1
|
Martins CS, Iv F, Suman SK, Panagiotou TC, Sidor C, Ruso-López M, Plancke CN, Omi S, Pagès R, Gomes M, Llewellyn A, Bandi SR, Ramond L, Arbizzani F, Rimoli CV, Schnorrer F, Robin F, Wilde A, LeGoff L, Pedelacq JD, Jégou A, Cabantous S, Rincon SA, Chandre C, Brasselet S, Mavrakis M. Genetically encoded reporters of actin filament organization in living cells and tissues. Cell 2025; 188:2540-2559.e27. [PMID: 40179884 DOI: 10.1016/j.cell.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 12/09/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
The cytoskeletal protein actin is crucial for cell shape and integrity throughout eukaryotes. Actin filaments perform essential biological functions, including muscle contraction, cell division, and tissue morphogenesis. These diverse activities are achieved through the ability of actin filaments to be arranged into precise architectures. Much progress has been made in defining the proteome of the actin cytoskeleton, but a detailed appreciation of the dynamic organizational state of the actin filaments themselves has been hindered by available tools. Fluorescence polarization microscopy is uniquely placed for measuring actin filament organization by exploiting the sensitivity of polarized light excitation to the orientation of fluorophores attached to actin filaments. By engineering fusions of five widely used actin localization reporters to fluorescent proteins with constrained mobility, we have succeeded in developing genetically encoded, green- and red-fluorescent-protein-based reporters for non-invasive, quantitative measurements of actin filament organization in living cells and tissues by fluorescence polarization microscopy.
Collapse
Affiliation(s)
- Carla Silva Martins
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - François Iv
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Shashi Kumar Suman
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Thomas C Panagiotou
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1M1, Canada
| | - Clara Sidor
- Aix Marseille Univ, CNRS, IBDM, Turing Centre for Living Systems, 13009 Marseille, France
| | - María Ruso-López
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca 37007, Spain
| | - Camille N Plancke
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Shizue Omi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Rebecca Pagès
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Maxime Gomes
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Alexander Llewellyn
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Sourish Reddy Bandi
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Laurie Ramond
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | | | - Caio Vaz Rimoli
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Frank Schnorrer
- Aix Marseille Univ, CNRS, IBDM, Turing Centre for Living Systems, 13009 Marseille, France
| | - François Robin
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 75005 Paris, France
| | - Andrew Wilde
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1M1, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1M1, Canada
| | - Loïc LeGoff
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France
| | - Jean-Denis Pedelacq
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 31077 Toulouse, France
| | - Antoine Jégou
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Stéphanie Cabantous
- Centre de Recherche en Cancérologie de Toulouse (CRCT), Inserm, Université Paul Sabatier - Toulouse III, CNRS, 31037 Toulouse, France
| | - Sergio A Rincon
- Instituto de Biología Funcional y Genómica and Departamento de Microbiología y Genética, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad de Salamanca, Salamanca 37007, Spain
| | | | - Sophie Brasselet
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France.
| | - Manos Mavrakis
- Institut Fresnel, CNRS UMR7249, Aix Marseille Univ, Centrale Med, 13013 Marseille, France.
| |
Collapse
|
2
|
Sorensen RM, Savić-Zdravković D, Jovanović B. Changes in the wing shape and size in fruit flies exposed to micro and nanoplastics. CHEMOSPHERE 2024; 363:142821. [PMID: 38986775 DOI: 10.1016/j.chemosphere.2024.142821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/12/2024]
Abstract
Geometric morphometrics analysis (GMA) is a well-known technique to identify minute changes in Drosophila wings. This study aimed to determine potential changes in Drosophila wings shape and size after exposure to polystyrene nanoplastics (NPs) (50 nm) and microplastics (MPs) (1 μm). Flies were exposed from eggs to pupal eclosion and analyzed using GMA. Results revealed a difference in shape and size between male and female wings, as expected, due to sexual dimorphism. Therefore, wings were analyzed by sex. Wings of MPs and NPs treated females were elongated compared to controls and had a constriction of the wing joint. Additionally, MPs treated female flies had the most dissimilar shape compared to controls. In male flies, NPs flies had smaller wings compared to MPs and control flies. Compared to control, NPs wings of males were shrunken at the joint and in the entire proximal region of the wing. However, male MPs wings had a narrower anal region and were slightly elongated. These results reveal that wing shape and size can change in a different way based on the sex of the flies and size of plastic particles that larvae interacted with. All the changes in the wings occurred only within the normally allowed wing variation and treatment with NPs/MPs did not cause development of the aberrant phenotypes. Results can pave the way for further understanding of how MPs and NPs can alter phenotypes of flies.
Collapse
Affiliation(s)
- Rachel M Sorensen
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, USA.
| | - Dimitrija Savić-Zdravković
- Department of Biology and Ecology, Faculty of Sciences and Mathematics, University of Niš, Višegradska 33, Niš, 18000, Serbia.
| | - Boris Jovanović
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, USA.
| |
Collapse
|
3
|
Fischer F, Ernst L, Frey A, Holstein K, Prasad D, Weichselberger V, Balaji R, Classen AK. A mismatch in the expression of cell surface molecules induces tissue-intrinsic defense against aberrant cells. Curr Biol 2024; 34:980-996.e6. [PMID: 38350446 DOI: 10.1016/j.cub.2024.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 09/29/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
Tissue-intrinsic error correction enables epithelial cells to detect abnormal neighboring cells and facilitate their removal from the tissue. One of these pathways, "interface surveillance," is triggered by cells with aberrant developmental and cell-fate-patterning pathways. It remains unknown which molecular mechanisms provide cells with the ability to compare fate between neighboring cells. We demonstrate that Drosophila imaginal discs express an array of cell surface molecules previously implicated in neuronal axon guidance processes. They include members of the Robo, Teneurin, Ephrin, Toll-like, or atypical cadherin families. Importantly, a mismatch in expression levels of these cell surface molecules between adjacent cells is sufficient to induce interface surveillance, indicating that differences in expression levels between neighboring cells, rather than their absolute expression levels, are crucial. Specifically, a mismatch in Robo2 and Robo3, but not Robo1, induces enrichment of actin, myosin II, and Ena/Vasp, as well as activation of JNK and apoptosis at clonal interfaces. Moreover, Robo2 can induce interface surveillance independently of its cytosolic domain and without the need for the Robo-ligand Slit. The expression of Robo2 and other cell surface molecules, such as Teneurins or the Ephrin receptor is regulated by fate-patterning pathways intrinsic and extrinsic to the wing disc, as well as by expression of oncogenic RasV12. Combined, we demonstrate that neighboring cells respond to a mismatch in surface code patterns mediated by specific transmembrane proteins and reveal a novel function for these cell surface proteins in cell fate recognition and removal of aberrant cells during development and homeostasis of epithelial tissues.
Collapse
Affiliation(s)
- Friedericke Fischer
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Laurin Ernst
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Anna Frey
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Katrin Holstein
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Deepti Prasad
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Vanessa Weichselberger
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Aix Marseille University, CNRS, UMR 7288, IBDM, 13288 Marseille, France
| | - Ramya Balaji
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
4
|
Liu SP, Yin HD, Li WJ, Qin ZH, Yang Y, Huang ZZ, Zong L, Liu XK, Du Z, Fan WL, Zhang YQ, Zhang D, Zhang YE, Liu XY, Yang D, Ge SQ. The Morphological Transformation of the Thorax during the Eclosion of Drosophila melanogaster (Diptera: Drosophilidae). INSECTS 2023; 14:893. [PMID: 37999092 PMCID: PMC10671814 DOI: 10.3390/insects14110893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
The model organism Drosophila melanogaster, as a species of Holometabola, undergoes a series of transformations during metamorphosis. To deeply understand its development, it is crucial to study its anatomy during the key developmental stages. We describe the anatomical systems of the thorax, including the endoskeleton, musculature, nervous ganglion, and digestive system, from the late pupal stage to the adult stage, based on micro-CT and 3D visualizations. The development of the endoskeleton causes original and insertional changes in muscles. Several muscles change their shape during development in a non-uniform manner with respect to both absolute and relative size; some become longer and broader, while others shorten and become narrower. Muscular shape may vary during development. The number of muscular bundles also increases or decreases. Growing muscles are probably anchored by the tissues in the stroma. Some muscles and tendons are absent in the adult stage, possibly due to the hardened sclerites. Nearly all flight muscles are present by the third day of the pupal stage, which may be due to the presence of more myofibers with enough mitochondria to support flight power. There are sexual differences in the same developmental period. In contrast to the endodermal digestive system, the functions of most thoracic muscles change in the development from the larva to the adult in order to support more complex locomotion under the control of a more structured ventral nerve cord based on the serial homology proposed herein.
Collapse
Affiliation(s)
- Si-Pei Liu
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
| | - Hao-Dong Yin
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| | - Wen-Jie Li
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| | - Zhuang-Hui Qin
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
| | - Yi Yang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| | - Zheng-Zhong Huang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
| | - Le Zong
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| | - Xiao-Kun Liu
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| | - Zhong Du
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| | - Wei-Li Fan
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
| | - Ya-Qiong Zhang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| | - Dan Zhang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Yong E. Zhang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
| | - Xing-Yue Liu
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China; (X.-Y.L.); (D.Y.)
| | - Ding Yang
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China; (X.-Y.L.); (D.Y.)
| | - Si-Qin Ge
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (S.-P.L.); (H.-D.Y.); (W.-J.L.); (Z.-H.Q.); (Y.Y.); (Z.-Z.H.); (L.Z.); (X.-K.L.); (Z.D.); (W.-L.F.); (Y.-Q.Z.); (D.Z.); (Y.E.Z.)
- University of Chinese Academy of Sciences, Beijing 100086, China
| |
Collapse
|
5
|
R S, K N. Teratogenic impacts of Antiepileptic drugs on development, behavior and reproduction in Drosophila melanogaster. Neurotoxicol Teratol 2023; 100:107305. [PMID: 37805079 DOI: 10.1016/j.ntt.2023.107305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/27/2023] [Accepted: 10/02/2023] [Indexed: 10/09/2023]
Abstract
Clobazam (CLB) and Vigabatrin (VGB) are the two widely used Antiepileptic drugs, which may have teratogenic potentiality and it has been evaluated in the fruit fly Drosophila melanogaster. These different concentrations of CLB (0.156, 0.25, and 0.312 μg/ml) and VGB (17.6, 22, and 44 μg/ml) were used to evaluate the life-history parameters, developmental, and behavioral abnormalities. The results revealed that life-history parameters (fecundity, fertility, larval and pupal mortality) were significantly affected along with varied developmental duration, and pupal and adult deformities in flies on exposure of CLB and VGB in concentration dependent manner. The present study demonstrated that the prenatal treatment of CLB and VGB has displayed clear teratogenic potentiality with various deformities in the fruit fly. The findings could be correlated with the various abnormalities in human caused by the use of AEDs.
Collapse
Affiliation(s)
- Shamapari R
- Department of PG Studies and Research in Applied Zoology, Kuvempu University, Jnana Sahyadri, Shankaraghatta, Karnataka 577451, India
| | - Nagaraj K
- Department of PG Studies and Research in Applied Zoology, Kuvempu University, Jnana Sahyadri, Shankaraghatta, Karnataka 577451, India.
| |
Collapse
|
6
|
Chen Y, Li H, Yi TC, Shen J, Zhang J. Notch Signaling in Insect Development: A Simple Pathway with Diverse Functions. Int J Mol Sci 2023; 24:14028. [PMID: 37762331 PMCID: PMC10530718 DOI: 10.3390/ijms241814028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Notch signaling is an evolutionarily conserved pathway which functions between adjacent cells to establish their distinct identities. Despite operating in a simple mechanism, Notch signaling plays remarkably diverse roles in development to regulate cell fate determination, organ growth and tissue patterning. While initially discovered and characterized in the model insect Drosophila melanogaster, recent studies across various insect species have revealed the broad involvement of Notch signaling in shaping insect tissues. This review focuses on providing a comprehensive picture regarding the roles of the Notch pathway in insect development. The roles of Notch in the formation and patterning of the insect embryo, wing, leg, ovary and several specific structures, as well as in physiological responses, are summarized. These results are discussed within the developmental context, aiming to deepen our understanding of the diversified functions of the Notch signaling pathway in different insect species.
Collapse
Affiliation(s)
- Yao Chen
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| | - Haomiao Li
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| | - Tian-Ci Yi
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, Institute of Entomology, Guizhou University, Guiyang 550025, China
| | - Jie Shen
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| | - Junzheng Zhang
- Department of Plant Biosecurity and MOA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China; (Y.C.)
| |
Collapse
|
7
|
Ostalé CM, Vega-Cuesta P, González T, López-Varea A, de Celis JF. RNAi screen in the Drosophila wing of genes encoding proteins related to cytoskeleton organization and cell division. Dev Biol 2023; 498:61-76. [PMID: 37015290 DOI: 10.1016/j.ydbio.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/04/2023]
Abstract
Cell division and cytoskeleton organization are fundamental processes participating in the development of Drosophila imaginal discs. In this manuscript we describe the phenotypes in the adult fly wing generated by knockdowns of 85% of Drosophila genes encoding proteins likely related to the regulation of cell division and cytoskeleton organization. We also compile a molecular classification of these proteins into classes that describe their expected or known main biochemical characteristics, as well as mRNA expression in the wing disc and likely protein subcellular localization for a subset of these genes. Finally, we analyze in more detail one protein family of cytoskeleton genes (Arp2/3 complex), and define the consequences of interfering with cell division for wing growth and patterning.
Collapse
|
8
|
Diaz-Torres E, Muñoz-Nava LM, Nahmad M. Coupling cell proliferation rates to the duration of recruitment controls final size of the Drosophila wing. Proc Biol Sci 2022; 289:20221167. [PMID: 36476003 PMCID: PMC9554725 DOI: 10.1098/rspb.2022.1167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022] Open
Abstract
Organ growth driven by cell proliferation is an exponential process. As a result, even small variations in proliferation rates, when integrated over a relatively long developmental time, will lead to large differences in size. How organs robustly control their final size despite perturbations in cell proliferation rates throughout development is a long-standing question in biology. Using a mathematical model, we show that in the developing wing of the fruit fly, Drosophila melanogaster, variations in proliferation rates of wing-committed cells are inversely proportional to the duration of cell recruitment, a differentiation process in which a population of undifferentiated cells adopt the wing fate by expressing the selector gene, vestigial. A time-course experiment shows that vestigial-expressing cells increase exponentially while recruitment takes place, but slows down when recruitable cells start to vanish, suggesting that undifferentiated cells may be driving proliferation of wing-committed cells. When this observation is incorporated in our model, we show that the duration of cell recruitment robustly determines a final wing size even when cell proliferation rates of wing-committed cells are perturbed. Finally, we show that this control mechanism fails when perturbations in proliferation rates affect both wing-committed and recruitable cells, providing an experimentally testable hypothesis of our model.
Collapse
Affiliation(s)
- Elizabeth Diaz-Torres
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Luis Manuel Muñoz-Nava
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| |
Collapse
|
9
|
Valencia-Expósito A, Gómez-Lamarca MJ, Widmann TJ, Martín-Bermudo MD. Integrins Cooperate With the EGFR/Ras Pathway to Preserve Epithelia Survival and Architecture in Development and Oncogenesis. Front Cell Dev Biol 2022; 10:892691. [PMID: 35769262 PMCID: PMC9234701 DOI: 10.3389/fcell.2022.892691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Adhesion to the extracellular matrix (ECM) is required for normal epithelial cell survival. Disruption of this interaction leads to a specific type of apoptosis known as anoikis. Yet, there are physiological and pathological situations in which cells not connected to the ECM are protected from anoikis, such as during cell migration or metastasis. The main receptors transmitting signals from the ECM are members of the integrin family. However, although integrin-mediated cell-ECM anchorage has been long recognized as crucial for epithelial cell survival, the in vivo significance of this interaction remains to be weighed. In this work, we have used the Drosophila wing imaginal disc epithelium to analyze the importance of integrins as survival factors during epithelia morphogenesis. We show that reducing integrin expression in the wing disc induces caspase-dependent cell death and basal extrusion of the dead cells. In this case, anoikis is mediated by the activation of the JNK pathway, which in turn triggers expression of the proapoptotic protein Hid. In addition, our results strongly suggest that, during wing disc morphogenesis, the EGFR pathway protects cells undergoing cell shape changes upon ECM detachment from anoikis. Furthermore, we show that oncogenic activation of the EGFR/Ras pathway in integrin mutant cells rescues them from apoptosis while promoting their extrusion from the epithelium. Altogether, our results support the idea that integrins promote cell survival during normal tissue morphogenesis and prevent the extrusion of transformed cells.
Collapse
Affiliation(s)
| | - M. Jesús Gómez-Lamarca
- Centro Andaluz de Biología del Desarrollo CSIC-Universidad Pablo de Olavide, Sevilla, Spain
- Departamento de Biología Celular, Universidad de Sevilla, Sevilla, Spain
| | | | - María D. Martín-Bermudo
- Centro Andaluz de Biología del Desarrollo CSIC-Universidad Pablo de Olavide, Sevilla, Spain
- *Correspondence: María D. Martín-Bermudo,
| |
Collapse
|
10
|
Kumar N, Huizar FJ, Farfán-Pira KJ, Brodskiy PA, Soundarrajan DK, Nahmad M, Zartman JJ. MAPPER: An Open-Source, High-Dimensional Image Analysis Pipeline Unmasks Differential Regulation of Drosophila Wing Features. Front Genet 2022; 13:869719. [PMID: 35480325 PMCID: PMC9035675 DOI: 10.3389/fgene.2022.869719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Phenomics requires quantification of large volumes of image data, necessitating high throughput image processing approaches. Existing image processing pipelines for Drosophila wings, a powerful genetic model for studying the underlying genetics for a broad range of cellular and developmental processes, are limited in speed, precision, and functional versatility. To expand on the utility of the wing as a phenotypic screening system, we developed MAPPER, an automated machine learning-based pipeline that quantifies high-dimensional phenotypic signatures, with each dimension quantifying a unique morphological feature of the Drosophila wing. MAPPER magnifies the power of Drosophila phenomics by rapidly quantifying subtle phenotypic differences in sample populations. We benchmarked MAPPER's accuracy and precision in replicating manual measurements to demonstrate its widespread utility. The morphological features extracted using MAPPER reveal variable sexual dimorphism across Drosophila species and unique underlying sex-specific differences in morphogen signaling in male and female wings. Moreover, the length of the proximal-distal axis across the species and sexes shows a conserved scaling relationship with respect to the wing size. In sum, MAPPER is an open-source tool for rapid, high-dimensional analysis of large imaging datasets. These high-content phenomic capabilities enable rigorous and systematic identification of genotype-to-phenotype relationships in a broad range of screening and drug testing applications and amplify the potential power of multimodal genomic approaches.
Collapse
Affiliation(s)
- Nilay Kumar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Francisco J. Huizar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Keity J. Farfán-Pira
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnical Institute (Cinvestav), Mexico City, Mexico
| | - Pavel A. Brodskiy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Dharsan K. Soundarrajan
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the National Polytechnical Institute (Cinvestav), Mexico City, Mexico
| | - Jeremiah J. Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
11
|
Antson H, Tõnissoo T, Shimmi O. The developing wing crossvein of Drosophila melanogaster: a fascinating model for signaling and morphogenesis. Fly (Austin) 2022; 16:118-127. [PMID: 35302430 PMCID: PMC8942417 DOI: 10.1080/19336934.2022.2040316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The Drosophila wing has been used as a model for studying tissue growth, morphogenesis and pattern formation. The wing veins of Drosophila are composed of two distinct structures, longitudinal veins and crossveins. Although positional information of longitudinal veins is largely defined in the wing imaginal disc during the larval stage, crossvein primordial cells appear to be naive until the early pupal stage. Here, we first review how wing crossveins have been investigated in the past. Then, the developmental mechanisms underlying crossvein formation are summarized. This review focuses on how a conserved trafficking mechanism of BMP ligands is utilized for crossvein formation, and how various co-factors play roles in sustaining BMP signalling. Recent findings further reveal that crossvein development serves as an excellent model to address how BMP signal and dynamic cellular processes are coupled. This comprehensive review illustrates the uniqueness, scientific value and future perspectives of wing crossvein development as a model.
Collapse
Affiliation(s)
- Hanna Antson
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Tambet Tõnissoo
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Osamu Shimmi
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia.,Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
12
|
Yang S, Wu X, Daoutidou EI, Zhang Y, Shimell M, Chuang KH, Peterson AJ, O'Connor MB, Zheng X. The NDNF-like factor Nord is a Hedgehog-induced extracellular BMP modulator that regulates Drosophila wing patterning and growth. eLife 2022; 11:e73357. [PMID: 35037619 PMCID: PMC8856659 DOI: 10.7554/elife.73357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/15/2022] [Indexed: 11/23/2022] Open
Abstract
Hedgehog (Hh) and Bone Morphogenetic Proteins (BMPs) pattern the developing Drosophila wing by functioning as short- and long-range morphogens, respectively. Here, we show that a previously unknown Hh-dependent mechanism fine-tunes the activity of BMPs. Through genome-wide expression profiling of the Drosophila wing imaginal discs, we identify nord as a novel target gene of the Hh signaling pathway. Nord is related to the vertebrate Neuron-Derived Neurotrophic Factor (NDNF) involved in congenital hypogonadotropic hypogonadism and several types of cancer. Loss- and gain-of-function analyses implicate Nord in the regulation of wing growth and proper crossvein patterning. At the molecular level, we present biochemical evidence that Nord is a secreted BMP-binding protein and localizes to the extracellular matrix. Nord binds to Decapentaplegic (Dpp) or the heterodimer Dpp-Glass-bottom boat (Gbb) to modulate their release and activity. Furthermore, we demonstrate that Nord is a dosage-dependent BMP modulator, where low levels of Nord promote and high levels inhibit BMP signaling. Taken together, we propose that Hh-induced Nord expression fine-tunes both the range and strength of BMP signaling in the developing Drosophila wing.
Collapse
Affiliation(s)
- Shu Yang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Xuefeng Wu
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Euphrosyne I Daoutidou
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Ya Zhang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - MaryJane Shimell
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Kun-Han Chuang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Aidan J Peterson
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Michael B O'Connor
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| |
Collapse
|
13
|
López-Varea A, Ostalé CM, Vega-Cuesta P, Ruiz-Gómez A, Organista MF, Martín M, Hevia CF, Molnar C, de Celis J, Culi J, Esteban N, de Celis JF. Genome-wide Phenotypic RNAi Screen in the Drosophila Wing: Global Parameters. G3-GENES GENOMES GENETICS 2021; 11:6380435. [PMID: 34599819 PMCID: PMC8962446 DOI: 10.1093/g3journal/jkab351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022]
Abstract
We have screened a collection of UAS-RNAi lines targeting 10,920 Drosophila protein-coding genes for phenotypes in the adult wing. We identified 3653 genes (33%) whose knockdown causes either larval/pupal lethality or a mutant phenotype affecting the formation of a normal wing. The most frequent phenotypes consist of changes in wing size, vein differentiation, and patterning, defects in the wing margin and in the apposition of the dorsal and ventral wing surfaces. We also defined 16 functional categories encompassing the most relevant aspect of each protein function and assigned each Drosophila gene to one of these functional groups. This allowed us to identify which mutant phenotypes are enriched within each functional group. Finally, we used previously published gene expression datasets to determine which genes are or are not expressed in the wing disc. Integrating expression, phenotypic and molecular information offers considerable precision to identify the relevant genes affecting wing formation and the biological processes regulated by them.
Collapse
Affiliation(s)
- Ana López-Varea
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Cristina M Ostalé
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Patricia Vega-Cuesta
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ana Ruiz-Gómez
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - María F Organista
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Mercedes Martín
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Covadonga F Hevia
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Cristina Molnar
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Jesús de Celis
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Joaquim Culi
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Nuria Esteban
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Jose F de Celis
- Centro de Biología Molecular "Severo Ochoa", CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
14
|
Chafino S, Martín D, Franch-Marro X. Activation of EGFR signaling by Tc-Vein and Tc-Spitz regulates the metamorphic transition in the red flour beetle Tribolium castaneum. Sci Rep 2021; 11:18807. [PMID: 34552169 PMCID: PMC8458297 DOI: 10.1038/s41598-021-98334-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023] Open
Abstract
Animal development relies on a sequence of specific stages that allow the formation of adult structures with a determined size. In general, juvenile stages are dedicated mainly to growth, whereas last stages are devoted predominantly to the maturation of adult structures. In holometabolous insects, metamorphosis marks the end of the growth period as the animals stops feeding and initiate the final differentiation of the tissues. This transition is controlled by the steroid hormone ecdysone produced in the prothoracic gland. In Drosophila melanogaster different signals have been shown to regulate the production of ecdysone, such as PTTH/Torso, TGFß and Egfr signaling. However, to which extent the roles of these signals are conserved remains unknown. Here, we study the role of Egfr signaling in post-embryonic development of the basal holometabolous beetle Tribolium castaneum. We show that Tc-Egfr and Tc-pointed are required to induced a proper larval-pupal transition through the control of the expression of ecdysone biosynthetic genes. Furthermore, we identified an additional Tc-Egfr ligand in the Tribolium genome, the neuregulin-like protein Tc-Vein (Tc-Vn), which contributes to induce larval-pupal transition together with Tc-Spitz (Tc-Spi). Interestingly, we found that in addition to the redundant role in the control of pupa formation, each ligand possesses different functions in organ morphogenesis. Whereas Tc-Spi acts as the main ligand in urogomphi and gin traps, Tc-Vn is required in wings and elytra. Altogether, our findings show that in Tribolium, post-embryonic Tc-Egfr signaling activation depends on the presence of two ligands and that its role in metamorphic transition is conserved in holometabolous insects.
Collapse
Affiliation(s)
- Sílvia Chafino
- grid.507636.10000 0004 0424 5398Institute of Evolutionary Biology (IBE, CSIC-Universitat Pompeu Fabra), Passeig de la Barceloneta 37, 08003 Barcelona, Catalonia Spain
| | - David Martín
- grid.507636.10000 0004 0424 5398Institute of Evolutionary Biology (IBE, CSIC-Universitat Pompeu Fabra), Passeig de la Barceloneta 37, 08003 Barcelona, Catalonia Spain
| | - Xavier Franch-Marro
- grid.507636.10000 0004 0424 5398Institute of Evolutionary Biology (IBE, CSIC-Universitat Pompeu Fabra), Passeig de la Barceloneta 37, 08003 Barcelona, Catalonia Spain
| |
Collapse
|
15
|
Atypical laminin spots and pull-generated microtubule-actin projections mediate Drosophila wing adhesion. Cell Rep 2021; 36:109667. [PMID: 34496252 DOI: 10.1016/j.celrep.2021.109667] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 06/11/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
During Drosophila metamorphosis, dorsal and ventral wing surfaces adhere, separate, and reappose in a paradoxical process involving cell-matrix adhesion, matrix production and degradation, and long cellular projections. The identity of the intervening matrix, the logic behind the adhesion-reapposition cycle, and the role of projections are unknown. We find that laminin matrix spots devoid of other main basement membrane components mediate wing adhesion. Through live imaging, we show that long microtubule-actin cables grow from those adhesion spots because of hydrostatic pressure that pushes wing surfaces apart. Formation of cables resistant to pressure requires spectraplakin, Patronin, septins, and Sdb, a SAXO1/2 microtubule stabilizer expressed under control of wing intervein-selector SRF. Silkworms and dead-leaf butterflies display similar dorso-ventral projections and expression of Sdb in intervein SRF-like patterns. Our study supports the morphogenetic importance of atypical basement-membrane-related matrices and dissects matrix-cytoskeleton coordination in a process of great evolutionary significance.
Collapse
|
16
|
Biodiversity-based development and evolution: the emerging research systems in model and non-model organisms. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1236-1280. [PMID: 33893979 DOI: 10.1007/s11427-020-1915-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Evolutionary developmental biology, or Evo-Devo for short, has become an established field that, broadly speaking, seeks to understand how changes in development drive major transitions and innovation in organismal evolution. It does so via integrating the principles and methods of many subdisciplines of biology. Although we have gained unprecedented knowledge from the studies on model organisms in the past decades, many fundamental and crucially essential processes remain a mystery. Considering the tremendous biodiversity of our planet, the current model organisms seem insufficient for us to understand the evolutionary and physiological processes of life and its adaptation to exterior environments. The currently increasing genomic data and the recently available gene-editing tools make it possible to extend our studies to non-model organisms. In this review, we review the recent work on the regulatory signaling of developmental and regeneration processes, environmental adaptation, and evolutionary mechanisms using both the existing model animals such as zebrafish and Drosophila, and the emerging nonstandard model organisms including amphioxus, ascidian, ciliates, single-celled phytoplankton, and marine nematode. In addition, the challenging questions and new directions in these systems are outlined as well.
Collapse
|
17
|
Different diets can affect attractiveness of Drosophila melanogaster males via changes in wing morphology. Anim Behav 2021. [DOI: 10.1016/j.anbehav.2020.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
18
|
Alba V, Carthew JE, Carthew RW, Mani M. Global constraints within the developmental program of the Drosophila wing. eLife 2021; 10:66750. [PMID: 34180394 PMCID: PMC8257256 DOI: 10.7554/elife.66750] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
Organismal development is a complex process, involving a vast number of molecular constituents interacting on multiple spatio-temporal scales in the formation of intricate body structures. Despite this complexity, development is remarkably reproducible and displays tolerance to both genetic and environmental perturbations. This robustness implies the existence of hidden simplicities in developmental programs. Here, using the Drosophila wing as a model system, we develop a new quantitative strategy that enables a robust description of biologically salient phenotypic variation. Analyzing natural phenotypic variation across a highly outbred population and variation generated by weak perturbations in genetic and environmental conditions, we observe a highly constrained set of wing phenotypes. Remarkably, the phenotypic variants can be described by a single integrated mode that corresponds to a non-intuitive combination of structural variations across the wing. This work demonstrates the presence of constraints that funnel environmental inputs and genetic variation into phenotypes stretched along a single axis in morphological space. Our results provide quantitative insights into the nature of robustness in complex forms while yet accommodating the potential for evolutionary variations. Methodologically, we introduce a general strategy for finding such invariances in other developmental contexts.
Collapse
Affiliation(s)
- Vasyl Alba
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States,NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
| | - James E Carthew
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States
| | - Richard W Carthew
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States,Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Madhav Mani
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States,NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States,Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| |
Collapse
|
19
|
Maier D. Membrane-Anchored Hairless Protein Restrains Notch Signaling Activity. Genes (Basel) 2020; 11:genes11111315. [PMID: 33171957 PMCID: PMC7694644 DOI: 10.3390/genes11111315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022] Open
Abstract
The Notch signaling pathway governs cell-to-cell communication in higher eukaryotes. In Drosophila, after cleavage of the transmembrane receptor Notch, the intracellular domain of Notch (ICN) binds to the transducer Suppressor of Hairless (Su(H)) and shuttles into the nucleus to activate Notch target genes. Similarly, the Notch antagonist Hairless transfers Su(H) into the nucleus to repress Notch target genes. With the aim to prevent Su(H) nuclear translocation, Hairless was fused to a transmembrane domain to anchor the protein at membranes. Indeed, endogenous Su(H) co-localized with membrane-anchored Hairless, demonstrating their binding in the cytoplasm. Moreover, adult phenotypes uncovered a loss of Notch activity, in support of membrane-anchored Hairless sequestering Su(H) in the cytosol. A combined overexpression of membrane-anchored Hairless with Su(H) lead to tissue proliferation, which is in contrast to the observed apoptosis after ectopic co-overexpression of the wild-type genes, indicating a shift to a gain of Notch activity. A mixed response, general de-repression of Notch signaling output, plus inhibition at places of highest Notch activity, perhaps reflects Su(H)’s role as activator and repressor, supported by results obtained with the Hairless-binding deficient Su(H)LLL mutant, inducing activation only. Overall, the results strengthen the idea of Su(H) and Hairless complex formation within the cytosolic compartment.
Collapse
Affiliation(s)
- Dieter Maier
- Deptartment of General Genetics 190g, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
20
|
Stapornwongkul KS, de Gennes M, Cocconi L, Salbreux G, Vincent JP. Patterning and growth control in vivo by an engineered GFP gradient. Science 2020; 370:321-327. [PMID: 33060356 PMCID: PMC7611032 DOI: 10.1126/science.abb8205] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Morphogen gradients provide positional information during development. To uncover the minimal requirements for morphogen gradient formation, we have engineered a synthetic morphogen in Drosophila wing primordia. We show that an inert protein, green fluorescent protein (GFP), can form a detectable diffusion-based gradient in the presence of surface-associated anti-GFP nanobodies, which modulate the gradient by trapping the ligand and limiting leakage from the tissue. We next fused anti-GFP nanobodies to the receptors of Dpp, a natural morphogen, to render them responsive to extracellular GFP. In the presence of these engineered receptors, GFP could replace Dpp to organize patterning and growth in vivo. Concomitant expression of glycosylphosphatidylinositol (GPI)-anchored nonsignaling receptors further improved patterning, to near-wild-type quality. Theoretical arguments suggest that GPI anchorage could be important for these receptors to expand the gradient length scale while at the same time reducing leakage.
Collapse
Affiliation(s)
| | - Marc de Gennes
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Luca Cocconi
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Imperial College, Department of Mathematics, London, UK
| | | | | |
Collapse
|
21
|
Nagel AC, Maier D, Scharpf J, Ketelhut M, Preiss A. Limited Availability of General Co-Repressors Uncovered in an Overexpression Context during Wing Venation in Drosophila melanogaster. Genes (Basel) 2020; 11:genes11101141. [PMID: 32998295 PMCID: PMC7601384 DOI: 10.3390/genes11101141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022] Open
Abstract
Cell fate is determined by the coordinated activity of different pathways, including the conserved Notch pathway. Activation of Notch results in the transcription of Notch targets that are otherwise silenced by repressor complexes. In Drosophila, the repressor complex comprises the transcription factor Suppressor of Hairless (Su(H)) bound to the Notch antagonist Hairless (H) and the general co-repressors Groucho (Gro) and C-terminal binding protein (CtBP). The latter two are shared by different repressors from numerous pathways, raising the possibility that they are rate-limiting. We noted that the overexpression during wing development of H mutants HdNT and HLD compromised in Su(H)-binding induced ectopic veins. On the basis of the role of H as Notch antagonist, overexpression of Su(H)-binding defective H isoforms should be without consequence, implying different mechanisms but repression of Notch signaling activity. Perhaps excess H protein curbs general co-repressor availability. Supporting this model, nearly normal wings developed upon overexpression of H mutant isoforms that bound neither Su(H) nor co-repressor Gro and CtBP. Excessive H protein appeared to sequester general co-repressors, resulting in specific vein defects, indicating their limited availability during wing vein development. In conclusion, interpretation of overexpression phenotypes requires careful consideration of possible dominant negative effects from interception of limiting factors.
Collapse
|
22
|
Wang Y, Wang J, Xia X, Wu G. Functional Identification of Px-fringe and Px-engrailed Genes under Heat Stress in Chlorpyrifos-Resistant and -Susceptible Plutela xylostella (Lepidoptera: Plutellidae). INSECTS 2020; 11:insects11050287. [PMID: 32392846 PMCID: PMC7290670 DOI: 10.3390/insects11050287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 11/16/2022]
Abstract
In our previous research, the fitness cost of resistance of the diamondback moth (DBM), Plutella xylostella found in insecticide-resistant DBM (Rc-DBM) under heat stress was based on heavier damage to wing veins when compared to insecticide-susceptible DBM (Sm-DBM). To investigate the molecular mechanism of the damage to the veins between Rc- and Sm-DBM, the full-length sequences of two related genes involved in the development of wing veins, fringe (Px-fng) and engrailed (Px-en) of DBM were cloned, and the mRNA expressions of both Px-fng and Px-en were studied. The Px-fng and Px-en cDNA contained 1038 bp and 1152 bp of open reading frames (ORFs), respectively, which encoded a putative protein comprising 345 and 383 amino acids with a calculated molecular weight of 39.59 kDa and 42.69 kDa. Significantly down regulated expressions of Px-fng and Px-en under heat stress were found in pupae and adults of Rc-DBM compared to Sm-DBM, and a result of higher damage to wing veins in Rc-DBM under heat stress. Based on RNAi experiments, significant inhibitions on expressions of Px-fng and Px-en in both Sm-DBM and Rc-DBM were found when the pupae were infected by dsFng or dsEn. Corresponding to these, infections of dsFng or dsEn resulted in significant decrease of eclosion rate and increase malformation rate of DBM. Our results suggest that the higher damage of wing veins in DBM might be related to the heavier inhibitions of Px-fng and Px-en expression, and the Px-fng and Px-en are involved in the development of wings and veins.
Collapse
Affiliation(s)
- Yu Wang
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jingnan Wang
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaofeng Xia
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Correspondence: (X.X.); (G.W.)
| | - Gang Wu
- Key Laboratory of Biopesticide and Chemical Biology (Ministry of Education), Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.W.); (J.W.)
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Correspondence: (X.X.); (G.W.)
| |
Collapse
|
23
|
Li X, Liu M, Ren X, Loncle N, Wang Q, Hemba-Waduge RUS, Yu SH, Boube M, Bourbon HMG, Ni JQ, Ji JY. The Mediator CDK8-Cyclin C complex modulates Dpp signaling in Drosophila by stimulating Mad-dependent transcription. PLoS Genet 2020; 16:e1008832. [PMID: 32463833 PMCID: PMC7282676 DOI: 10.1371/journal.pgen.1008832] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/09/2020] [Accepted: 05/05/2020] [Indexed: 11/19/2022] Open
Abstract
Dysregulation of CDK8 (Cyclin-Dependent Kinase 8) and its regulatory partner CycC (Cyclin C), two subunits of the conserved Mediator (MED) complex, have been linked to diverse human diseases such as cancer. Thus, it is essential to understand the regulatory network modulating the CDK8-CycC complex in both normal development and tumorigenesis. To identify upstream regulators or downstream effectors of CDK8, we performed a dominant modifier genetic screen in Drosophila based on the defects in vein patterning caused by specific depletion or overexpression of CDK8 or CycC in developing wing imaginal discs. We identified 26 genomic loci whose haploinsufficiency can modify these CDK8- or CycC-specific phenotypes. Further analysis of two overlapping deficiency lines and mutant alleles led us to identify genetic interactions between the CDK8-CycC pair and the components of the Decapentaplegic (Dpp, the Drosophila homolog of TGFβ, or Transforming Growth Factor-β) signaling pathway. We observed that CDK8-CycC positively regulates transcription activated by Mad (Mothers against dpp), the primary transcription factor downstream of the Dpp/TGFβ signaling pathway. CDK8 can directly interact with Mad in vitro through the linker region between the DNA-binding MH1 (Mad homology 1) domain and the carboxy terminal MH2 (Mad homology 2) transactivation domain. Besides CDK8 and CycC, further analyses of other subunits of the MED complex have revealed six additional subunits that are required for Mad-dependent transcription in the wing discs: Med12, Med13, Med15, Med23, Med24, and Med31. Furthermore, our analyses confirmed the positive roles of CDK9 and Yorkie in regulating Mad-dependent gene expression in vivo. These results suggest that CDK8 and CycC, together with a few other subunits of the MED complex, may coordinate with other transcription cofactors in regulating Mad-dependent transcription during wing development in Drosophila.
Collapse
Affiliation(s)
- Xiao Li
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Mengmeng Liu
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Xingjie Ren
- School of Medicine, Tsinghua University, Beijing, China
| | - Nicolas Loncle
- Centre de Biologie Intégrative, Centre de Biologie du Développement, UMR5544 du CNRS, Université de Toulouse, Toulouse, France
| | - Qun Wang
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Rajitha-Udakara-Sampath Hemba-Waduge
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Stephen H. Yu
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
| | - Muriel Boube
- Centre de Biologie Intégrative, Centre de Biologie du Développement, UMR5544 du CNRS, Université de Toulouse, Toulouse, France
| | - Henri-Marc G. Bourbon
- Centre de Biologie Intégrative, Centre de Biologie du Développement, UMR5544 du CNRS, Université de Toulouse, Toulouse, France
| | - Jian-Quan Ni
- School of Medicine, Tsinghua University, Beijing, China
| | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, United States of America
- Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
24
|
Kushnir T, Bar-Cohen S, Mooshayef N, Lange R, Bar-Sinai A, Rozen H, Salzberg A, Engelberg D, Paroush Z. An Activating Mutation in ERK Causes Hyperplastic Tumors in a scribble Mutant Tissue in Drosophila. Genetics 2020; 214:109-120. [PMID: 31740452 PMCID: PMC6944410 DOI: 10.1534/genetics.119.302794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
Receptor tyrosine kinase signaling plays prominent roles in tumorigenesis, and activating oncogenic point mutations in the core pathway components Ras, Raf, or MEK are prevalent in many types of cancer. Intriguingly, however, analogous oncogenic mutations in the downstream effector kinase ERK have not been described or validated in vivo To determine if a point mutation could render ERK intrinsically active and oncogenic, we have assayed in Drosophila the effects of a mutation that confers constitutive activity upon a yeast ERK ortholog and has also been identified in a few human tumors. Our analyses indicate that a fly ERK ortholog harboring this mutation alone (RolledR80S), and more so in conjunction with the known sevenmaker mutation (RolledR80S+D334N), suppresses multiple phenotypes caused by loss of Ras-Raf-MEK pathway activity, consistent with an intrinsic activity that is independent of upstream signaling. Moreover, expression of RolledR80S and RolledR80S+D334N induces tissue overgrowth in an established Drosophila cancer model. Our findings thus demonstrate that activating mutations can bestow ERK with pro-proliferative, tumorigenic capabilities and suggest that Drosophila represents an effective experimental system for determining the oncogenicity of ERK mutants and their response to therapy.
Collapse
Affiliation(s)
- Tatyana Kushnir
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Shaked Bar-Cohen
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Navit Mooshayef
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Singapore-Hebrew University of Jerusalem Alliance for Research and Enterprise, Molecular Mechanisms of Inflammatory Diseases Interdisciplinary Research Group, Campus for Research Excellence and Technological Enterprise, 138602, Singapore
| | - Rotem Lange
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Allan Bar-Sinai
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Helit Rozen
- Faculty of Medicine in the Galilee, Bar-Ilan University, Safed 1311502, 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
| | - David Engelberg
- Department of Biological Chemistry, Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Singapore-Hebrew University of Jerusalem Alliance for Research and Enterprise, Molecular Mechanisms of Inflammatory Diseases Interdisciplinary Research Group, Campus for Research Excellence and Technological Enterprise, 138602, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Ze'ev Paroush
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
25
|
Banerjee TD, Monteiro A. Molecular mechanisms underlying simplification of venation patterns in holometabolous insects. Development 2020; 147:dev.196394. [DOI: 10.1242/dev.196394] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/21/2020] [Indexed: 01/07/2023]
Abstract
How mechanisms of pattern formation evolve has remained a central research theme in the field of evolutionary and developmental biology. The mechanism of wing vein differentiation in Drosophila is a classic text-book example of pattern formation using a system of positional-information, yet very little is known about how species with a different number of veins pattern their wings, and how insect venation patterns evolved. Here, we examine the expression pattern of genes previously implicated in vein differentiation in Drosophila in two butterfly species with more complex venation Bicyclus anynana and Pieris canidia. We also test the function of some of these genes in B. anynana. We identify both conserved as well as new domains of decapentaplegic, engrailed, invected, spalt, optix, wingless, armadillo, blistered, and rhomboid gene expression in butterflies, and propose how the simplified venation in Drosophila might have evolved via loss of decapentaplegic, spalt and optix gene expression domains, silencing of vein inducing programs at Spalt-expression boundaries, and changes in gene expression of vein maintenance genes.
Collapse
Affiliation(s)
- Tirtha Das Banerjee
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, Singapore
- Yale-NUS College, Singapore
| |
Collapse
|
26
|
Mahmud AKMF, Yang D, Stenberg P, Ioshikhes I, Nandi S. Exploring a Drosophila Transcription Factor Interaction Network to Identify Cis-Regulatory Modules. J Comput Biol 2019; 27:1313-1328. [PMID: 31855461 DOI: 10.1089/cmb.2018.0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiple transcription factors (TFs) bind to specific sites in the genome and interact among themselves to form the cis-regulatory modules (CRMs). They are essential in modulating the expression of genes, and it is important to study this interplay to understand gene regulation. In the present study, we integrated experimentally identified TF binding sites collected from published studies with computationally predicted TF binding sites to identify Drosophila CRMs. Along with the detection of the previously known CRMs, this approach identified novel protein combinations. We determined high-occupancy target sites, where a large number of TFs bind. Investigating these sites revealed that Giant, Dichaete, and Knirp are highly enriched in these locations. A common TAG team motif was observed at these sites, which might play a role in recruiting other TFs. While comparing the binding sites at distal and proximal promoters, we found that certain regulatory TFs, such as Zelda, were highly enriched in enhancers. Our study has shown that, from the information available concerning the TF binding sites, the real CRMs could be predicted accurately and efficiently. Although we only may claim co-occurrence of these proteins in this study, it may actually point to their interaction (as known interaction proteins typically co-occur together). Such an integrative approach can, therefore, help us to provide a better understanding of the interplay among the factors, even though further experimental verification is required.
Collapse
Affiliation(s)
| | - Doo Yang
- Ottawa Institute of Computational Biology and Bioinformatics (OICBB) and Ottawa Institute of Systems Biology (OISB) and Department of Biochemistry, Microbiology and Immunology (BMI), Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Per Stenberg
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Ilya Ioshikhes
- Ottawa Institute of Computational Biology and Bioinformatics (OICBB) and Ottawa Institute of Systems Biology (OISB) and Department of Biochemistry, Microbiology and Immunology (BMI), Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Soumyadeep Nandi
- Life Sciences Division, Institute of Advanced Study in Science and Technology, Vigyan Path, Paschim Boragaon, Guwahati, India; Amity University Haryana, Gurugram, India
| |
Collapse
|
27
|
Pascoal S, Risse JE, Zhang X, Blaxter M, Cezard T, Challis RJ, Gharbi K, Hunt J, Kumar S, Langan E, Liu X, Rayner JG, Ritchie MG, Snoek BL, Trivedi U, Bailey NW. Field cricket genome reveals the footprint of recent, abrupt adaptation in the wild. Evol Lett 2019; 4:19-33. [PMID: 32055408 PMCID: PMC7006468 DOI: 10.1002/evl3.148] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/21/2019] [Accepted: 11/13/2019] [Indexed: 12/31/2022] Open
Abstract
Evolutionary adaptation is generally thought to occur through incremental mutational steps, but large mutational leaps can occur during its early stages. These are challenging to study in nature due to the difficulty of observing new genetic variants as they arise and spread, but characterizing their genomic dynamics is important for understanding factors favoring rapid adaptation. Here, we report genomic consequences of recent, adaptive song loss in a Hawaiian population of field crickets (Teleogryllus oceanicus). A discrete genetic variant, flatwing, appeared and spread approximately 15 years ago. Flatwing erases sound‐producing veins on male wings. These silent flatwing males are protected from a lethal, eavesdropping parasitoid fly. We sequenced, assembled and annotated the cricket genome, produced a linkage map, and identified a flatwing quantitative trait locus covering a large region of the X chromosome. Gene expression profiling showed that flatwing is associated with extensive genome‐wide effects on embryonic gene expression. We found that flatwing male crickets express feminized chemical pheromones. This male feminizing effect, on a different sexual signaling modality, is genetically associated with the flatwing genotype. Our findings suggest that the early stages of evolutionary adaptation to extreme pressures can be accompanied by greater genomic and phenotypic disruption than previously appreciated, and highlight how abrupt adaptation might involve suites of traits that arise through pleiotropy or genomic hitchhiking.
Collapse
Affiliation(s)
- Sonia Pascoal
- Department of Zoology University of Cambridge Cambridge CB2 3EJ United Kingdom
| | - Judith E Risse
- Division of Bioinformatics, Department of Plant Sciences Wageningen University & Research Wageningen 6708 PB The Netherlands.,Animal Ecology Netherlands Institute of Ecology Wageningen 6700 AB The Netherlands
| | - Xiao Zhang
- School of Biology University of St Andrews St Andrews Fife KY16 9TH United Kingdom
| | - Mark Blaxter
- Edinburgh Genomics University of Edinburgh Edinburgh EH9 3JT United Kingdom.,Institute of Evolutionary Biology University of Edinburgh Edinburgh EH9 3JT United Kingdom
| | - Timothee Cezard
- Edinburgh Genomics University of Edinburgh Edinburgh EH9 3JT United Kingdom
| | - Richard J Challis
- Edinburgh Genomics University of Edinburgh Edinburgh EH9 3JT United Kingdom
| | - Karim Gharbi
- Edinburgh Genomics University of Edinburgh Edinburgh EH9 3JT United Kingdom.,Earlham Institute Norwich Research Park Norwich NR4 7UZ United Kingdom
| | - John Hunt
- School of Science and Health and the Hawkesbury Institute for the Environment Western Sydney University Penrith NSW 2751 Australia.,Centre for Ecology and Conservation University of Exeter Penryn TR10 9FE United Kingdom
| | - Sujai Kumar
- Edinburgh Genomics University of Edinburgh Edinburgh EH9 3JT United Kingdom
| | - Emma Langan
- Edinburgh Genomics University of Edinburgh Edinburgh EH9 3JT United Kingdom.,School of Environmental Sciences University of East Anglia Norwich NR4 7UZ United Kingdom
| | - Xuan Liu
- Centre for Genomic Research University of Liverpool Liverpool L69 7ZB United Kingdom
| | - Jack G Rayner
- School of Biology University of St Andrews St Andrews Fife KY16 9TH United Kingdom
| | - Michael G Ritchie
- School of Biology University of St Andrews St Andrews Fife KY16 9TH United Kingdom
| | - Basten L Snoek
- Theoretical Biology and Bioinformatics Utrecht University Utrecht 3584 CH The Netherlands.,Terrestrial Ecology Netherlands Institute of Ecology Wageningen 6700 AB The Netherlands
| | - Urmi Trivedi
- Edinburgh Genomics University of Edinburgh Edinburgh EH9 3JT United Kingdom
| | - Nathan W Bailey
- School of Biology University of St Andrews St Andrews Fife KY16 9TH United Kingdom
| |
Collapse
|
28
|
McCulloch GA, Oliphant A, Dearden PK, Veale AJ, Ellen CW, Waters JM. Comparative transcriptomic analysis of a wing-dimorphic stonefly reveals candidate wing loss genes. EvoDevo 2019; 10:21. [PMID: 31516688 PMCID: PMC6728979 DOI: 10.1186/s13227-019-0135-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/24/2019] [Indexed: 01/28/2023] Open
Abstract
Background The genetic basis of wing development has been well characterised for model insect species, but remains poorly understood in phylogenetically divergent, non-model taxa. Wing-polymorphic insect species potentially provide ideal systems for unravelling the genetic basis of secondary wing reduction. Stoneflies (Plecoptera) represent an anciently derived insect assemblage for which the genetic basis of wing polymorphism remains unclear. We undertake quantitative RNA-seq of sympatric full-winged versus vestigial-winged nymphs of a widespread wing-dimorphic New Zealand stonefly, Zelandoperla fenestrata, to identify genes potentially involved in wing development and secondary wing loss. Results Our analysis reveals substantial differential expression of wing-development genes between full-winged versus vestigial-winged stonefly ecotypes. Specifically, of 23 clusters showing significant similarity to Drosophila wing development-related genes and their pea aphid orthologues, nine were significantly upregulated in full-winged stonefly ecotypes, whereas only one cluster (teashirt) was substantially upregulated in the vestigial-winged ecotype. Conclusions These findings suggest remarkable conservation of key wing-development pathways throughout 400 Ma of insect evolution. The finding that two Juvenile Hormone pathway clusters were significantly upregulated in vestigial-winged Zelandoperla supports the hypothesis that Juvenile Hormone may play a key role in modulating insect wing polymorphism, as has previously been suggested for other insect lineages.
Collapse
Affiliation(s)
- Graham A McCulloch
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Andrew Oliphant
- 2Genomics Aotearoa and Department of Biochemistry, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Peter K Dearden
- 2Genomics Aotearoa and Department of Biochemistry, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Andrew J Veale
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand.,3Landcare Research, Private Bag 92170, Auckland Mail Centre, Auckland, 1142 New Zealand
| | - Charles W Ellen
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| | - Jonathan M Waters
- 1Department of Zoology, University of Otago, P.O. Box 56, Dunedin, 9054 New Zealand
| |
Collapse
|
29
|
Barik BK, Mishra M. Nanoparticles as a potential teratogen: a lesson learnt from fruit fly. Nanotoxicology 2018; 13:258-284. [DOI: 10.1080/17435390.2018.1530393] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Bedanta Kumar Barik
- Neural Developmental Biology Lab, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Monalisa Mishra
- Neural Developmental Biology Lab, Department of Life Science, National Institute of Technology, Rourkela, India
| |
Collapse
|
30
|
Belus MT, Rogers MA, Elzubeir A, Josey M, Rose S, Andreeva V, Yelick PC, Bates EA. Kir2.1 is important for efficient BMP signaling in mammalian face development. Dev Biol 2018; 444 Suppl 1:S297-S307. [PMID: 29571612 PMCID: PMC6148416 DOI: 10.1016/j.ydbio.2018.02.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 12/23/2022]
Abstract
Mutations that disrupt the inwardly rectifying potassium channel Kir2.1 lead to Andersen-Tawil syndrome that includes periodic paralysis, cardiac arrhythmia, cognitive deficits, craniofacial dysmorphologies and limb defects. The molecular mechanism that underlies the developmental consequences of inhibition of these channels has remained a mystery. We show that while loss of Kir2.1 function does not affect expression of several early facial patterning genes, the domain in which Pou3f3 is expressed in the maxillary arch is reduced. Pou3f3 is important for development of the jugal and squamosal bones. The reduced expression domain of Pou3f3 is consistent with the reduction in the size of the squamosal and jugal bones in Kcnj2KO/KO animals, however it does not account for the diverse craniofacial defects observed in Kcnj2KO/KO animals. We show that Kir2.1 function is required in the cranial neural crest for morphogenesis of several craniofacial structures including palate closure. We find that while the palatal shelves of Kir2.1-null embryos elevate properly, they are reduced in size due to decreased proliferation of the palatal mesenchyme. While we find no reduction in expression of BMP ligands, receptors, and associated Smads in this setting, loss of Kir2.1 reduces the efficacy of BMP signaling as shown by the reduction of phosphorylated Smad 1/5/8 and reduced expression of BMP targets Smad6 and Satb2.
Collapse
Affiliation(s)
- Matthew T Belus
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Madison A Rogers
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Alaaeddin Elzubeir
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Megan Josey
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Steven Rose
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Viktoria Andreeva
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University, Boston, MA 02111, United States
| | - Pamela C Yelick
- Department of Orthodontics, Division of Craniofacial and Molecular Genetics, Tufts University, Boston, MA 02111, United States
| | - Emily A Bates
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States.
| |
Collapse
|
31
|
Ando T, Matsuda T, Goto K, Hara K, Ito A, Hirata J, Yatomi J, Kajitani R, Okuno M, Yamaguchi K, Kobayashi M, Takano T, Minakuchi Y, Seki M, Suzuki Y, Yano K, Itoh T, Shigenobu S, Toyoda A, Niimi T. Repeated inversions within a pannier intron drive diversification of intraspecific colour patterns of ladybird beetles. Nat Commun 2018; 9:3843. [PMID: 30242156 PMCID: PMC6155092 DOI: 10.1038/s41467-018-06116-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 08/15/2018] [Indexed: 11/16/2022] Open
Abstract
How genetic information is modified to generate phenotypic variation within a species is one of the central questions in evolutionary biology. Here we focus on the striking intraspecific diversity of >200 aposematic elytral (forewing) colour patterns of the multicoloured Asian ladybird beetle, Harmonia axyridis, which is regulated by a tightly linked genetic locus h. Our loss-of-function analyses, genetic association studies, de novo genome assemblies, and gene expression data reveal that the GATA transcription factor gene pannier is the major regulatory gene located at the h locus, and suggest that repeated inversions and cis-regulatory modifications at pannier led to the expansion of colour pattern variation in H. axyridis. Moreover, we show that the colour-patterning function of pannier is conserved in the seven-spotted ladybird beetle, Coccinella septempunctata, suggesting that H. axyridis’ extraordinary intraspecific variation may have arisen from ancient modifications in conserved elytral colour-patterning mechanisms in ladybird beetles. The harlequin ladybird beetle, Harmonia axyridis, has remarkable phenotypic diversity, with over 200 colour patterns. Here, Ando et al. show that this patterning is regulated by the transcription factor gene pannier and has diversified by repeated inversions and cis-regulatory modifications of pannier.
Collapse
Affiliation(s)
- Toshiya Ando
- Division of Evolutionary Developmental Biology, National Institute for Basic Biology, Okazaki, Aichi, 444-8585, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, 444-8585, Japan
| | - Takeshi Matsuda
- Laboratory of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Kumiko Goto
- Laboratory of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Kimiko Hara
- Laboratory of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Akinori Ito
- Laboratory of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Junya Hirata
- Laboratory of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Joichiro Yatomi
- Laboratory of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Rei Kajitani
- Department of Biological Information, Tokyo Institute of Technology, Meguro-ku, Tokyo, 152-8550, Japan
| | - Miki Okuno
- Department of Biological Information, Tokyo Institute of Technology, Meguro-ku, Tokyo, 152-8550, Japan
| | - Katsushi Yamaguchi
- NIBB Core Research Facilities, National Institute for Basic Biology, Okazaki, Aichi, 444-8585, Japan
| | - Masaaki Kobayashi
- Bioinformatics Laboratory, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan
| | - Tomoyuki Takano
- Bioinformatics Laboratory, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan
| | - Yohei Minakuchi
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Masahide Seki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8562, Japan
| | - Yutaka Suzuki
- Laboratory of Systems Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8562, Japan
| | - Kentaro Yano
- Bioinformatics Laboratory, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, Kanagawa, 214-8571, Japan
| | - Takehiko Itoh
- Department of Biological Information, Tokyo Institute of Technology, Meguro-ku, Tokyo, 152-8550, Japan
| | - Shuji Shigenobu
- Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, 444-8585, Japan.,NIBB Core Research Facilities, National Institute for Basic Biology, Okazaki, Aichi, 444-8585, Japan
| | - Atsushi Toyoda
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan.,Advanced Genomics Center, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Teruyuki Niimi
- Division of Evolutionary Developmental Biology, National Institute for Basic Biology, Okazaki, Aichi, 444-8585, Japan. .,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi, 444-8585, Japan. .,Laboratory of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan.
| |
Collapse
|
32
|
Ruiz-Losada M, Blom-Dahl D, Córdoba S, Estella C. Specification and Patterning of Drosophila Appendages. J Dev Biol 2018; 6:jdb6030017. [PMID: 30011921 PMCID: PMC6162442 DOI: 10.3390/jdb6030017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/10/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023] Open
Abstract
Appendages are external projections of the body that serve the animal for locomotion, feeding, or environment exploration. The appendages of the fruit fly Drosophilamelanogaster are derived from the imaginal discs, epithelial sac-like structures specified in the embryo that grow and pattern during larva development. In the last decades, genetic and developmental studies in the fruit fly have provided extensive knowledge regarding the mechanisms that direct the formation of the appendages. Importantly, many of the signaling pathways and patterning genes identified and characterized in Drosophila have similar functions during vertebrate appendage development. In this review, we will summarize the genetic and molecular mechanisms that lead to the specification of appendage primordia in the embryo and their posterior patterning during imaginal disc development. The identification of the regulatory logic underlying appendage specification in Drosophila suggests that the evolutionary origin of the insect wing is, in part, related to the development of ventral appendages.
Collapse
Affiliation(s)
- Mireya Ruiz-Losada
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - David Blom-Dahl
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - Sergio Córdoba
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| | - Carlos Estella
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM/CSIC), Nicolás Cabrera 1, 28049 Madrid, Spain.
| |
Collapse
|
33
|
Thuma L, Carter D, Weavers H, Martin P. Drosophila immune cells extravasate from vessels to wounds using Tre1 GPCR and Rho signaling. J Cell Biol 2018; 217:3045-3056. [PMID: 29941473 PMCID: PMC6122984 DOI: 10.1083/jcb.201801013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/04/2018] [Accepted: 05/29/2018] [Indexed: 12/17/2022] Open
Abstract
In contrast to vertebrates, adult Drosophila melanogaster have an open cardiovascular system. However, Thuma et al. find that in late pupation, hemolymph flows through Drosophila wing veins, providing a unique genetic and live-imaging opportunity to investigate the mechanisms driving immune cell extravasation from vessels to wounds and reveal new roles for Tre1 and Rho signaling in this process. Inflammation is pivotal to fight infection, clear debris, and orchestrate repair of injured tissues. Although Drosophila melanogaster have proven invaluable for studying extravascular recruitment of innate immune cells (hemocytes) to wounds, they have been somewhat neglected as viable models to investigate a key rate-limiting component of inflammation—that of immune cell extravasation across vessel walls—due to their open circulation. We have now identified a period during pupal development when wing hearts pulse hemolymph, including circulating hemocytes, through developing wing veins. Wounding near these vessels triggers local immune cell extravasation, enabling live imaging and correlative light-electron microscopy of these events in vivo. We show that RNAi knockdown of immune cell integrin blocks diapedesis, just as in vertebrates, and we uncover a novel role for Rho-like signaling through the GPCR Tre1, a gene previously implicated in the trans-epithelial migration of germ cells. We believe this new Drosophila model complements current murine models and provides new mechanistic insight into immune cell extravasation.
Collapse
Affiliation(s)
- Leila Thuma
- Department of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK
| | - Deborah Carter
- Department of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK
| | - Helen Weavers
- School of Cellular and Molecular Medicine, Biomedical Sciences, University of Bristol, Bristol, UK .,School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, UK
| | - Paul Martin
- Department of Physiology, Pharmacology and Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK .,School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, UK.,School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
34
|
Ahmed-de-Prado S, Diaz-Garcia S, Baonza A. JNK and JAK/STAT signalling are required for inducing loss of cell fate specification during imaginal wing discs regeneration in Drosophila melanogaster. Dev Biol 2018; 441:31-41. [PMID: 29870691 DOI: 10.1016/j.ydbio.2018.05.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 05/21/2018] [Accepted: 05/24/2018] [Indexed: 11/25/2022]
Abstract
The regenerative process after tissue damage relies on a variety of cellular responses that includes compensatory cell proliferation and cell fate re-specification. The identification of the signalling networks regulating these cellular events is a central question in regenerative biology. Tissue regeneration models in Drosophila have shown that two of the signals that play a fundamental role during the early stages of regeneration are the c-Jun N-terminal kinase (JNK) and JAK/STAT signalling pathways. These pathways have been shown to be required for controlling regenerative proliferation, however their contribution to the processes of cellular reprogramming and cell fate re-specification that take place during regeneration are largely unknown. Here, we present evidence for a previously unrecognised function of the cooperative activities of JNK and JAK/STAT signalling pathways in inducing loss of cell fate specification in imaginal discs. We show that co-activation of these signalling pathways induces both the cell fate changes in injured areas, as well as in adjacent cells. We have also found that this function relies on the activity of the Caspase initiator encoded by the gene dronc.
Collapse
Affiliation(s)
- Sara Ahmed-de-Prado
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, C/Nicolás Cabrera 1, Madrid 28049, Spain
| | - Sandra Diaz-Garcia
- University of California, San Diego Section of Cell&Developmental Biology, La Jolla, CA 92093-0349, USA
| | - Antonio Baonza
- Centro de Biología Molecular "Severo Ochoa", CSIC/UAM, C/Nicolás Cabrera 1, Madrid 28049, Spain.
| |
Collapse
|
35
|
Preiss A, Nagel AC, Praxenthaler H, Maier D. Complex genetic interactions of novel Suppressor of Hairless alleles deficient in co-repressor binding. PLoS One 2018; 13:e0193956. [PMID: 29509808 PMCID: PMC5839567 DOI: 10.1371/journal.pone.0193956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/21/2018] [Indexed: 11/18/2022] Open
Abstract
Throughout the animal kingdom, the Notch signalling pathway allows cells to acquire diversified cell fates. Notch signals are translated into activation of Notch target genes by CSL transcription factors. In the absence of Notch signals, CSL together with co-repressors functions as a transcriptional repressor. In Drosophila, repression of Notch target genes involves the CSL homologue Suppressor of Hairless (Su(H)) and the Notch (N) antagonist Hairless (H) that together form a repressor complex. Guided by crystal structure, three mutations Su(H)LL, Su(H)LLF and Su(H)LLL were generated that specifically affect interactions with the repressor H, and were introduced into the endogenous Su(H) locus by gene engineering. In contrast to the wild type isoform, these Su(H) mutants are incapable of repressor complex formation. Accordingly, Notch signalling activity is dramatically elevated in the homozygotes, resembling complete absence of H activity. It was noted, however, that heterozygotes do not display a dominant H loss of function phenotype. In this work we addressed genetic interactions the three H-binding deficient Su(H) mutants display in combination with H and N null alleles. We included a null mutant of Delta (Dl), encoding the ligand of the Notch receptor, as well as of Su(H) itself in our genetic analyses. H, N or Dl mutations cause dominant wing phenotypes that are sensitive to gene dose of the others. Moreover, H heterozygotes lack bristle organs and develop bristle sockets instead of shafts. The latter phenotype is suppressed by Su(H) null alleles but not by H-binding deficient Su(H) alleles which we attribute to the socket cell specific activity of Su(H). Modification of the dominant wing phenotypes of either H, N or Dl, however, suggested some lack of repressor activity in the Su(H) null allele and likewise in the H-binding deficient Su(H) alleles. Overall, Su(H) mutants are recessive perhaps reflecting self-adjusting availability of Su(H) protein.
Collapse
Affiliation(s)
- Anette Preiss
- Institute of Genetics (240), University of Hohenheim, Stuttgart, Germany
| | - Anja C. Nagel
- Institute of Genetics (240), University of Hohenheim, Stuttgart, Germany
| | - Heiko Praxenthaler
- Institute of Genetics (240), University of Hohenheim, Stuttgart, Germany
| | - Dieter Maier
- Institute of Genetics (240), University of Hohenheim, Stuttgart, Germany
- * E-mail:
| |
Collapse
|
36
|
Guo S, Zhao Z, Liu L, Li Z, Shen J. Comparative Transcriptome Analyses Uncover Key Candidate Genes Mediating Flight Capacity in Bactrocera dorsalis (Hendel) and Bactrocera correcta (Bezzi) (Diptera: Tephritidae). Int J Mol Sci 2018; 19:E396. [PMID: 29385681 PMCID: PMC5855618 DOI: 10.3390/ijms19020396] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/08/2018] [Accepted: 01/22/2018] [Indexed: 11/16/2022] Open
Abstract
Flight capacity is important for invasive pests during entry, establishment and spreading. Both Bactroceradorsalis Hendel and Bactroceracorrecta Bezzi are invasive fruit flies but their flight capacities differ. Here, a tethered flight mill test demonstrated that B. dorsalis exhibits a greater flight capacity than B. correcta. RNA-Seq was used to determine the transcriptomic differences associated with the flight capacity of two Bactrocera species. Transcriptome data showed that 6392 unigenes were differentially expressed between the two species in the larval stage, whereas in the adult stage, 4104 differentially expressed genes (DEGs) were identified in females, and 3445 DEGs were observed in males. The flight capacity appeared to be correlated with changes in the transcriptional levels of genes involved in wing formation, flight muscle structure, energy metabolism, and hormonal control. Using RNA interference (RNAi) to verify the function of one DEG, the epidermal growth factor receptor (EGFR), we confirmed the role of this gene in regulating wing development, and thereby flight capacity, in both species. This work reveals the flight mechanism of fruit flies and provides insight into fundamental transcriptomics for further studies on the flight performance of insects.
Collapse
Affiliation(s)
- Shaokun Guo
- Key Laboratory of Ministry of Agriculture for Monitoring and Green Management of Crop Pests, Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Zihua Zhao
- Key Laboratory of Ministry of Agriculture for Monitoring and Green Management of Crop Pests, Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Lijun Liu
- Key Laboratory of Ministry of Agriculture for Monitoring and Green Management of Crop Pests, Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Zhihong Li
- Key Laboratory of Ministry of Agriculture for Monitoring and Green Management of Crop Pests, Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| | - Jie Shen
- Key Laboratory of Ministry of Agriculture for Monitoring and Green Management of Crop Pests, Department of Entomology, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
37
|
Stewart DP, Marada S, Bodeen WJ, Truong A, Sakurada SM, Pandit T, Pruett-Miller SM, Ogden SK. Cleavage activates dispatched for Sonic Hedgehog ligand release. eLife 2018; 7:31678. [PMID: 29359685 PMCID: PMC5811216 DOI: 10.7554/elife.31678] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/22/2018] [Indexed: 12/12/2022] Open
Abstract
Hedgehog ligands activate an evolutionarily conserved signaling pathway that provides instructional cues during tissue morphogenesis, and when corrupted, contributes to developmental disorders and cancer. The transmembrane protein Dispatched is an essential component of the machinery that deploys Hedgehog family ligands from producing cells, and is absolutely required for signaling to long-range targets. Despite this crucial role, regulatory mechanisms controlling Dispatched activity remain largely undefined. Herein, we reveal vertebrate Dispatched is activated by proprotein convertase-mediated cleavage at a conserved processing site in its first extracellular loop. Dispatched processing occurs at the cell surface to instruct its membrane re-localization in polarized epithelial cells. Cleavage site mutation alters Dispatched membrane trafficking and reduces ligand release, leading to compromised pathway activity in vivo. As such, convertase-mediated cleavage is required for Dispatched maturation and functional competency in Hedgehog ligand-producing cells.
Collapse
Affiliation(s)
- Daniel P Stewart
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Suresh Marada
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - William J Bodeen
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Integrated Program in Biomedical Sciences, University of Tennessee Health Sciences Center, Memphis, United States
| | - Ashley Truong
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Sadie Miki Sakurada
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - Tanushree Pandit
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, United States
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, United States
| |
Collapse
|
38
|
Calpena E, López Del Amo V, Chakraborty M, Llamusí B, Artero R, Espinós C, Galindo MI. The Drosophila junctophilin gene is functionally equivalent to its four mammalian counterparts and is a modifier of a Huntingtin poly-Q expansion and the Notch pathway. Dis Model Mech 2018; 11:dmm.029082. [PMID: 29208631 PMCID: PMC5818072 DOI: 10.1242/dmm.029082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 11/08/2017] [Indexed: 12/13/2022] Open
Abstract
Members of the Junctophilin (JPH) protein family have emerged as key actors in all excitable cells, with crucial implications for human pathophysiology. In mammals, this family consists of four members (JPH1-JPH4) that are differentially expressed throughout excitable cells. The analysis of knockout mice lacking JPH subtypes has demonstrated their essential contribution to physiological functions in skeletal and cardiac muscles and in neurons. Moreover, mutations in the human JPH2 gene are associated with hypertrophic and dilated cardiomyopathies; mutations in JPH3 are responsible for the neurodegenerative Huntington's disease-like-2 (HDL2), whereas JPH1 acts as a genetic modifier in Charcot–Marie–Tooth 2K peripheral neuropathy. Drosophila melanogaster has a single junctophilin (jp) gene, as is the case in all invertebrates, which might retain equivalent functions of the four homologous JPH genes present in mammalian genomes. Therefore, owing to the lack of putatively redundant genes, a jpDrosophila model could provide an excellent platform to model the Junctophilin-related diseases, to discover the ancestral functions of the JPH proteins and to reveal new pathways. By up- and downregulation of Jp in a tissue-specific manner in Drosophila, we show that altering its levels of expression produces a phenotypic spectrum characterized by muscular deficits, dilated cardiomyopathy and neuronal alterations. Importantly, our study has demonstrated that Jp modifies the neuronal degeneration in a Drosophila model of Huntington's disease, and it has allowed us to uncover an unsuspected functional relationship with the Notch pathway. Therefore, this Drosophila model has revealed new aspects of Junctophilin function that can be relevant for the disease mechanisms of their human counterparts. Summary: This work reveals that the Drosophila Junctophilin protein has similar functions to its mammalian homologues and uncovers new interactions of potential biomedical interest with Huntingtin and Notch signalling.
Collapse
Affiliation(s)
- Eduardo Calpena
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain
| | - Víctor López Del Amo
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain
| | - Mouli Chakraborty
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menendez Pelayo 4 acc 46010, Valencia, Spain.,Department of Genetics and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, c/ Dr Moliner 50, 46100 Burjasot, Spain
| | - Beatriz Llamusí
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menendez Pelayo 4 acc 46010, Valencia, Spain.,Department of Genetics and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, c/ Dr Moliner 50, 46100 Burjasot, Spain
| | - Rubén Artero
- Translational Genomics Group, Incliva Health Research Institute, Avda. Menendez Pelayo 4 acc 46010, Valencia, Spain.,Department of Genetics and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, c/ Dr Moliner 50, 46100 Burjasot, Spain
| | - Carmen Espinós
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain.,UPV-CIPF Joint Unit Disease Mechanisms and Nanomedicine, 46012 Valencia, Spain
| | - Máximo I Galindo
- Program in Molecular Mechanisms of Disease, Centro de Investigación Príncipe Felipe (CIPF), c/ Eduardo Primo Yúfera no. 3, 46012 Valencia, Spain .,UPV-CIPF Joint Unit Disease Mechanisms and Nanomedicine, 46012 Valencia, Spain.,Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46022 Valencia, Spain
| |
Collapse
|
39
|
Pappus SA, Mishra M. A Drosophila Model to Decipher the Toxicity of Nanoparticles Taken Through Oral Routes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1048:311-322. [DOI: 10.1007/978-3-319-72041-8_18] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Bolatto C, Parada C, Colmenares V. A Rapid and Efficient Method to Dissect Pupal Wings of Drosophila Suitable for Immunodetections or PCR Assays. J Vis Exp 2017. [PMID: 29364201 DOI: 10.3791/55854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Wing development in Drosophila melanogaster is an ideal model for studying morphogenesis at tissue level. These appendages develop from a group of cells named wing imaginal discs formed during embryonic development. In the larval stages the imaginal discs grow, increasing its number of cells and forming monolayered epithelial structures. Inside the pupal case, the imaginal discs bud out and fold into bilayers along a line that becomes the future margin of the wing. During this process, the longitudinal primodia veins originate vein cells on the prospective dorsal and ventral surfaces of the wing. During the pupal stage the stripes of vein cells of each surface communicate in order to generate tight tubes; at the same time, the cross-veins begin their formation. With the help of appropriate molecular markers, it is possible to identify the major elements composing the wing during its development. For this reason, the ability to accurately detect transcripts or proteins in this structure is critical for studying their abundance and localization related to the development process of the wing. The procedure described here focuses on manipulating pupal wings, providing detailed instructions on how to dissect the wing during the pupal stage. The dissection of pupal tissue is more difficult to perform than their counterparts in third instar larvae. This is why this approach was developed, to obtain rapid and efficient high quality samples. Details of how to immunostain and mount these wing samples, to allow the visualization of proteins or cell components, are provided in the protocol. With little expertise it is possible to collect 8-10 high quality pupal wings in a short amount of time.
Collapse
Affiliation(s)
- Carmen Bolatto
- Departamento de Histología y Embriología, Facultad de Medicina, Laboratorio de Biología del Desarrollo;
| | - Cristina Parada
- Departamento de Histología y Embriología, Facultad de Medicina, Laboratorio de Biología del Desarrollo
| | - Victoria Colmenares
- Departamento de Histología y Embriología, Facultad de Medicina, Laboratorio de Biología del Desarrollo
| |
Collapse
|
41
|
Nagel AC, Auer JS, Schulz A, Pfannstiel J, Yuan Z, Collins CE, Kovall RA, Preiss A. Phosphorylation of Suppressor of Hairless impedes its DNA-binding activity. Sci Rep 2017; 7:11820. [PMID: 28928428 PMCID: PMC5605572 DOI: 10.1038/s41598-017-11952-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/01/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signalling activity governs cellular differentiation in higher metazoa, where Notch signals are transduced by the transcription factor CSL, called Suppressor of Hairless [Su(H)] in Drosophila. Su(H) operates as molecular switch on Notch target genes: within activator complexes, including intracellular Notch, or within repressor complexes, including the antagonist Hairless. Mass spectrometry identified phosphorylation on Serine 269 in Su(H), potentially serving as a point of cross-regulation by other signalling pathways. To address the biological significance, we generated phospho-deficient [Su(H)S269A] and phospho-mimetic [Su(H)S269D] variants: the latter displayed reduced transcriptional activity despite unaltered protein interactions with co-activators and -repressors. Based on the Su(H) structure, Ser269 phosphorylation may interfere with DNA-binding, which we confirmed by electro-mobility shift assay and isothermal titration calorimetry. Overexpression of Su(H)S269D during fly development demonstrated reduced transcriptional regulatory activity, similar to the previously reported DNA-binding defective mutant Su(H)R266H. As both are able to bind Hairless and Notch proteins, Su(H)S269D and Su(H)R266H provoked dominant negative effects upon overexpression. Our data imply that Ser269 phosphorylation impacts Notch signalling activity by inhibiting DNA-binding of Su(H), potentially affecting both activation and repression. Ser269 is highly conserved in vertebrate CSL homologues, opening the possibility of a general and novel mechanism of modulating Notch signalling activity.
Collapse
Affiliation(s)
- Anja C Nagel
- Institut für Genetik (240), University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany.
| | - Jasmin S Auer
- Institut für Genetik (240), University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| | - Adriana Schulz
- Institut für Genetik (240), University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| | - Jens Pfannstiel
- Core Facility Hohenheim, Mass Spectrometry Unit University of Hohenheim, 70599, Stuttgart, Germany
| | - Zhenyu Yuan
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Courtney E Collins
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Rhett A Kovall
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Anette Preiss
- Institut für Genetik (240), University of Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| |
Collapse
|
42
|
Upadhyay A, Moss-Taylor L, Kim MJ, Ghosh AC, O'Connor MB. TGF-β Family Signaling in Drosophila. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022152. [PMID: 28130362 DOI: 10.1101/cshperspect.a022152] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The transforming growth factor β (TGF-β) family signaling pathway is conserved and ubiquitous in animals. In Drosophila, fewer representatives of each signaling component are present compared with vertebrates, simplifying mechanistic study of the pathway. Although there are fewer family members, the TGF-β family pathway still regulates multiple and diverse functions in Drosophila. In this review, we focus our attention on several of the classic and best-studied functions for TGF-β family signaling in regulating Drosophila developmental processes such as embryonic and imaginal disc patterning, but we also describe several recently discovered roles in regulating hormonal, physiological, neuronal, innate immunity, and tissue homeostatic processes.
Collapse
Affiliation(s)
- Ambuj Upadhyay
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Lindsay Moss-Taylor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Arpan C Ghosh
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
43
|
Ishimoto Y, Sugimura K. A mechanical model for diversified insect wing margin shapes. J Theor Biol 2017; 427:17-27. [PMID: 28549619 DOI: 10.1016/j.jtbi.2017.05.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 11/24/2022]
Abstract
The wings in different insect species are morphologically distinct with regards to their size, outer contour (margin) shape, venation, and pigmentation. The basis of the diversity of wing margin shapes remains unknown, despite the fact that gene networks governing the Drosophila wing development have been well characterised. Among the different types of wing margin shapes, smoothly curved contour is the most frequently found and implies the existence of a highly organised, multicellular mechanical structure. Here, we developed a mechanical model for diversified insect wing margin shapes, in which non-uniform bending stiffness of the wing margin is considered. We showed that a variety of spatial distribution of the bending stiffness could reproduce diverse wing margin shapes. Moreover, the inference of the distribution of the bending stiffness from experimental images indicates a common spatial profile among insects tested. We further studied the effect of the intrinsic tension of the wing blade on the margin shape and on the inferred bending stiffness. Finally, we implemented the bending stiffness of the wing margin in the cell vertex model of the wing blade, and confirmed that the hybrid model retains the essential feature of the margin model. We propose that in addition to morphogenetic processes in the wing blade, the spatial profile of the bending stiffness in the wing margin can play a pivotal role in shaping insect wings.
Collapse
Affiliation(s)
- Yukitaka Ishimoto
- Department of Machine Intelligence and Systems Engineering, Akita Prefectural University, Akita 015-0055, Japan.
| | - Kaoru Sugimura
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan; JST PRESTO, Tokyo 102-0075, Japan
| |
Collapse
|
44
|
Scarpella E. The logic of plant vascular patterning. Polarity, continuity and plasticity in the formation of the veins and of their networks. Curr Opin Genet Dev 2017; 45:34-43. [DOI: 10.1016/j.gde.2017.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 10/20/2022]
|
45
|
Martín M, Ostalé CM, de Celis JF. Patterning of the Drosophila L2 vein is driven by regulatory interactions between region-specific transcription factors expressed in response to Dpp signalling. Development 2017; 144:3168-3176. [PMID: 28760811 DOI: 10.1242/dev.143461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 07/25/2017] [Indexed: 01/31/2023]
Abstract
Pattern formation relies on the generation of transcriptional landscapes regulated by signalling pathways. A paradigm of epithelial patterning is the distribution of vein territories in the Drosophila wing disc. In this tissue, Decapentaplegic signalling regulates its target genes at different distances from the source of the ligand. The transformation of signalling into coherent territories of gene expression requires regulatory cross-interactions between these target genes. Here, we analyse the mechanisms generating the domain of knirps expression in the presumptive L2 vein of the wing imaginal disc. We find that knirps is regulated by four Decapentaplegic target genes encoding the transcription factors aristaless, spalt major, spalt-related and optix The expression of optix is activated by Dpp and repressed by the Spalt proteins, becoming restricted to the most anterior region of the wing blade. In turn, the expression of knirps is activated by Aristaless and repressed by Optix and the Spalt proteins. In this manner, the expression of knirps becomes restricted to those cells where Spalt levels are sufficient to repress optix, but not sufficient to repress knirps.
Collapse
Affiliation(s)
- Mercedes Martín
- Centro de Biología Molecular 'Severo Ochoa', CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Cristina M Ostalé
- Centro de Biología Molecular 'Severo Ochoa', CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Jose F de Celis
- Centro de Biología Molecular 'Severo Ochoa', CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
46
|
Dahal GR, Pradhan SJ, Bates EA. Inwardly rectifying potassium channels influence Drosophila wing morphogenesis by regulating Dpp release. Development 2017; 144:2771-2783. [PMID: 28684627 DOI: 10.1242/dev.146647] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 06/14/2017] [Indexed: 12/23/2022]
Abstract
Loss of embryonic ion channel function leads to morphological defects, but the underlying reason for these defects remains elusive. Here, we show that inwardly rectifying potassium (Irk) channels regulate release of the Drosophila bone morphogenetic protein Dpp in the developing fly wing and that this is necessary for developmental signaling. Inhibition of Irk channels decreases the incidence of distinct Dpp-GFP release events above baseline fluorescence while leading to a broader distribution of Dpp-GFP. Work by others in different cell types has shown that Irk channels regulate peptide release by modulating membrane potential and calcium levels. We found calcium transients in the developing wing, and inhibition of Irk channels reduces the duration and amplitude of calcium transients. Depolarization with high extracellular potassium evokes Dpp release. Taken together, our data implicate Irk channels as a requirement for regulated release of Dpp, highlighting the importance of the temporal pattern of Dpp presentation for morphogenesis of the wing.
Collapse
Affiliation(s)
- Giri Raj Dahal
- University of Colorado Denver School of Medicine, 12800 E 19th Avenue, Aurora, CO 80045, USA
| | - Sarala Joshi Pradhan
- University of Colorado Denver School of Medicine, 12800 E 19th Avenue, Aurora, CO 80045, USA
| | - Emily Anne Bates
- University of Colorado Denver School of Medicine, 12800 E 19th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
47
|
Abstract
Although the insect wing is a textbook example of morphological novelty, the origin of insect wings remains a mystery and is regarded as a chief conundrum in biology. Centuries of debates have culminated into two prominent hypotheses: the tergal origin hypothesis and the pleural origin hypothesis. However, between these two hypotheses, there is little consensus in regard to the origin tissue of the wing as well as the evolutionary route from the origin tissue to the functional flight device. Recent evolutionary developmental (evo-devo) studies have shed new light on the origin of insect wings. A key concept in these studies is “serial homology”. In this review, we discuss how the wing serial homologs identified in recent evo-devo studies have provided a new angle through which this century-old conundrum can be explored. We also review what we have learned so far from wing serial homologs and discuss what we can do to go beyond simply identifying wing serial homologs and delve further into the developmental and genetic mechanisms that have facilitated the evolution of insect wings.
Collapse
Affiliation(s)
- Yoshinori Tomoyasu
- Department of Biology, Miami University, Pearson Hall, 700E High Street, Oxford, OH 45056, USA
| | - Takahiro Ohde
- Division of Evolutionary Developmental Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki 444-8585, Japan.,Department of Basic Biology, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 38 Nishigonaka Myodaiji, Okazaki 444-8585, Japan
| | - Courtney Clark-Hachtel
- Department of Biology, Miami University, Pearson Hall, 700E High Street, Oxford, OH 45056, USA
| |
Collapse
|
48
|
Sabat D, Patnaik A, Ekka B, Dash P, Mishra M. Investigation of titania nanoparticles on behaviour and mechanosensory organ of Drosophila melanogaster. Physiol Behav 2016; 167:76-85. [DOI: 10.1016/j.physbeh.2016.08.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/06/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
|
49
|
Hadjivasiliou Z, Hunter GL, Baum B. A new mechanism for spatial pattern formation via lateral and protrusion-mediated lateral signalling. J R Soc Interface 2016; 13:20160484. [PMID: 27807273 PMCID: PMC5134009 DOI: 10.1098/rsif.2016.0484] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 10/11/2016] [Indexed: 02/06/2023] Open
Abstract
Tissue organization and patterning are critical during development when genetically identical cells take on different fates. Lateral signalling plays an important role in this process by helping to generate self-organized spatial patterns in an otherwise uniform collection of cells. Recent data suggest that lateral signalling can be mediated both by junctional contacts between neighbouring cells and via cellular protrusions that allow non-neighbouring cells to interact with one another at a distance. However, it remains unclear precisely how signalling mediated by these distinct types of cell-cell contact can physically contribute to the generation of complex patterns without the assistance of diffusible morphogens or pre-patterns. To explore this question, in this work we develop a model of lateral signalling based on a single receptor/ligand pair as exemplified by Notch and Delta. We show that allowing the signalling kinetics to differ at junctional versus protrusion-mediated contacts, an assumption inspired by recent data which show that the cleavage of Notch in several systems requires both Delta binding and the application of mechanical force, permits individual cells to act to promote both lateral activation and lateral inhibition. Strikingly, under this model, in which Delta can sequester Notch, a variety of patterns resembling those typical of reaction-diffusion systems is observed, together with more unusual patterns that arise when we consider changes in signalling kinetics, and in the length and distribution of protrusions. Importantly, these patterns are self-organizing-so that local interactions drive tissue-scale patterning. Together, these data show that protrusions can, in principle, generate different types of patterns in addition to contributing to long-range signalling and to pattern refinement.
Collapse
Affiliation(s)
- Zena Hadjivasiliou
- Centre for Mathematics, Physics, and Engineering in the Life Sciences and Experimental Biology, University College London, London WC1E 6BT, UK
- Department of Genetics, Evolution and Environment, University College London, London WC1E 6BT, UK
| | - Ginger L Hunter
- MRC-Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
- Institute of Physics of Living Systems, University College London, London WC1E 6BT, UK
| | - Buzz Baum
- MRC-Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
- Institute of Physics of Living Systems, University College London, London WC1E 6BT, UK
| |
Collapse
|
50
|
Smylla TK, Preiss A, Maier D. In vivo analysis of internal ribosome entry at the Hairless locus by genome engineering in Drosophila. Sci Rep 2016; 6:34881. [PMID: 27713501 PMCID: PMC5054391 DOI: 10.1038/srep34881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/21/2016] [Indexed: 12/23/2022] Open
Abstract
Cell communication in metazoans requires the highly conserved Notch signaling pathway, which is subjected to strict regulation of both activation and silencing. In Drosophila melanogaster, silencing involves the assembly of a repressor complex by Hairless (H) on Notch target gene promoters. We previously found an in-frame internal ribosome entry site in the full length H transcript resulting in two H protein isoforms (Hp120 and Hp150). Hence, H may repress Notch signalling activity in situations where cap-dependent translation is inhibited. Here we demonstrate the in vivo importance of both H isoforms for proper fly development. To this end, we replaced the endogenous H locus by constructs specifically affecting translation of either Hp150 or Hp120 isoforms using genome engineering. Our findings indicate the functional relevance of both H proteins. Based on bristle phenotypes, the predominant isoform Hp150 appears to be of particular importance. In contrast, growth regulation and venation of the wing require the concomitant activity of both isoforms. Finally, the IRES dependent production of Hp120 during mitosis was verified in vivo. Together our data confirm IRES mediated translation of H protein in vivo, supporting strict regulation of Notch in different cellular settings.
Collapse
Affiliation(s)
- Thomas K Smylla
- Universität Hohenheim, Institut für Genetik (240), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anette Preiss
- Universität Hohenheim, Institut für Genetik (240), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Dieter Maier
- Universität Hohenheim, Institut für Genetik (240), Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|