
112
|
Vigano MA, Ell CM, Kustermann MMM, Aguilar G, Matsuda S, Zhao N, Stasevich TJ, Affolter M, Pyrowolakis G. Protein manipulation using single copies of short peptide tags in cultured cells and in Drosophila melanogaster. Development 2021; 148:dev191700. [PMID: 33593816 PMCID: PMC7990863 DOI: 10.1242/dev.191700] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 02/09/2021] [Indexed: 01/01/2023]
Abstract
Cellular development and function rely on highly dynamic molecular interactions among proteins distributed in all cell compartments. Analysis of these interactions has been one of the main topics in cellular and developmental research, and has been mostly achieved by the manipulation of proteins of interest (POIs) at the genetic level. Although genetic strategies have significantly contributed to our current understanding, targeting specific interactions of POIs in a time- and space-controlled manner or analysing the role of POIs in dynamic cellular processes, such as cell migration or cell division, would benefit from more-direct approaches. The recent development of specific protein binders, which can be expressed and function intracellularly, along with advancement in synthetic biology, have contributed to the creation of a new toolbox for direct protein manipulations. Here, we have selected a number of short-tag epitopes for which protein binders from different scaffolds have been generated and showed that single copies of these tags allowed efficient POI binding and manipulation in living cells. Using Drosophila, we also find that single short tags can be used for POI manipulation in vivo.
Collapse
Affiliation(s)
- M Alessandra Vigano
- Growth and Development, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland
| | - Clara-Maria Ell
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
- Institute for Biology I, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104 Freiburg, Germany
| | - Manuela M M Kustermann
- Institute for Biology I, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104 Freiburg, Germany
| | - Gustavo Aguilar
- Growth and Development, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland
| | - Shinya Matsuda
- Growth and Development, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland
| | - Ning Zhao
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Markus Affolter
- Growth and Development, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland
| | - George Pyrowolakis
- Institute for Biology I, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
- Center for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104 Freiburg, Germany
| |
Collapse
|
113
|
Mohr SE, Tattikota SG, Xu J, Zirin J, Hu Y, Perrimon N. Methods and tools for spatial mapping of single-cell RNAseq clusters in Drosophila. Genetics 2021; 217:6156631. [PMID: 33713129 DOI: 10.1093/genetics/iyab019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/02/2021] [Indexed: 01/26/2023] Open
Abstract
Single-cell RNA sequencing (scRNAseq) experiments provide a powerful means to identify clusters of cells that share common gene expression signatures. A major challenge in scRNAseq studies is to map the clusters to specific anatomical regions along the body and within tissues. Existing data, such as information obtained from large-scale in situ RNA hybridization studies, cell type specific transcriptomics, gene expression reporters, antibody stainings, and fluorescent tagged proteins, can help to map clusters to anatomy. However, in many cases, additional validation is needed to precisely map the spatial location of cells in clusters. Several approaches are available for spatial resolution in Drosophila, including mining of existing datasets, and use of existing or new tools for direct or indirect detection of RNA, or direct detection of proteins. Here, we review available resources and emerging technologies that will facilitate spatial mapping of scRNAseq clusters at high resolution in Drosophila. Importantly, we discuss the need, available approaches, and reagents for multiplexing gene expression detection in situ, as in most cases scRNAseq clusters are defined by the unique coexpression of sets of genes.
Collapse
Affiliation(s)
- Stephanie E Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sudhir Gopal Tattikota
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Xu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan Zirin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.,Howard Hughes Medical Institute, Boston, MA 02115, USA
| |
Collapse
|
114
|
Zhang Z, Denans N, Liu Y, Zhulyn O, Rosenblatt HD, Wernig M, Barna M. Optogenetic manipulation of cellular communication using engineered myosin motors. Nat Cell Biol 2021; 23:198-208. [PMID: 33526902 PMCID: PMC7880895 DOI: 10.1038/s41556-020-00625-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022]
Abstract
Cells achieve highly efficient and accurate communication through cellular projections such as neurites and filopodia, yet there is a lack of genetically encoded tools that can selectively manipulate their composition and dynamics. Here, we present a versatile optogenetic toolbox of artificial multi-headed myosin motors that can move bidirectionally within long cellular extensions and allow for the selective transport of GFP-tagged cargo with light. Utilizing these engineered motors, we could transport bulky transmembrane receptors and organelles as well as actin remodellers to control the dynamics of both filopodia and neurites. Using an optimized in vivo imaging scheme, we further demonstrate that, upon limb amputation in axolotls, a complex array of filopodial extensions is formed. We selectively modulated these filopodial extensions and showed that they re-establish a Sonic Hedgehog signalling gradient during regeneration. Considering the ubiquitous existence of actin-based extensions, this toolbox shows the potential to manipulate cellular communication with unprecedented accuracy.
Collapse
Affiliation(s)
- Zijian Zhang
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Nicolas Denans
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Yingfei Liu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Olena Zhulyn
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Hannah D Rosenblatt
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Maria Barna
- Department of Developmental Biology, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
115
|
Aughey GN, Delandre C, McMullen JPD, Southall TD, Marshall OJ. FlyORF-TaDa allows rapid generation of new lines for in vivo cell-type-specific profiling of protein-DNA interactions in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2021; 11:6044134. [PMID: 33561239 PMCID: PMC8022459 DOI: 10.1093/g3journal/jkaa005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/07/2020] [Indexed: 12/01/2022]
Abstract
Targeted DamID (TaDa) is an increasingly popular method of generating cell-type-specific DNA-binding profiles in vivo. Although sensitive and versatile, TaDa requires the generation of new transgenic fly lines for every protein that is profiled, which is both time-consuming and costly. Here, we describe the FlyORF-TaDa system for converting an existing FlyORF library of inducible open reading frames (ORFs) to TaDa lines via a genetic cross, with recombinant progeny easily identifiable by eye color. Profiling the binding of the H3K36me3-associated chromatin protein MRG15 in larval neural stem cells using both FlyORF-TaDa and conventional TaDa demonstrates that new lines generated using this system provide accurate and highly reproducible DamID-binding profiles. Our data further show that MRG15 binds to a subset of active chromatin domains in vivo. Courtesy of the large coverage of the FlyORF library, the FlyORF-TaDa system enables the easy creation of TaDa lines for 74% of all transcription factors and chromatin-modifying proteins within the Drosophila genome.
Collapse
Affiliation(s)
- Gabriel N Aughey
- Imperial College London, Sir Ernst Chain Building, South Kensington Campus, London SW7 2AZ, UK
| | - Caroline Delandre
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool St, Hobart 7000, Australia
| | - John P D McMullen
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool St, Hobart 7000, Australia
| | - Tony D Southall
- Imperial College London, Sir Ernst Chain Building, South Kensington Campus, London SW7 2AZ, UK
| | - Owen J Marshall
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool St, Hobart 7000, Australia
| |
Collapse
|
116
|
Gallicchio L, Griffiths-Jones S, Ronshaugen M. Single-cell visualization of mir-9a and Senseless co-expression during Drosophila melanogaster embryonic and larval peripheral nervous system development. G3-GENES GENOMES GENETICS 2021; 11:6044132. [PMID: 33561238 PMCID: PMC7849905 DOI: 10.1093/g3journal/jkaa010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/16/2020] [Indexed: 11/17/2022]
Abstract
The Drosophila melanogaster peripheral nervous system (PNS) comprises the sensory organs that allow the fly to detect environmental factors such as temperature and pressure. PNS development is a highly specified process where each sensilla originates from a single sensory organ precursor (SOP) cell. One of the major genetic orchestrators of PNS development is Senseless, which encodes a zinc finger transcription factor (Sens). Sens is both necessary and sufficient for SOP differentiation. Senseless expression and SOP number are regulated by the microRNA miR-9a. However, the reciprocal dynamics of Senseless and miR-9a are still obscure. By coupling single-molecule FISH with immunofluorescence, we are able to visualize transcription of the mir-9a locus and expression of Sens simultaneously. During embryogenesis, we show that the expression of mir-9a in SOP cells is rapidly lost as Senseless expression increases. However, this mutually exclusive expression pattern is not observed in the third instar imaginal wing disc, where some Senseless-expressing cells show active sites of mir-9a transcription. These data challenge and extend previous models of Senseless regulation and show complex co-expression dynamics between mir-9a and Senseless. The differences in this dynamic relationship between embryonic and larval PNS development suggest a possible switch in miR-9a function. Our work brings single-cell resolution to the understanding of dynamic regulation of PNS development by Senseless and miR-9a.
Collapse
Affiliation(s)
- Lorenzo Gallicchio
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Sam Griffiths-Jones
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - Matthew Ronshaugen
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| |
Collapse
|
117
|
Kaya-Çopur A, Marchiano F, Hein MY, Alpern D, Russeil J, Luis NM, Mann M, Deplancke B, Habermann BH, Schnorrer F. The Hippo pathway controls myofibril assembly and muscle fiber growth by regulating sarcomeric gene expression. eLife 2021; 10:e63726. [PMID: 33404503 PMCID: PMC7815313 DOI: 10.7554/elife.63726] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/05/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscles are composed of gigantic cells called muscle fibers, packed with force-producing myofibrils. During development, the size of individual muscle fibers must dramatically enlarge to match with skeletal growth. How muscle growth is coordinated with growth of the contractile apparatus is not understood. Here, we use the large Drosophila flight muscles to mechanistically decipher how muscle fiber growth is controlled. We find that regulated activity of core members of the Hippo pathway is required to support flight muscle growth. Interestingly, we identify Dlg5 and Slmap as regulators of the STRIPAK phosphatase, which negatively regulates Hippo to enable post-mitotic muscle growth. Mechanistically, we show that the Hippo pathway controls timing and levels of sarcomeric gene expression during development and thus regulates the key components that physically mediate muscle growth. Since Dlg5, STRIPAK and the Hippo pathway are conserved a similar mechanism may contribute to muscle or cardiomyocyte growth in humans.
Collapse
Affiliation(s)
- Aynur Kaya-Çopur
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Fabio Marchiano
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
| | - Marco Y Hein
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Daniel Alpern
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Julie Russeil
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Nuno Miguel Luis
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
| | - Matthias Mann
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Bart Deplancke
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Bianca H Habermann
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Center for Living SystemsMarseilleFrance
- Max Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
118
|
Parkhitko AA, Ramesh D, Wang L, Leshchiner D, Filine E, Binari R, Olsen AL, Asara JM, Cracan V, Rabinowitz JD, Brockmann A, Perrimon N. Downregulation of the tyrosine degradation pathway extends Drosophila lifespan. eLife 2020; 9:58053. [PMID: 33319750 PMCID: PMC7744100 DOI: 10.7554/elife.58053] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 11/28/2020] [Indexed: 12/31/2022] Open
Abstract
Aging is characterized by extensive metabolic reprogramming. To identify metabolic pathways associated with aging, we analyzed age-dependent changes in the metabolomes of long-lived Drosophila melanogaster. Among the metabolites that changed, levels of tyrosine were increased with age in long-lived flies. We demonstrate that the levels of enzymes in the tyrosine degradation pathway increase with age in wild-type flies. Whole-body and neuronal-specific downregulation of enzymes in the tyrosine degradation pathway significantly extends Drosophila lifespan, causes alterations of metabolites associated with increased lifespan, and upregulates the levels of tyrosine-derived neuromediators. Moreover, feeding wild-type flies with tyrosine increased their lifespan. Mechanistically, we show that suppression of ETC complex I drives the upregulation of enzymes in the tyrosine degradation pathway, an effect that can be rescued by tigecycline, an FDA-approved drug that specifically suppresses mitochondrial translation. In addition, tyrosine supplementation partially rescued lifespan of flies with ETC complex I suppression. Altogether, our study highlights the tyrosine degradation pathway as a regulator of longevity.
Collapse
Affiliation(s)
- Andrey A Parkhitko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States.,Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, United States
| | - Divya Ramesh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India.,Department of Biology, University of Konstanz, Konstanz, Germany
| | - Lin Wang
- Department of Chemistry, Princeton University, Princeton, United States.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, United States
| | - Dmitry Leshchiner
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Elizabeth Filine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Richard Binari
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| | - Abby L Olsen
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, United States
| | - Valentin Cracan
- Scintillon Institute, San Diego, United States.,Department of Chemistry, The Scripps Research Institute, La Jolla, United States
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, United States.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, United States
| | - Axel Brockmann
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| |
Collapse
|
119
|
Rondelet A, Pozniakovsky A, Namboodiri D, Cardoso da Silva R, Singh D, Leuschner M, Poser I, Ssykor A, Berlitz J, Schmidt N, Röhder L, Vader G, Hyman AA, Bird AW. ESI mutagenesis: a one-step method for introducing mutations into bacterial artificial chromosomes. Life Sci Alliance 2020; 4:4/2/e202000836. [PMID: 33293335 PMCID: PMC7756954 DOI: 10.26508/lsa.202000836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 01/23/2023] Open
Abstract
A simple and efficient recombineering-based method for introducing point mutations into bacterial artificial chromosomes using an artificial intron cassette. Bacterial artificial chromosome (BAC)–based transgenes have emerged as a powerful tool for controlled and conditional interrogation of protein function in higher eukaryotes. Although homologous recombination-based recombineering methods have streamlined the efficient integration of protein tags onto BAC transgenes, generating precise point mutations has remained less efficient and time-consuming. Here, we present a simplified method for inserting point mutations into BAC transgenes requiring a single recombineering step followed by antibiotic selection. This technique, which we call exogenous/synthetic intronization (ESI) mutagenesis, relies on co-integration of a mutation of interest along with a selectable marker gene, the latter of which is harboured in an artificial intron adjacent to the mutation site. Cell lines generated from ESI-mutated BACs express the transgenes equivalently to the endogenous gene, and all cells efficiently splice out the synthetic intron. Thus, ESI mutagenesis provides a robust and effective single-step method with high precision and high efficiency for mutating BAC transgenes.
Collapse
Affiliation(s)
- Arnaud Rondelet
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andrei Pozniakovsky
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | - Divya Singh
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Marit Leuschner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ina Poser
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrea Ssykor
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Julian Berlitz
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Nadine Schmidt
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Lea Röhder
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Gerben Vader
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Anthony A Hyman
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | |
Collapse
|
120
|
Cai H, Holleufer A, Simonsen B, Schneider J, Lemoine A, Gad HH, Huang J, Huang J, Chen D, Peng T, Marques JT, Hartmann R, Martins NE, Imler JL. 2'3'-cGAMP triggers a STING- and NF-κB-dependent broad antiviral response in Drosophila. Sci Signal 2020; 13:13/660/eabc4537. [PMID: 33262294 DOI: 10.1126/scisignal.abc4537] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
We previously reported that an ortholog of STING regulates infection by picorna-like viruses in Drosophila In mammals, STING is activated by the cyclic dinucleotide 2'3'-cGAMP produced by cGAS, which acts as a receptor for cytosolic DNA. Here, we showed that injection of flies with 2'3'-cGAMP induced the expression of dSTING-regulated genes. Coinjection of 2'3'-cGAMP with a panel of RNA or DNA viruses resulted in substantially reduced viral replication. This 2'3'-cGAMP-mediated protection was still observed in flies with mutations in Atg7 and AGO2, genes that encode key components of the autophagy and small interfering RNA pathways, respectively. By contrast, this protection was abrogated in flies with mutations in the gene encoding the NF-κB transcription factor Relish. Transcriptomic analysis of 2'3'-cGAMP-injected flies revealed a complex response pattern in which genes were rapidly induced, induced after a delay, or induced in a sustained manner. Our results reveal that dSTING regulates an NF-κB-dependent antiviral program that predates the emergence of interferons in vertebrates.
Collapse
Affiliation(s)
- Hua Cai
- Sino-French Hoffmann Institute, State Key Laboratory of Respiratory Disease, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.,Université de Strasbourg, CNRS UPR 9022, 67084 Strasbourg, France
| | - Andreas Holleufer
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Bine Simonsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Aurélie Lemoine
- Université de Strasbourg, CNRS UPR 9022, 67084 Strasbourg, France
| | - Hans Henrik Gad
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Jingxian Huang
- Sino-French Hoffmann Institute, State Key Laboratory of Respiratory Disease, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Jieqing Huang
- Sino-French Hoffmann Institute, State Key Laboratory of Respiratory Disease, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Di Chen
- Sino-French Hoffmann Institute, State Key Laboratory of Respiratory Disease, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Tao Peng
- Sino-French Hoffmann Institute, State Key Laboratory of Respiratory Disease, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - João T Marques
- Université de Strasbourg, CNRS UPR 9022, INSERM U1257, 67084 Strasbourg, France.,Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais CEP 31270901, Brazil
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark.
| | - Nelson E Martins
- Université de Strasbourg, CNRS UPR 9022, 67084 Strasbourg, France.
| | - Jean-Luc Imler
- Sino-French Hoffmann Institute, State Key Laboratory of Respiratory Disease, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.,Université de Strasbourg, CNRS UPR 9022, 67084 Strasbourg, France
| |
Collapse
|
121
|
Spannl S, Buhl T, Nellas I, Zeidan SA, Iyer KV, Khaliullina H, Schultz C, Nadler A, Dye NA, Eaton S. Glycolysis regulates Hedgehog signalling via the plasma membrane potential. EMBO J 2020; 39:e101767. [PMID: 33021744 PMCID: PMC7604625 DOI: 10.15252/embj.2019101767] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/19/2020] [Accepted: 08/25/2020] [Indexed: 01/04/2023] Open
Abstract
Changes in cell metabolism and plasma membrane potential have been linked to shifts between tissue growth and differentiation, and to developmental patterning. How such changes mediate these effects is poorly understood. Here, we use the developing wing of Drosophila to investigate the interplay between cell metabolism and a key developmental regulator-the Hedgehog (Hh) signalling pathway. We show that reducing glycolysis both lowers steady-state levels of ATP and stabilizes Smoothened (Smo), the 7-pass transmembrane protein that transduces the Hh signal. As a result, the transcription factor Cubitus interruptus accumulates in its full-length, transcription activating form. We show that glycolysis is required to maintain the plasma membrane potential and that plasma membrane depolarization blocks cellular uptake of N-acylethanolamides-lipoprotein-borne Hh pathway inhibitors required for Smo destabilization. Similarly, pharmacological inhibition of glycolysis in mammalian cells induces ciliary translocation of Smo-a key step in pathway activation-in the absence of Hh. Thus, changes in cell metabolism alter Hh signalling through their effects on plasma membrane potential.
Collapse
Affiliation(s)
- Stephanie Spannl
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Present address:
Department of BiochemistryFaculty of MedicineUniversity of TorontoTorontoONCanada
| | - Tomasz Buhl
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
| | - Ioannis Nellas
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
| | - Salma A Zeidan
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
| | - K Venkatesan Iyer
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Max Planck Institute for the Physics of Complex SystemsDresdenGermany
| | - Helena Khaliullina
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Present address:
Department of PhysiologyDevelopment and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Carsten Schultz
- Cell Biology and Biophysics UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Department of Chemical Physiology and BiochemistryOregon Health and Science UniversityPortlandORUSA
| | - André Nadler
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Natalie A Dye
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Suzanne Eaton
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Biotechnologisches ZentrumTechnische Universität DresdenDresdenGermany
- Center for Systems Biology DresdenDresdenGermany
| |
Collapse
|
122
|
Miscopein Saler L, Hauser V, Bartoletti M, Mallart C, Malartre M, Lebrun L, Pret AM, Théodore L, Chalvet F, Netter S. The Bric-à-Brac BTB/POZ transcription factors are necessary in niche cells for germline stem cells establishment and homeostasis through control of BMP/DPP signaling in the Drosophila melanogaster ovary. PLoS Genet 2020; 16:e1009128. [PMID: 33151937 PMCID: PMC7643948 DOI: 10.1371/journal.pgen.1009128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/22/2020] [Indexed: 11/26/2022] Open
Abstract
Many studies have focused on the mechanisms of stem cell maintenance via their interaction with a particular niche or microenvironment in adult tissues, but how formation of a functional niche is initiated, including how stem cells within a niche are established, is less well understood. Adult Drosophila melanogaster ovary Germline Stem Cell (GSC) niches are comprised of somatic cells forming a stack called a Terminal Filament (TF) and associated Cap and Escort Cells (CCs and ECs, respectively), which are in direct contact with GSCs. In the adult ovary, the transcription factor Engrailed is specifically expressed in niche cells where it directly controls expression of the decapentaplegic (dpp) gene encoding a member of the Bone Morphogenetic Protein (BMP) family of secreted signaling molecules, which are key factors for GSC maintenance. In larval ovaries, in response to BMP signaling from newly formed niches, adjacent primordial germ cells become GSCs. The bric-à-brac paralogs (bab1 and bab2) encode BTB/POZ domain-containing transcription factors that are expressed in developing niches of larval ovaries. We show here that their functions are necessary specifically within precursor cells for TF formation during these stages. We also identify a new function for Bab1 and Bab2 within developing niches for GSC establishment in the larval ovary and for robust GSC maintenance in the adult. Moreover, we show that the presence of Bab proteins in niche cells is necessary for activation of transgenes reporting dpp expression as of larval stages in otherwise correctly specified Cap Cells, independently of Engrailed and its paralog Invected (En/Inv). Moreover, strong reduction of engrailed/invected expression during larval stages does not impair TF formation and only partially reduces GSC numbers. In the adult ovary, Bab proteins are also required for dpp reporter expression in CCs. Finally, when bab2 was overexpressed at this stage in somatic cells outside of the niche, there were no detectable levels of ectopic En/Inv, but ectopic expression of a dpp transgene was found in these cells and BMP signaling activation was induced in adjacent germ cells, which produced GSC-like tumors. Together, these results indicate that Bab transcription factors are positive regulators of BMP signaling in niche cells for establishment and homeostasis of GSCs in the Drosophila ovary.
Collapse
Affiliation(s)
- Laurine Miscopein Saler
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Virginie Hauser
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Mathieu Bartoletti
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Charlotte Mallart
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Marianne Malartre
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Laura Lebrun
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anne-Marie Pret
- Université Paris-Saclay, UVSQ, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Laurent Théodore
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Fabienne Chalvet
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Sophie Netter
- Université Paris-Saclay, UVSQ, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| |
Collapse
|
123
|
Hadjieconomou D, King G, Gaspar P, Mineo A, Blackie L, Ameku T, Studd C, de Mendoza A, Diao F, White BH, Brown AEX, Plaçais PY, Préat T, Miguel-Aliaga I. Enteric neurons increase maternal food intake during reproduction. Nature 2020; 587:455-459. [PMID: 33116314 PMCID: PMC7610780 DOI: 10.1038/s41586-020-2866-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/04/2020] [Indexed: 01/01/2023]
Abstract
Reproduction induces increased food intake across females of many animal species1-4, providing a physiologically relevant paradigm for the exploration of appetite regulation. Here, by examining the diversity of enteric neurons in Drosophila melanogaster, we identify a key role for gut-innervating neurons with sex- and reproductive state-specific activity in sustaining the increased food intake of mothers during reproduction. Steroid and enteroendocrine hormones functionally remodel these neurons, which leads to the release of their neuropeptide onto the muscles of the crop-a stomach-like organ-after mating. Neuropeptide release changes the dynamics of crop enlargement, resulting in increased food intake, and preventing the post-mating remodelling of enteric neurons reduces both reproductive hyperphagia and reproductive fitness. The plasticity of enteric neurons is therefore key to reproductive success. Our findings provide a mechanism to attain the positive energy balance that sustains gestation, dysregulation of which could contribute to infertility or weight gain.
Collapse
Affiliation(s)
- Dafni Hadjieconomou
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - George King
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Pedro Gaspar
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Alessandro Mineo
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Laura Blackie
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Tomotsune Ameku
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Chris Studd
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Alex de Mendoza
- Australian Research Council Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Fengqiu Diao
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin H White
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - André E X Brown
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Pierre-Yves Plaçais
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Thomas Préat
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, London, UK.
- Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
124
|
Fendl S, Vieira RM, Borst A. Conditional protein tagging methods reveal highly specific subcellular distribution of ion channels in motion-sensing neurons. eLife 2020; 9:62953. [PMID: 33079061 PMCID: PMC7655108 DOI: 10.7554/elife.62953] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/14/2020] [Indexed: 11/25/2022] Open
Abstract
Neurotransmitter receptors and ion channels shape the biophysical properties of neurons, from the sign of the response mediated by neurotransmitter receptors to the dynamics shaped by voltage-gated ion channels. Therefore, knowing the localizations and types of receptors and channels present in neurons is fundamental to our understanding of neural computation. Here, we developed two approaches to visualize the subcellular localization of specific proteins in Drosophila: The flippase-dependent expression of GFP-tagged receptor subunits in single neurons and ‘FlpTag’, a versatile new tool for the conditional labelling of endogenous proteins. Using these methods, we investigated the subcellular distribution of the receptors GluClα, Rdl, and Dα7 and the ion channels para and Ih in motion-sensing T4/T5 neurons of the Drosophila visual system. We discovered a strictly segregated subcellular distribution of these proteins and a sequential spatial arrangement of glutamate, acetylcholine, and GABA receptors along the dendrite that matched the previously reported EM-reconstructed synapse distributions.
Collapse
Affiliation(s)
- Sandra Fendl
- Max Planck Institute of Neurobiology, Martinsried, Germany.,Graduate School of Systemic Neurosciences, LMU Munich, Martinsried, Germany
| | | | - Alexander Borst
- Max Planck Institute of Neurobiology, Martinsried, Germany.,Graduate School of Systemic Neurosciences, LMU Munich, Martinsried, Germany
| |
Collapse
|
125
|
Horne-Badovinac S. The Drosophila micropyle as a system to study how epithelia build complex extracellular structures. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190561. [PMID: 32829690 PMCID: PMC7482212 DOI: 10.1098/rstb.2019.0561] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2020] [Indexed: 02/02/2023] Open
Abstract
Dynamic rearrangements of epithelial cells play central roles in shaping tissues and organs during development. There are also scenarios, however, in which epithelial cell movements synergize with the secretion of extracellular matrix to build rigid, acellular structures that persist long after the cells are gone. The formation of the Drosophila micropyle provides an elegant example of this epithelial craftsmanship. The micropyle is a cone-shaped projection of the eggshell through which the sperm will enter to fertilize the oocyte. Though simple on the surface, both the inner structure and construction of the micropyle are remarkably complex. In this review, I first provide an overview of egg development, focusing on the key events required to understand micropyle formation. I then describe the structure of the micropyle, the cellular contributions to its morphogenesis and some interesting open questions about this process. There is a brief discussion of micropyle formation in other insects and fish to highlight the potential for comparative studies. Finally, I discuss how new studies of micropyle formation could reveal general mechanisms that epithelia use to build complex extracellular structures. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Sally Horne-Badovinac
- Department of Molecular Genetics and Cell Biology, The University of Chicago, 920 East 58th Street, Chicago, IL 60637, USA
| |
Collapse
|
126
|
Arefin B, Parvin F, Bahrampour S, Stadler CB, Thor S. Drosophila Neuroblast Selection Is Gated by Notch, Snail, SoxB, and EMT Gene Interplay. Cell Rep 2020; 29:3636-3651.e3. [PMID: 31825841 DOI: 10.1016/j.celrep.2019.11.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/20/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
In the developing Drosophila central nervous system (CNS), neural progenitor (neuroblast [NB]) selection is gated by lateral inhibition, controlled by Notch signaling and proneural genes. However, proneural mutants still generate many NBs, indicating the existence of additional proneural genes. Moreover, recent studies reveal involvement of key epithelial-mesenchymal transition (EMT) genes in NB selection, but the regulatory interplay between Notch signaling and the EMT machinery is unclear. We find that SoxNeuro (SoxB family) and worniu (Snail family) are integrated with the Notch pathway, and constitute the missing proneural genes. Notch signaling, the proneural, SoxNeuro, and worniu genes regulate key EMT genes to orchestrate the NB selection process. Hence, we uncover an expanded lateral inhibition network for NB selection and demonstrate its link to key players in the EMT machinery. The evolutionary conservation of the genes involved suggests that the Notch-SoxB-Snail-EMT network may control neural progenitor selection in many other systems.
Collapse
Affiliation(s)
- Badrul Arefin
- Department of Clinical and Experimental Medicine, Linkoping University, 58185 Linkoping, Sweden
| | - Farjana Parvin
- Department of Clinical and Experimental Medicine, Linkoping University, 58185 Linkoping, Sweden
| | - Shahrzad Bahrampour
- Department of Clinical and Experimental Medicine, Linkoping University, 58185 Linkoping, Sweden
| | - Caroline Bivik Stadler
- Department of Clinical and Experimental Medicine, Linkoping University, 58185 Linkoping, Sweden
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linkoping University, 58185 Linkoping, Sweden; School of Biomedical Sciences, University of Queensland, St. Lucia, QLD 4072, Australia.
| |
Collapse
|
127
|
Bansal P, Madlung J, Schaaf K, Macek B, Bono F. An Interaction Network of RNA-Binding Proteins Involved in Drosophila Oogenesis. Mol Cell Proteomics 2020; 19:1485-1502. [PMID: 32554711 PMCID: PMC8143644 DOI: 10.1074/mcp.ra119.001912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/26/2020] [Indexed: 12/31/2022] Open
Abstract
During Drosophila oogenesis, the localization and translational regulation of maternal transcripts relies on RNA-binding proteins (RBPs). Many of these RBPs localize several mRNAs and may have additional direct interaction partners to regulate their functions. Using immunoprecipitation from whole Drosophila ovaries coupled to mass spectrometry, we examined protein-protein associations of 6 GFP-tagged RBPs expressed at physiological levels. Analysis of the interaction network and further validation in human cells allowed us to identify 26 previously unknown associations, besides recovering several well characterized interactions. We identified interactions between RBPs and several splicing factors, providing links between nuclear and cytoplasmic events of mRNA regulation. Additionally, components of the translational and RNA decay machineries were selectively co-purified with some baits, suggesting a mechanism for how RBPs may regulate maternal transcripts. Given the evolutionary conservation of the studied RBPs, the interaction network presented here provides the foundation for future functional and structural studies of mRNA localization across metazoans.
Collapse
Affiliation(s)
- Prashali Bansal
- Living Systems Institute, University of Exeter, Exeter, UK; Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Johannes Madlung
- Proteome Center Tübingen, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | - Kristina Schaaf
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, Interfaculty Institute for Cell Biology, Eberhard Karls University, Tübingen, Germany
| | - Fulvia Bono
- Living Systems Institute, University of Exeter, Exeter, UK; Max Planck Institute for Developmental Biology, Tübingen, Germany.
| |
Collapse
|
128
|
Mo D, Chen Y, Jiang N, Shen J, Zhang J. Investigation of Isoform Specific Functions of the V-ATPase a Subunit During Drosophila Wing Development. Front Genet 2020; 11:723. [PMID: 32754202 PMCID: PMC7365883 DOI: 10.3389/fgene.2020.00723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
The vacuolar ATPases (V-ATPases) are ATP-dependent proton pumps that play vital roles in eukaryotic cells. Insect V-ATPases are required in nearly all epithelial tissues to regulate a multiplicity of processes including receptor-mediated endocytosis, protein degradation, fluid secretion, and neurotransmission. Composed of fourteen different subunits, several V-ATPase subunits exist in distinct isoforms to perform cell type specific functions. The 100 kD a subunit (Vha100) of V-ATPases are encoded by a family of five genes in Drosophila, but their assignments are not fully understood. Here we report an experimental survey of the Vha100 gene family during Drosophila wing development. A combination of CRISPR-Cas9 mutagenesis, somatic clonal analysis and in vivo RNAi assays is used to characterize the requirement of Vha100 isoforms, and mutants of Vha100-2, Vha100-3, Vha100-4, and Vha100-5 genes were generated. We show that Vha100-3 and Vha100-5 are dispensable for fly development, while Vha100-1 is not critically required in the wing. As for the other two isoforms, we find that Vha100-2 regulates wing cuticle maturation, while Vha100-4 is the single isoform involved in developmental patterning. More specifically, Vha100-4 is required for proper activation of the Wingless signaling pathway during fly wing development. Interestingly, we also find a specific genetic interaction between Vha100-1 and Vha100-4 during wing development. Our results revealed the distinct roles of Vha100 isoforms during insect wing development, providing a rationale for understanding the diverse roles of V-ATPases.
Collapse
Affiliation(s)
- Dongqing Mo
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yao Chen
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Na Jiang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Shen
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Junzheng Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
129
|
Carayon A, Bataillé L, Lebreton G, Dubois L, Pelletier A, Carrier Y, Wystrach A, Vincent A, Frendo JL. Intrinsic control of muscle attachment sites matching. eLife 2020; 9:57547. [PMID: 32706334 PMCID: PMC7431191 DOI: 10.7554/elife.57547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/23/2020] [Indexed: 11/13/2022] Open
Abstract
Myogenesis is an evolutionarily conserved process. Little known, however, is how the morphology of each muscle is determined, such that movements relying upon contraction of many muscles are both precise and coordinated. Each Drosophila larval muscle is a single multinucleated fibre whose morphology reflects expression of distinctive identity Transcription Factors (iTFs). By deleting transcription cis-regulatory modules of one iTF, Collier, we generated viable muscle identity mutants, allowing live imaging and locomotion assays. We show that both selection of muscle attachment sites and muscle/muscle matching is intrinsic to muscle identity and requires transcriptional reprogramming of syncytial nuclei. Live-imaging shows that the staggered muscle pattern involves attraction to tendon cells and heterotypic muscle-muscle adhesion. Unbalance leads to formation of branched muscles, and this correlates with locomotor behavior deficit. Thus, engineering Drosophila muscle identity mutants allows to investigate, in vivo, physiological and mechanical properties of abnormal muscles.
Collapse
Affiliation(s)
- Alexandre Carayon
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Laetitia Bataillé
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Gaëlle Lebreton
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Laurence Dubois
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Aurore Pelletier
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yannick Carrier
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Antoine Wystrach
- Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Alain Vincent
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Jean-Louis Frendo
- Centre de Biologie du Développement (CBD), Toulouse, France.,Centre de Recherche sur la Cognition Animale (CRCA), Toulouse, France.,Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
130
|
Hassan A, Sapir L, Nitsan I, Greenblatt Ben-El RT, Halachmi N, Salzberg A, Tzlil S. A Change in ECM Composition Affects Sensory Organ Mechanics and Function. Cell Rep 2020; 27:2272-2280.e4. [PMID: 31116974 DOI: 10.1016/j.celrep.2019.04.092] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 02/12/2019] [Accepted: 04/18/2019] [Indexed: 11/26/2022] Open
Abstract
Proprioception requires the transduction of muscle-generated deformations into sensory neuronal impulses. In proprioceptive organs, the mechanical coupling between the sensory neuron and the muscle is mediated by a connective structure composed of accessory cells and an extracellular matrix (ECM). Here, we use the fly chordotonal organ (ChO) to investigate how the mechanical properties of the connective element affect mechanosensing. We show that the loss of Pericardin, a major constituent of the ChO ECM, alters the mechanical properties of the ChO resulting in short-wavelength buckling of the accessory cells upon muscle contraction and low compressive strain within the organ. We explain these results using a simplified theoretical model of an elastic beam interacting with an elastic network under a compressive force. We further demonstrate that the transition from compression to bending interferes with the ability of the accessory cells to propagate muscle-generated deformations correctly to the neuron and hence with proper sensing.
Collapse
Affiliation(s)
- Abeer Hassan
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Liel Sapir
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Ido Nitsan
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Rina T Greenblatt Ben-El
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 3109601, Israel
| | - Naomi Halachmi
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 3109601, Israel
| | - Adi Salzberg
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 3109601, Israel
| | - Shelly Tzlil
- Faculty of Mechanical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
131
|
Upregulated TNF/Eiger signaling mediates stem cell recovery and tissue homeostasis during nutrient resupply in Drosophila testis. Sci Rep 2020; 10:11674. [PMID: 32669615 PMCID: PMC7363678 DOI: 10.1038/s41598-020-68313-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Stem cell activity and cell differentiation is robustly influenced by the nutrient availability in the gonads. The signal that connects nutrient availability to gonadal stem cell activity remains largely unknown. In this study, we show that tumor necrosis factor Eiger (Egr) is upregulated in testicular smooth muscles as a response to prolonged protein starvation in Drosophila testis. While Egr is not essential for starvation-induced changes in germline and somatic stem cell numbers, Egr and its receptor Grindelwald influence the recovery dynamics of somatic cyst stem cells (CySCs) upon protein refeeding. Moreover, Egr is also involved in the refeeding-induced, ectopic expression of the CySC self-renewal protein and the accumulation of early germ cells. Egr primarily acts through the Jun N-terminal kinase (JNK) signaling in Drosophila. We show that inhibition of JNK signaling in cyst cells suppresses the refeeding-induced abnormality in both somatic and germ cells. In conclusion, our study reveals both beneficial and detrimental effects of Egr upregulation in the recovery of stem cells and spermatogenesis from prolonged protein starvation.
Collapse
|
132
|
Rapid Homeostatic Turnover of Embryonic ECM during Tissue Morphogenesis. Dev Cell 2020; 54:33-42.e9. [PMID: 32585131 PMCID: PMC7332994 DOI: 10.1016/j.devcel.2020.06.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/27/2020] [Accepted: 06/02/2020] [Indexed: 12/28/2022]
Abstract
The extracellular matrix (ECM) is a polymer network hypothesized to form a stable cellular scaffold. While the ECM can undergo acute remodeling during embryogenesis, it is experimentally difficult to determine whether basal turnover is also important. Most studies of homeostatic turnover assume an initial steady-state balance of production and degradation and measure half-life by quantifying the rate of decay after experimental intervention (e.g., pulse labeling). Here, we present an intervention-free approach to mathematically model basal ECM turnover during embryogenesis by exploiting our ability to live image de novo ECM development in Drosophila to quantify production from initiation to homeostasis. This reveals rapid turnover (half-life ∼7–10 h), which we confirmed by in vivo pulse-chase experiments. Moreover, ECM turnover is partially dependent on proteolysis and network interactions, and slowing turnover affects tissue morphogenesis. These data demonstrate that embryonic ECM undergoes constant replacement, which is likely necessary to maintain network plasticity to accommodate growth and morphogenesis. Labeled ECM in fly embryos can be examined from initiation to homeostasis Quantifying ECM levels to homeostasis allows for modeling of basal turnover rate Embryonic ECM has a half-life of ∼10 h, which was confirmed by pulse-chase analysis Inhibiting MMPs or ECM interactions alters the basal turnover rate
Collapse
|
133
|
Tamamouna V, Panagi M, Theophanous A, Demosthenous M, Michail M, Papadopoulou M, Teloni S, Pitsouli C, Apidianakis Y. Evidence of two types of balance between stem cell mitosis and enterocyte nucleus growth in the Drosophila midgut. Development 2020; 147:147/11/dev189472. [PMID: 32513656 DOI: 10.1242/dev.189472] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/17/2020] [Indexed: 12/21/2022]
Abstract
Systemic and stem cell niche-emanating cytokines and growth factors can promote regeneration, through mitosis. High mitosis, however, predisposes for all types of cancer and, thus, a trade-off exists between regeneration capacity and tissue homeostasis. Here, we study the role of tissue-intrinsic regenerative signaling in stem cell mitosis of adult Drosophila midgut of different genetic backgrounds. We provide evidence of two naturally occurring types of balance between mitosis and enterocyte nucleus growth: one based mostly on stem cell mitosis producing new cells and the other based mostly on the degree of young enterocyte nucleus size increase. Mitosis promotes intestinal host defense to infection, but predisposes for dysplasia in the form of stem cell-like clusters. Enterocyte nucleus growth also promotes host defense, without the drawback of promoting dysplasia. Through quantitative genetics, we identified eiger as an autocrine and paracrine inducer of stem cell mitosis. eiger expression in immature epithelial cells tilts the balance towards mitosis and dysplasia via a positive-feedback loop of highly mitotic stem cells sustaining more small nucleus enterocytes, which in turn supply more Eiger.
Collapse
Affiliation(s)
- Vasilia Tamamouna
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| | - Myrofora Panagi
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| | - Andria Theophanous
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| | - Maria Demosthenous
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| | - Maria Michail
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| | | | - Savvas Teloni
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| | - Yiorgos Apidianakis
- Department of Biological Sciences, University of Cyprus, 2109 Nicosia, Cyprus
| |
Collapse
|
134
|
DeAngelis MW, McGhie EW, Coolon JD, Johnson RI. Mask, a component of the Hippo pathway, is required for Drosophila eye morphogenesis. Dev Biol 2020; 464:53-70. [PMID: 32464117 DOI: 10.1016/j.ydbio.2020.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022]
Abstract
Hippo signaling is an important regulator of tissue size, but it also has a lesser-known role in tissue morphogenesis. Here we use the Drosophila pupal eye to explore the role of the Hippo effector Yki and its cofactor Mask in morphogenesis. We found that Mask is required for the correct distribution and accumulation of adherens junctions and appropriate organization of the cytoskeleton. Accordingly, disrupting mask expression led to severe mis-patterning and similar defects were observed when yki was reduced or in response to ectopic wts. Further, the patterning defects generated by reducing mask expression were modified by Hippo pathway activity. RNA-sequencing revealed a requirement for Mask for appropriate expression of numerous genes during eye morphogenesis. These included genes implicated in cell adhesion and cytoskeletal organization, a comprehensive set of genes that promote cell survival, and numerous signal transduction genes. To validate our transcriptome analyses, we then considered two loci that were modified by Mask activity: FER and Vinc, which have established roles in regulating adhesion. Modulating the expression of either locus modified mask mis-patterning and adhesion phenotypes. Further, expression of FER and Vinc was modified by Yki. It is well-established that the Hippo pathway is responsive to changes in cell adhesion and the cytoskeleton, but our data indicate that Hippo signaling also regulates these structures.
Collapse
Affiliation(s)
- Miles W DeAngelis
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Emily W McGhie
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Joseph D Coolon
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| | - Ruth I Johnson
- Wesleyan University Department of Biology, Middletown CT, 06457, USA.
| |
Collapse
|
135
|
Hudry B, de Goeij E, Mineo A, Gaspar P, Hadjieconomou D, Studd C, Mokochinski JB, Kramer HB, Plaçais PY, Preat T, Miguel-Aliaga I. Sex Differences in Intestinal Carbohydrate Metabolism Promote Food Intake and Sperm Maturation. Cell 2020; 178:901-918.e16. [PMID: 31398343 PMCID: PMC6700282 DOI: 10.1016/j.cell.2019.07.029] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 05/31/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023]
Abstract
Physiology and metabolism are often sexually dimorphic, but the underlying mechanisms remain incompletely understood. Here, we use the intestine of Drosophila melanogaster to investigate how gut-derived signals contribute to sex differences in whole-body physiology. We find that carbohydrate handling is male-biased in a specific portion of the intestine. In contrast to known sexual dimorphisms in invertebrates, the sex differences in intestinal carbohydrate metabolism are extrinsically controlled by the adjacent male gonad, which activates JAK-STAT signaling in enterocytes within this intestinal portion. Sex reversal experiments establish roles for this male-biased intestinal metabolic state in controlling food intake and sperm production through gut-derived citrate. Our work uncovers a male gonad-gut axis coupling diet and sperm production, revealing that metabolic communication across organs is physiologically important. The instructive role of citrate in inter-organ communication might be significant in more biological contexts than previously recognized. Intestinal carbohydrate metabolism is male-biased and region-specific Testes masculinize gut sugar handling by promoting enterocyte JAK-STAT signaling The male intestine secretes citrate to the adjacent testes Gut-derived citrate promotes food intake and sperm maturation
Collapse
Affiliation(s)
- Bruno Hudry
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK; Université Côte d'Azur, CNRS, INSERM, iBV, France.
| | - Eva de Goeij
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Alessandro Mineo
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Pedro Gaspar
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Dafni Hadjieconomou
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Chris Studd
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Joao B Mokochinski
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Holger B Kramer
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Pierre-Yves Plaçais
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - Thomas Preat
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
136
|
Chen Y, Kotian N, Aranjuez G, Chen L, Messer CL, Burtscher A, Sawant K, Ramel D, Wang X, McDonald JA. Protein phosphatase 1 activity controls a balance between collective and single cell modes of migration. eLife 2020; 9:52979. [PMID: 32369438 PMCID: PMC7200163 DOI: 10.7554/elife.52979] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Collective cell migration is central to many developmental and pathological processes. However, the mechanisms that keep cell collectives together and coordinate movement of multiple cells are poorly understood. Using the Drosophila border cell migration model, we find that Protein phosphatase 1 (Pp1) activity controls collective cell cohesion and migration. Inhibition of Pp1 causes border cells to round up, dissociate, and move as single cells with altered motility. We present evidence that Pp1 promotes proper levels of cadherin-catenin complex proteins at cell-cell junctions within the cluster to keep border cells together. Pp1 further restricts actomyosin contractility to the cluster periphery rather than at individual internal border cell contacts. We show that the myosin phosphatase Pp1 complex, which inhibits non-muscle myosin-II (Myo-II) activity, coordinates border cell shape and cluster cohesion. Given the high conservation of Pp1 complexes, this study identifies Pp1 as a major regulator of collective versus single cell migration.
Collapse
Affiliation(s)
- Yujun Chen
- Division of Biology, Kansas State University, Manhattan, United States
| | - Nirupama Kotian
- Division of Biology, Kansas State University, Manhattan, United States
| | - George Aranjuez
- Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Lin Chen
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - C Luke Messer
- Division of Biology, Kansas State University, Manhattan, United States
| | - Ashley Burtscher
- Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Ketki Sawant
- Division of Biology, Kansas State University, Manhattan, United States
| | - Damien Ramel
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Xiaobo Wang
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | | |
Collapse
|
137
|
Homeostatic maintenance and age-related functional decline in the Drosophila ear. Sci Rep 2020; 10:7431. [PMID: 32366993 PMCID: PMC7198581 DOI: 10.1038/s41598-020-64498-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/13/2020] [Indexed: 01/12/2023] Open
Abstract
Age-related hearing loss (ARHL) is a threat to future human wellbeing. Multiple factors contributing to the terminal auditory decline have been identified; but a unified understanding of ARHL - or the homeostatic maintenance of hearing before its breakdown - is missing. We here present an in-depth analysis of homeostasis and ageing in the antennal ears of the fruit fly Drosophila melanogaster. We show that Drosophila, just like humans, display ARHL. By focusing on the phase of dynamic stability prior to the eventual hearing loss we discovered a set of evolutionarily conserved homeostasis genes. The transcription factors Onecut (closest human orthologues: ONECUT2, ONECUT3), Optix (SIX3, SIX6), Worniu (SNAI2) and Amos (ATOH1, ATOH7, ATOH8, NEUROD1) emerged as key regulators, acting upstream of core components of the fly’s molecular machinery for auditory transduction and amplification. Adult-specific manipulation of homeostatic regulators in the fly’s auditory neurons accelerated - or protected against - ARHL.
Collapse
|
138
|
Bataillé L, Colombié N, Pelletier A, Paululat A, Lebreton G, Carrier Y, Frendo JL, Vincent A. Alary muscles and thoracic alary-related muscles are atypical striated muscles involved in maintaining the position of internal organs. Development 2020; 147:dev.185645. [PMID: 32188630 DOI: 10.1242/dev.185645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Alary muscles (AMs) have been described as a component of the cardiac system in various arthropods. Lineage-related thoracic muscles (TARMs), linking the exoskeleton to specific gut regions, have recently been discovered in Drosophila Asymmetrical attachments of AMs and TARMs, to the exoskeleton on one side and internal organs on the other, suggested an architectural function in moving larvae. Here, we analysed the shape and sarcomeric organisation of AMs and TARMs, and imaged their atypical deformability in crawling larvae. We then selectively eliminated AMs and TARMs by targeted apoptosis. Elimination of AMs revealed that AMs are required for suspending the heart in proper intra-haemocelic position and for opening of the heart lumen, and that AMs constrain the curvature of the respiratory tracheal system during crawling; TARMs are required for proper positioning of visceral organs and efficient food transit. AM/TARM cardiac versus visceral attachment depends on Hox control, with visceral attachment being the ground state. TARMs and AMs are the first example of multinucleate striated muscles connecting the skeleton to the cardiac and visceral systems in bilaterians, with multiple physiological functions.
Collapse
Affiliation(s)
- Laetitia Bataillé
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Nathalie Colombié
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Aurore Pelletier
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Achim Paululat
- University of Osnabrück, Department of Biology/Chemistry, Zoology and Developmental Biology, Barbarastraße 11, 49076 Osnabrück, Germany
| | - Gaëlle Lebreton
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Yannick Carrier
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Jean-Louis Frendo
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Alain Vincent
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse 3, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| |
Collapse
|
139
|
Cerqueira Campos F, Dennis C, Alégot H, Fritsch C, Isabella A, Pouchin P, Bardot O, Horne-Badovinac S, Mirouse V. Oriented basement membrane fibrils provide a memory for F-actin planar polarization via the Dystrophin-Dystroglycan complex during tissue elongation. Development 2020; 147:dev.186957. [PMID: 32156755 DOI: 10.1242/dev.186957] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/24/2020] [Indexed: 12/31/2022]
Abstract
How extracellular matrix contributes to tissue morphogenesis is still an open question. In the Drosophila ovarian follicle, it has been proposed that after Fat2-dependent planar polarization of the follicle cell basal domain, oriented basement membrane (BM) fibrils and F-actin stress fibers constrain follicle growth, promoting its axial elongation. However, the relationship between BM fibrils and stress fibers and their respective impact on elongation are unclear. We found that Dystroglycan (Dg) and Dystrophin (Dys) are involved in BM fibril deposition. Moreover, they also orient stress fibers, by acting locally and in parallel to Fat2. Importantly, Dg-Dys complex-mediated cell-autonomous control of F-actin fiber orientation relies on the preceding BM fibril deposition, indicating two distinct but interdependent functions. Thus, the Dg-Dys complex works as a crucial organizer of the epithelial basal domain, regulating both F-actin and BM. Furthermore, BM fibrils act as a persistent cue for the orientation of stress fibers that are the main effector of elongation.
Collapse
Affiliation(s)
- Fabiana Cerqueira Campos
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Cynthia Dennis
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Hervé Alégot
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Cornelia Fritsch
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Adam Isabella
- Committee on Development, Regeneration and Stem Cell Biology, and Department of Molecular Genetics and Cell Biology - The University of Chicago, 920 East 58th Street, Chicago IL 60653, USA
| | - Pierre Pouchin
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Olivier Bardot
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| | - Sally Horne-Badovinac
- Committee on Development, Regeneration and Stem Cell Biology, and Department of Molecular Genetics and Cell Biology - The University of Chicago, 920 East 58th Street, Chicago IL 60653, USA
| | - Vincent Mirouse
- iGReD (Institute of Genetics, Reproduction and Development), Université Clermont Auvergne - UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, 28 Place Henri-Dunant, 63000 Clermont-Ferrand, France
| |
Collapse
|
140
|
Li S, Tian A, Li S, Han Y, Wang B, Jiang J. Gilgamesh (Gish)/CK1γ regulates tissue homeostasis and aging in adult Drosophila midgut. J Cell Biol 2020; 219:133831. [PMID: 32328627 PMCID: PMC7147094 DOI: 10.1083/jcb.201909103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/03/2020] [Accepted: 01/16/2020] [Indexed: 12/26/2022] Open
Abstract
Adult tissues and organs rely on resident stem cells to generate new cells that replenish damaged cells. To maintain homeostasis, stem cell activity needs to be tightly controlled throughout the adult life. Here, we show that the membrane-associated kinase Gilgamesh (Gish)/CK1γ maintains Drosophila adult midgut homeostasis by restricting JNK pathway activity and that Gish is essential for intestinal stem cell (ISC) maintenance under stress conditions. Inactivation of Gish resulted in aberrant JNK pathway activation and excessive production of multiple cytokines and growth factors that drive ISC overproliferation. Mechanistically, Gish restricts JNK activation by phosphorylating and destabilizing a small GTPase, Rho1. Interestingly, we find that Gish expression is down-regulated in aging guts and that increasing Gish activity in aging guts can restore tissue homeostasis. Hence, our study identifies Gish/CK1γ as a novel regulator of Rho1 and gatekeeper of tissue homeostasis whose activity is compromised in aging guts.
Collapse
Affiliation(s)
- Shuangxi Li
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Aiguo Tian
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shuang Li
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yuhong Han
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Bing Wang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jin Jiang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
141
|
Li Y, Zhang F, Jiang N, Liu T, Shen J, Zhang J. Decapentaplegic signaling regulates Wingless ligand production and target activation during
Drosophila
wing development. FEBS Lett 2020; 594:1176-1186. [DOI: 10.1002/1873-3468.13713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Yunlong Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management College of Plant Protection China Agricultural University Beijing China
| | - Fengchao Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management College of Plant Protection China Agricultural University Beijing China
| | - Na Jiang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management College of Plant Protection China Agricultural University Beijing China
| | - Tong Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management College of Plant Protection China Agricultural University Beijing China
| | - Jie Shen
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management College of Plant Protection China Agricultural University Beijing China
| | - Junzheng Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management College of Plant Protection China Agricultural University Beijing China
| |
Collapse
|
142
|
Kato K, Orihara-Ono M, Awasaki T. Multiple lineages enable robust development of the neuropil-glia architecture in adult Drosophila. Development 2020; 147:dev184085. [PMID: 32051172 DOI: 10.1242/dev.184085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022]
Abstract
Neural remodeling is essential for the development of a functional nervous system and has been extensively studied in the metamorphosis of Drosophila Despite the crucial roles of glial cells in brain functions, including learning and behavior, little is known of how adult glial cells develop in the context of neural remodeling. Here, we show that the architecture of neuropil-glia in the adult Drosophila brain, which is composed of astrocyte-like glia (ALG) and ensheathing glia (EG), robustly develops from two different populations in the larva: the larval EG and glial cell missing-positive (gcm+ ) cells. Whereas gcm+ cells proliferate and generate adult ALG and EG, larval EG dedifferentiate, proliferate and redifferentiate into the same glial subtypes. Each glial lineage occupies a certain brain area complementary to the other, and together they form the adult neuropil-glia architecture. Both lineages require the FGF receptor Heartless to proliferate, and the homeoprotein Prospero to differentiate into ALG. Lineage-specific inhibition of gliogenesis revealed that each lineage compensates for deficiency in the proliferation of the other. Together, the lineages ensure the robust development of adult neuropil-glia, thereby ensuring a functional brain.
Collapse
Affiliation(s)
- Kentaro Kato
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| | - Minako Orihara-Ono
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| | - Takeshi Awasaki
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| |
Collapse
|
143
|
Bosch JA, Knight S, Kanca O, Zirin J, Yang-Zhou D, Hu Y, Rodiger J, Amador G, Bellen HJ, Perrimon N, Mohr SE. Use of the CRISPR-Cas9 System in Drosophila Cultured Cells to Introduce Fluorescent Tags into Endogenous Genes. CURRENT PROTOCOLS IN MOLECULAR BIOLOGY 2020; 130:e112. [PMID: 31869524 PMCID: PMC7213786 DOI: 10.1002/cpmb.112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The CRISPR-Cas9 system makes it possible to cause double-strand breaks in specific regions, inducing repair. In the presence of a donor construct, repair can involve insertion or 'knock-in' of an exogenous cassette. One common application of knock-in technology is to generate cell lines expressing fluorescently tagged endogenous proteins. The standard approach relies on production of a donor plasmid with ∼500 to 1000 bp of homology on either side of an insertion cassette that contains the fluorescent protein open reading frame (ORF). We present two alternative methods for knock-in of fluorescent protein ORFs into Cas9-expressing Drosophila S2R+ cultured cells, the single-stranded DNA (ssDNA) Drop-In method and the CRISPaint universal donor method. Both methods eliminate the need to clone a large plasmid donor for each target. We discuss the advantages and limitations of the standard, ssDNA Drop-In, and CRISPaint methods for fluorescent protein tagging in Drosophila cultured cells. © 2019 by John Wiley & Sons, Inc. Basic Protocol 1: Knock-in into Cas9-positive S2R+ cells using the ssDNA Drop-In approach Basic Protocol 2: Knock-in into Cas9-positive S2R+ cells by homology-independent insertion of universal donor plasmids that provide mNeonGreen (CRISPaint method) Support Protocol 1: sgRNA design and cloning Support Protocol 2: ssDNA donor synthesis Support Protocol 3: Transfection using Effectene Support Protocol 4: Electroporation of S2R+-MT::Cas9 Drosophila cells Support Protocol 5: Single-cell isolation of fluorescent cells using FACS.
Collapse
Affiliation(s)
- Justin A Bosch
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Shannon Knight
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
| | - Jonathan Zirin
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
| | - Donghui Yang-Zhou
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
| | - Jonathan Rodiger
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
| | - Gabriel Amador
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, Texas
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
- Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts
| | - Stephanie E Mohr
- Department of Genetics, Harvard Medical School, Boston, Massachusetts
- Drosophila RNAi Screening Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
144
|
Lepesant JMJ, Iampietro C, Galeota E, Augé B, Aguirrenbengoa M, Mercé C, Chaubet C, Rocher V, Haenlin M, Waltzer L, Pelizzola M, Di Stefano L. A dual role of dLsd1 in oogenesis: regulating developmental genes and repressing transposons. Nucleic Acids Res 2020; 48:1206-1224. [PMID: 31799607 PMCID: PMC7026653 DOI: 10.1093/nar/gkz1142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 11/05/2019] [Accepted: 11/23/2019] [Indexed: 11/14/2022] Open
Abstract
The histone demethylase LSD1 is a key chromatin regulator that is often deregulated in cancer. Its ortholog, dLsd1 plays a crucial role in Drosophila oogenesis; however, our knowledge of dLsd1 function is insufficient to explain its role in the ovary. Here, we have performed genome-wide analysis of dLsd1 binding in the ovary, and we document that dLsd1 is preferentially associated to the transcription start site of developmental genes. We uncovered an unanticipated interplay between dLsd1 and the GATA transcription factor Serpent and we report an unexpected role for Serpent in oogenesis. Besides, our transcriptomic data show that reducing dLsd1 levels results in ectopic transposable elements (TE) expression correlated with changes in H3K4me2 and H3K9me2 at TE loci. In addition, our results suggest that dLsd1 is required for Piwi dependent TE silencing. Hence, we propose that dLsd1 plays crucial roles in establishing specific gene expression programs and in repressing transposons during oogenesis.
Collapse
Affiliation(s)
- Julie M J Lepesant
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Carole Iampietro
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Eugenia Galeota
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan 20139, Italy
| | - Benoit Augé
- CBD, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Marion Aguirrenbengoa
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Clemèntine Mercé
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France.,School of Biological Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Camille Chaubet
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Vincent Rocher
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Marc Haenlin
- CBD, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Lucas Waltzer
- CBD, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France.,Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand F-63000, France
| | - Mattia Pelizzola
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan 20139, Italy
| | - Luisa Di Stefano
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| |
Collapse
|
145
|
Ramond E, Petrignani B, Dudzic JP, Boquete J, Poidevin M, Kondo S, Lemaitre B. The adipokine NimrodB5 regulates peripheral hematopoiesis in
Drosophila. FEBS J 2020; 287:3399-3426. [DOI: 10.1111/febs.15237] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/08/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Elodie Ramond
- Global Health Institute School of Life Science École Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Bianca Petrignani
- Global Health Institute School of Life Science École Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Jan Paul Dudzic
- Global Health Institute School of Life Science École Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Jean‐Philippe Boquete
- Global Health Institute School of Life Science École Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Mickaël Poidevin
- Centre de Génétique Moléculaire CNRS Université Pierre et Marie Curie Gif‐sur‐Yvette France
| | - Shu Kondo
- Invertebrate Genetics Laboratory Genetic Strains Research Center National Institute of Genetics Mishima Japan
| | - Bruno Lemaitre
- Global Health Institute School of Life Science École Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| |
Collapse
|
146
|
Nikonova E, Kao SY, Spletter ML. Contributions of alternative splicing to muscle type development and function. Semin Cell Dev Biol 2020; 104:65-80. [PMID: 32070639 DOI: 10.1016/j.semcdb.2020.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/30/2022]
Abstract
Animals possess a wide variety of muscle types that support different kinds of movements. Different muscles have distinct locations, morphologies and contractile properties, raising the question of how muscle diversity is generated during development. Normal aging processes and muscle disorders differentially affect particular muscle types, thus understanding how muscles normally develop and are maintained provides insight into alterations in disease and senescence. As muscle structure and basic developmental mechanisms are highly conserved, many important insights into disease mechanisms in humans as well as into basic principles of muscle development have come from model organisms such as Drosophila, zebrafish and mouse. While transcriptional regulation has been characterized to play an important role in myogenesis, there is a growing recognition of the contributions of alternative splicing to myogenesis and the refinement of muscle function. Here we review our current understanding of muscle type specific alternative splicing, using examples of isoforms with distinct functions from both vertebrates and Drosophila. Future exploration of the vast potential of alternative splicing to fine-tune muscle development and function will likely uncover novel mechanisms of isoform-specific regulation and a more holistic understanding of muscle development, disease and aging.
Collapse
Affiliation(s)
- Elena Nikonova
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, 82152 Martinsried-Planegg, Germany
| | - Shao-Yen Kao
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, 82152 Martinsried-Planegg, Germany
| | - Maria L Spletter
- Biomedical Center, Department of Physiological Chemistry, Ludwig-Maximilians-Universität München, Großhaderner Str. 9, 82152 Martinsried-Planegg, Germany; Center for Integrated Protein Science Munich (CIPSM) at the Department of Chemistry, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
147
|
Slaidina M, Banisch TU, Gupta S, Lehmann R. A single-cell atlas of the developing Drosophila ovary identifies follicle stem cell progenitors. Genes Dev 2020; 34:239-249. [PMID: 31919193 PMCID: PMC7000915 DOI: 10.1101/gad.330464.119] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022]
Abstract
Addressing the complexity of organogenesis at a system-wide level requires a complete understanding of adult cell types, their origin, and precursor relationships. The Drosophila ovary has been a model to study how coordinated stem cell units, germline, and somatic follicle stem cells maintain and renew an organ. However, lack of cell type-specific tools have limited our ability to study the origin of individual cell types and stem cell units. Here, we used a single-cell RNA sequencing approach to uncover all known cell types of the developing ovary, reveal transcriptional signatures, and identify cell type-specific markers for lineage tracing. Our study identifies a novel cell type corresponding to the elusive follicle stem cell precursors and predicts subtypes of known cell types. Altogether, we reveal a previously unanticipated complexity of the developing ovary and provide a comprehensive resource for the systematic analysis of ovary morphogenesis.
Collapse
Affiliation(s)
- Maija Slaidina
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York 10016, USA
| | - Torsten U Banisch
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York 10016, USA
| | - Selena Gupta
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York 10016, USA
| | - Ruth Lehmann
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York 10016, USA
| |
Collapse
|
148
|
Hoyer N, Zielke P, Hu C, Petersen M, Sauter K, Scharrenberg R, Peng Y, Kim CC, Han C, Parrish JZ, Soba P. Ret and Substrate-Derived TGF-β Maverick Regulate Space-Filling Dendrite Growth in Drosophila Sensory Neurons. Cell Rep 2020; 24:2261-2272.e5. [PMID: 30157422 DOI: 10.1016/j.celrep.2018.07.092] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/17/2018] [Accepted: 07/27/2018] [Indexed: 12/19/2022] Open
Abstract
Dendrite morphogenesis is a highly regulated process that gives rise to stereotyped receptive fields, which are required for proper neuronal connectivity and function. Specific classes of neurons, including Drosophila class IV dendritic arborization (C4da) neurons, also feature complete space-filling growth of dendrites. In this system, we have identified the substrate-derived TGF-β ligand maverick (mav) as a developmental signal promoting space-filling growth through the neuronal Ret receptor. Both are necessary for radial spreading of C4da neuron dendrites, and Ret is required for neuronal uptake of Mav. Moreover, local changes in Mav levels result in directed dendritic growth toward regions with higher ligand availability. Our results suggest that Mav acts as a substrate-derived secreted signal promoting dendrite growth within not-yet-covered areas of the receptive field to ensure space-filling dendritic growth.
Collapse
Affiliation(s)
- Nina Hoyer
- Research Group Neuronal Patterning and Connectivity, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Philip Zielke
- Research Group Neuronal Patterning and Connectivity, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Chun Hu
- Research Group Neuronal Patterning and Connectivity, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Meike Petersen
- Research Group Neuronal Patterning and Connectivity, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Kathrin Sauter
- Research Group Neuronal Patterning and Connectivity, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Robin Scharrenberg
- Research Group Neuronal Development, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Yun Peng
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | | | - Chun Han
- Weill Institute for Cell and Molecular Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Peter Soba
- Research Group Neuronal Patterning and Connectivity, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
149
|
Yatsenko AS, Kucherenko MM, Xie Y, Aweida D, Urlaub H, Scheibe RJ, Cohen S, Shcherbata HR. Profiling of the muscle-specific dystroglycan interactome reveals the role of Hippo signaling in muscular dystrophy and age-dependent muscle atrophy. BMC Med 2020; 18:8. [PMID: 31959160 PMCID: PMC6971923 DOI: 10.1186/s12916-019-1478-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dystroglycanopathies are a group of inherited disorders characterized by vast clinical and genetic heterogeneity and caused by abnormal functioning of the ECM receptor dystroglycan (Dg). Remarkably, among many cases of diagnosed dystroglycanopathies, only a small fraction can be linked directly to mutations in Dg or its regulatory enzymes, implying the involvement of other, not-yet-characterized, Dg-regulating factors. To advance disease diagnostics and develop new treatment strategies, new approaches to find dystroglycanopathy-related factors should be considered. The Dg complex is highly evolutionarily conserved; therefore, model genetic organisms provide excellent systems to address this challenge. In particular, Drosophila is amenable to experiments not feasible in any other system, allowing original insights about the functional interactors of the Dg complex. METHODS To identify new players contributing to dystroglycanopathies, we used Drosophila as a genetic muscular dystrophy model. Using mass spectrometry, we searched for muscle-specific Dg interactors. Next, in silico analyses allowed us to determine their association with diseases and pathological conditions in humans. Using immunohistochemical, biochemical, and genetic interaction approaches followed by the detailed analysis of the muscle tissue architecture, we verified Dg interaction with some of the discovered factors. Analyses of mouse muscles and myocytes were used to test if interactions are conserved in vertebrates. RESULTS The muscle-specific Dg complexome revealed novel components that influence the efficiency of Dg function in the muscles. We identified the closest human homologs for Dg-interacting partners, determined their significant enrichment in disease-associations, and verified some of the newly identified Dg interactions. We found that Dg associates with two components of the mechanosignaling Hippo pathway: the WW domain-containing proteins Kibra and Yorkie. Importantly, this conserved interaction manages adult muscle size and integrity. CONCLUSIONS The results presented in this study provide a new list of muscle-specific Dg interactors, further analysis of which could aid not only in the diagnosis of muscular dystrophies, but also in the development of new therapeutics. To regulate muscle fitness during aging and disease, Dg associates with Kibra and Yorkie and acts as a transmembrane Hippo signaling receptor that transmits extracellular information to intracellular signaling cascades, regulating muscle gene expression.
Collapse
Affiliation(s)
- Andriy S Yatsenko
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Mariya M Kucherenko
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Institute of Physiology, Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Yuanbin Xie
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: University Medical Center, Centre for Anatomy, Institute of Neuroanatomy, Georg-August-University Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Dina Aweida
- Faculty of Biology, Technion, 32000, Haifa, Israel
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Research Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Bioanalytics Institute for Clinical Chemistry, University Medical Center Goettingen, Robert Koch Strasse 40, 37075, Göttingen, Germany
| | - Renate J Scheibe
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | | | - Halyna R Shcherbata
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany. .,Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
150
|
Antioxidant Therapy in Parkinson's Disease: Insights from Drosophila melanogaster. Antioxidants (Basel) 2020; 9:antiox9010052. [PMID: 31936094 PMCID: PMC7023233 DOI: 10.3390/antiox9010052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) play an important role as endogenous mediators in several cellular signalling pathways. However, at high concentrations they can also exert deleterious effects by reacting with many macromolecules including DNA, proteins and lipids. The precise balance between ROS production and their removal via numerous enzymatic and nonenzymatic molecules is of fundamental importance for cell survival. Accordingly, many neurodegenerative disorders, including Parkinson’s disease (PD), are associated with excessive levels of ROS, which induce oxidative damage. With the aim of coping with the progression of PD, antioxidant compounds are currently receiving increasing attention as potential co-adjuvant molecules in the treatment of these diseases, and many studies have been performed to evaluate the purported protective effects of several antioxidant molecules. In the present review, we present and discuss the relevance of the use of Drosophila melanogaster as an animal model with which to evaluate the therapeutic potential of natural and synthetic antioxidants. The conservation of most of the PD-related genes between humans and D. melanogaster, along with the animal’s rapid life cycle and the versatility of genetic tools, makes fruit flies an ideal experimental system for rapid screening of antioxidant-based treatments.
Collapse
|