1
|
Li T, Shi W, Ho MS, Zhang YQ. A Pvr-AP-1-Mmp1 signaling pathway is activated in astrocytes upon traumatic brain injury. eLife 2024; 12:RP87258. [PMID: 39480704 PMCID: PMC11527428 DOI: 10.7554/elife.87258] [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] [Indexed: 11/02/2024] Open
Abstract
Traumatic brain injury (TBI) caused by external mechanical forces is a major health burden worldwide, but the underlying mechanism in glia remains largely unclear. We report herein that Drosophila adults exhibit a defective blood-brain barrier, elevated innate immune responses, and astrocyte swelling upon consecutive strikes with a high-impact trauma device. RNA sequencing (RNA-seq) analysis of these astrocytes revealed upregulated expression of genes encoding PDGF and VEGF receptor-related (Pvr, a receptor tyrosine kinase), adaptor protein complex 1 (AP-1, a transcription factor complex of the c-Jun N-terminal kinase pathway) composed of Jun-related antigen (Jra) and kayak (kay), and matrix metalloproteinase 1 (Mmp1) following TBI. Interestingly, Pvr is both required and sufficient for AP-1 and Mmp1 upregulation, while knockdown of AP-1 expression in the background of Pvr overexpression in astrocytes rescued Mmp1 upregulation upon TBI, indicating that Pvr acts as the upstream receptor for the downstream AP-1-Mmp1 transduction. Moreover, dynamin-associated endocytosis was found to be an important regulatory step in downregulating Pvr signaling. Our results identify a new Pvr-AP-1-Mmp1 signaling pathway in astrocytes in response to TBI, providing potential targets for developing new therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Tingting Li
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Wenwen Shi
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Margaret S Ho
- Institute of Neuroscience, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Brain Research Center, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yong Q Zhang
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
2
|
Lee JR, Boothe T, Mauksch C, Thommen A, Rink JC. Epidermal turnover in the planarian Schmidtea mediterranea involves basal cell extrusion and intestinal digestion. Cell Rep 2024; 43:114305. [PMID: 38906148 DOI: 10.1016/j.celrep.2024.114305] [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: 11/06/2023] [Revised: 03/21/2024] [Accepted: 05/15/2024] [Indexed: 06/23/2024] Open
Abstract
Planarian flatworms undergo continuous internal turnover, wherein old cells are replaced by the division progeny of adult pluripotent stem cells (neoblasts). How cell turnover is carried out at the organismal level remains an intriguing question in planarians and other systems. While previous studies have predominantly focused on neoblast proliferation, little is known about the processes that mediate cell loss during tissue homeostasis. Here, we use the planarian epidermis as a model to study the mechanisms of cell removal. We established a covalent dye-labeling assay and image analysis pipeline to quantify the cell turnover rate in the planarian epidermis. Our findings indicate that the ventral epidermis is highly dynamic and epidermal cells undergo internalization via basal extrusion, followed by a relocation toward the intestine and ultimately digestion by intestinal phagocytes. Overall, our study reveals a complex homeostatic process of cell clearance that may generally allow planarians to catabolize their own cells.
Collapse
Affiliation(s)
- Jun-Ru Lee
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany; Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, 37077 Göttingen, Germany
| | - Tobias Boothe
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Clemens Mauksch
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Albert Thommen
- Cancer Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jochen C Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
3
|
Garcia-Arias JM, Pinal N, Cristobal-Vargas S, Estella C, Morata G. Lack of apoptosis leads to cellular senescence and tumorigenesis in Drosophila epithelial cells. Cell Death Discov 2023; 9:281. [PMID: 37532716 PMCID: PMC10397273 DOI: 10.1038/s41420-023-01583-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023] Open
Abstract
Programmed cell death (apoptosis) is a homeostasis program of animal tissues designed to remove cells that are unwanted or are damaged by physiological insults. To assess the functional role of apoptosis, we have studied the consequences of subjecting Drosophila epithelial cells defective in apoptosis to stress or genetic perturbations that normally cause massive cell death. We find that many of those cells acquire persistent activity of the JNK pathway, which drives them into senescent status, characterized by arrest of cell division, cell hypertrophy, Senescent Associated ß-gal activity (SA-ß-gal), reactive oxygen species (ROS) production, Senescent Associated Secretory Phenotype (SASP) and migratory behaviour. We have identified two classes of senescent cells in the wing disc: 1) those that localize to the appendage part of the disc, express the upd, wg and dpp signalling genes and generate tumour overgrowths, and 2) those located in the thoracic region do not express wg and dpp nor they induce tumour overgrowths. Whether to become tumorigenic or non-tumorigenic depends on the original identity of the cell prior to the transformation. We also find that the p53 gene contributes to senescence by enhancing the activity of JNK.
Collapse
Affiliation(s)
- Juan Manuel Garcia-Arias
- Laboratory of Tumorogenesis and Regeneration. Centro de Biología Molecular CSIC-UAM, Madrid, Spain
| | - Noelia Pinal
- Laboratory of Tumorogenesis and Regeneration. Centro de Biología Molecular CSIC-UAM, Madrid, Spain
| | - Sara Cristobal-Vargas
- Laboratory of Gene expression control, patterning and growth during appendage development. Centro de Biología Molecular CSIC-UAM, Madrid, Spain
- Cancer Research Center-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Carlos Estella
- Laboratory of Gene expression control, patterning and growth during appendage development. Centro de Biología Molecular CSIC-UAM, Madrid, Spain.
| | - Ginés Morata
- Laboratory of Tumorogenesis and Regeneration. Centro de Biología Molecular CSIC-UAM, Madrid, Spain.
| |
Collapse
|
4
|
Lei Y, Huang Y, Yang K, Cao X, Song Y, Martín-Blanco E, Pastor-Pareja JC. FGF signaling promotes spreading of fat body precursors necessary for adult adipogenesis in Drosophila. PLoS Biol 2023; 21:e3002050. [PMID: 36947563 PMCID: PMC10069774 DOI: 10.1371/journal.pbio.3002050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 04/03/2023] [Accepted: 02/24/2023] [Indexed: 03/23/2023] Open
Abstract
Knowledge of adipogenetic mechanisms is essential to understand and treat conditions affecting organismal metabolism and adipose tissue health. In Drosophila, mature adipose tissue (fat body) exists in larvae and adults. In contrast to the well-known development of the larval fat body from the embryonic mesoderm, adult adipogenesis has remained mysterious. Furthermore, conclusive proof of its physiological significance is lacking. Here, we show that the adult fat body originates from a pool of undifferentiated mesodermal precursors that migrate from the thorax into the abdomen during metamorphosis. Through in vivo imaging, we found that these precursors spread from the ventral midline and cover the inner surface of the abdomen in a process strikingly reminiscent of embryonic mesoderm migration, requiring fibroblast growth factor (FGF) signaling as well. FGF signaling guides migration dorsally and regulates adhesion to the substrate. After spreading is complete, precursor differentiation involves fat accumulation and cell fusion that produces mature binucleate and tetranucleate adipocytes. Finally, we show that flies where adult adipogenesis is impaired by knock down of FGF receptor Heartless or transcription factor Serpent display ectopic fat accumulation in oenocytes and decreased resistance to starvation. Our results reveal that adult adipogenesis occurs de novo during metamorphosis and demonstrate its crucial physiological role.
Collapse
Affiliation(s)
- Yuting Lei
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuwei Huang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Ke Yang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xueya Cao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuzhao Song
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Enrique Martín-Blanco
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Científic de Barcelona, Barcelona, Spain
| | - José Carlos Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Institute of Neurosciences, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
5
|
Friesen S, Hariharan IK. Coordinated growth of linked epithelia is mediated by the Hippo pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.26.530099. [PMID: 36993542 PMCID: PMC10054945 DOI: 10.1101/2023.02.26.530099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
An epithelium in a living organism seldom develops in isolation. Rather, most epithelia are tethered to other epithelial or non-epithelial tissues, necessitating growth coordination between layers. We investigated how two tethered epithelial layers of the Drosophila larval wing imaginal disc, the disc proper (DP) and the peripodial epithelium (PE), coordinate their growth. DP growth is driven by the morphogens Hedgehog (Hh) and Dpp, but regulation of PE growth is poorly understood. We find that the PE adapts to changes in growth rates of the DP, but not vice versa, suggesting a "leader and follower" mechanism. Moreover, PE growth can occur by cell shape changes, even when proliferation is inhibited. While Hh and Dpp pattern gene expression in both layers, growth of the DP is exquisitely sensitive to Dpp levels, while growth of the PE is not; the PE can achieve an appropriate size even when Dpp signaling is inhibited. Instead, both the growth of the PE and its accompanying cell shape changes require the activity of two components of the mechanosensitive Hippo pathway, the DNA-binding protein Scalloped (Sd) and its co-activator (Yki), which could allow the PE to sense and respond to forces generated by DP growth. Thus, an increased reliance on mechanically-dependent growth mediated by the Hippo pathway, at the expense of morphogen-dependent growth, enables the PE to evade layer-intrinsic growth control mechanisms and coordinate its growth with the DP. This provides a potential paradigm for growth coordination between different components of a developing organ.
Collapse
Affiliation(s)
- Sophia Friesen
- Department of Molecular and Cell Biology, University of California, Berkeley
| | - Iswar K. Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley
| |
Collapse
|
6
|
Growth anisotropy of the extracellular matrix shapes a developing organ. Nat Commun 2023; 14:1220. [PMID: 36869053 PMCID: PMC9984492 DOI: 10.1038/s41467-023-36739-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Final organ size and shape result from volume expansion by growth and shape changes by contractility. Complex morphologies can also arise from differences in growth rate between tissues. We address here how differential growth guides the morphogenesis of the growing Drosophila wing imaginal disc. We report that 3D morphology results from elastic deformation due to differential growth anisotropy between the epithelial cell layer and its enveloping extracellular matrix (ECM). While the tissue layer grows in plane, growth of the bottom ECM occurs in 3D and is reduced in magnitude, thereby causing geometric frustration and tissue bending. The elasticity, growth anisotropy and morphogenesis of the organ are fully captured by a mechanical bilayer model. Moreover, differential expression of the Matrix metalloproteinase MMP2 controls growth anisotropy of the ECM envelope. This study shows that the ECM is a controllable mechanical constraint whose intrinsic growth anisotropy directs tissue morphogenesis in a developing organ.
Collapse
|
7
|
Karkali K, Saunders TE, Panayotou G, Martín-Blanco E. JNK signaling in pioneer neurons organizes ventral nerve cord architecture in Drosophila embryos. Nat Commun 2023; 14:675. [PMID: 36750572 PMCID: PMC9905486 DOI: 10.1038/s41467-023-36388-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Morphogenesis of the Central Nervous System (CNS) is a complex process that obeys precise architectural rules. Yet, the mechanisms dictating these rules remain unknown. Analyzing morphogenesis of the Drosophila embryo Ventral Nerve Cord (VNC), we observe that a tight control of JNK signaling is essential for attaining the final VNC architecture. JNK signaling in a specific subset of pioneer neurons autonomously regulates the expression of Fasciclin 2 (Fas 2) and Neurexin IV (Nrx IV) adhesion molecules, probably via the transcription factor zfh1. Interfering at any step in this cascade affects fasciculation along pioneer axons, leading to secondary cumulative scaffolding defects during the structural organization of the axonal network. The global disorder of architectural landmarks ultimately influences nervous system condensation. In summary, our data point to JNK signaling in a subset of pioneer neurons as a key element underpinning VNC architecture, revealing critical milestones on the mechanism of control of its structural organization.
Collapse
Affiliation(s)
- Katerina Karkali
- Instituto de Biología Molecular de Barcelona (CSIC), Parc Cientific de Barcelona, Baldiri Reixac 10-12, 08028, Barcelona, Spain
- Mechanobiology Institute and Department of Biological Sciences, 5 Engineering Drive 1, National University of Singapore, Singapore, 117411, Singapore
- BSRC Alexander Fleming, 34 Fleming Street, 16672, Vari, Greece
| | - Timothy E Saunders
- Mechanobiology Institute and Department of Biological Sciences, 5 Engineering Drive 1, National University of Singapore, Singapore, 117411, Singapore
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | | | - Enrique Martín-Blanco
- Instituto de Biología Molecular de Barcelona (CSIC), Parc Cientific de Barcelona, Baldiri Reixac 10-12, 08028, Barcelona, Spain.
- Mechanobiology Institute and Department of Biological Sciences, 5 Engineering Drive 1, National University of Singapore, Singapore, 117411, Singapore.
| |
Collapse
|
8
|
Bonche R, Smolen P, Chessel A, Boisivon S, Pisano S, Voigt A, Schaub S, Thérond P, Pizette S. Regulation of the collagen IV network by the basement membrane protein perlecan is crucial for squamous epithelial cell morphogenesis and organ architecture. Matrix Biol 2022; 114:35-66. [PMID: 36343860 DOI: 10.1016/j.matbio.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
All epithelia have their basal side in contact with a specialized extracellular matrix, the basement membrane (BM). During development, the BM contributes to the shaping of epithelial organs via its mechanical properties. These properties rely on two core components of the BM, collagen type IV and perlecan/HSPG2, which both interact with another core component, laminin, the initiator of BM assembly. While collagen type IV supplies the BM with rigidity to constrain the tissue, perlecan antagonizes this effect. Nevertheless, the number of organs that has been studied is still scarce, and given that epithelial tissues exhibit a wide array of shapes, their forms are bound to be regulated by distinct mechanisms. This is underscored by mounting evidence that BM composition and assembly/biogenesis is tissue-specific. Moreover, previous reports have essentially focused on the mechanical role of the BM in morphogenesis at the tissue scale, but not the cell scale. Here, we took advantage of the robust conservation of core BM proteins and the limited genetic redundancy of the Drosophila model system to address how this matrix shapes the wing imaginal disc, a complex organ comprising a squamous, a cuboidal and a columnar epithelium. With the use of a hypomorphic allele, we show that the depletion of Trol (Drosophila perlecan) affects the morphogenesis of the three epithelia, but particularly that of the squamous one. The planar surface of the squamous epithelium (SE) becomes extremely narrow, due to a function for Trol in the control of the squamous shape of its cells. Furthermore, we find that the lack of Trol impairs the biogenesis of the BM of the SE by modifying the structure of the collagen type IV lattice. Through atomic force microscopy and laser surgery, we demonstrate that Trol provides elasticity to the SE's BM, thereby regulating the mechanical properties of the SE. Moreover, we show that Trol acts via collagen type IV, since the global reduction in the trol mutant context of collagen type IV or the enzyme that cross-links its 7S -but not the enzyme that cross-links its NC1- domain substantially restores the morphogenesis of the SE. In addition, a stronger decrease in collagen type IV achieved by the overexpression of the matrix metalloprotease 2 exclusively in the BM of the SE, significantly rescues the organization of the two other epithelia. Our data thus sustain a model in which Trol counters the rigidity conveyed by collagen type IV to the BM of the SE, via the regulation of the NC1-dependant assembly of its scaffold, allowing the spreading of the squamous cells, spreading which is compulsory for the architecture of the whole organ.
Collapse
Affiliation(s)
| | - Prune Smolen
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| | | | | | | | - Aaron Voigt
- Department of Neurology, University Medical Center, RWTH Aachen University, Aachen 52074, Germany
| | | | | | | |
Collapse
|
9
|
Serras F. The sooner, the better: ROS, kinases and nutrients at the onset of the damage response in Drosophila. Front Cell Dev Biol 2022; 10:1047823. [PMID: 36353511 PMCID: PMC9637634 DOI: 10.3389/fcell.2022.1047823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/11/2022] [Indexed: 11/20/2022] Open
Abstract
One of the main topics in regeneration biology is the nature of the early signals that trigger the damage response. Recent advances in Drosophila point to the MAP3 kinase Ask1 as a molecular hub that integrates several signals at the onset of regeneration. It has been discovered that reactive oxygen species (ROS) produced in damaged imaginal discs and gut epithelia will activate the MAP3 kinase Ask1. Severely damaged and apoptotic cells produce an enormous amount of ROS, which ensures their elimination by activating Ask1 and in turn the pro-apoptotic function of JNK. However, this creates an oxidative stress environment with beneficial effects that is sensed by neighboring healthy cells. This environment, in addition to the Pi3K/Akt nutrient sensing pathway, can be integrated into Ask1 to launch regeneration. Ultimately the activity of Ask1 depends on these and other inputs and modulates its signaling to achieve moderate levels of p38 and low JNK signaling and thus promote survival and regeneration. This model based on the dual function of Ask1 for early response to damage is discussed here.
Collapse
Affiliation(s)
- Florenci Serras
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, and Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| |
Collapse
|
10
|
Smith SJ, Guillon E, Holley SA. The roles of inter-tissue adhesion in development and morphological evolution. J Cell Sci 2022; 135:275268. [PMID: 35522159 PMCID: PMC9264361 DOI: 10.1242/jcs.259579] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The study of how neighboring tissues physically interact with each other, inter-tissue adhesion, is an emerging field at the interface of cell biology, biophysics and developmental biology. Inter-tissue adhesion can be mediated by either cell-extracellular matrix adhesion or cell-cell adhesion, and both the mechanisms and consequences of inter-tissue adhesion have been studied in vivo in numerous vertebrate and invertebrate species. In this Review, we discuss recent progress in understanding the many functions of inter-tissue adhesion in development and evolution. Inter-tissue adhesion can couple the motion of adjacent tissues, be the source of mechanical resistance that constrains morphogenesis, and transmit tension required for normal development. Tissue-tissue adhesion can also create mechanical instability that leads to tissue folding or looping. Transient inter-tissue adhesion can facilitate tissue invasion, and weak tissue adhesion can generate friction that shapes and positions tissues within the embryo. Lastly, we review studies that reveal how inter-tissue adhesion contributes to the diversification of animal morphologies.
Collapse
Affiliation(s)
- Sarah Jacquelyn Smith
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Emilie Guillon
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
11
|
The Tbx6 Transcription Factor Dorsocross Mediates Dpp Signaling to Regulate Drosophila Thorax Closure. Int J Mol Sci 2022; 23:ijms23094543. [PMID: 35562934 PMCID: PMC9104307 DOI: 10.3390/ijms23094543] [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: 03/11/2022] [Revised: 04/08/2022] [Accepted: 04/17/2022] [Indexed: 11/23/2022] Open
Abstract
Movement and fusion of separate cell populations are critical for several developmental processes, such as neural tube closure in vertebrates or embryonic dorsal closure and pupal thorax closure in Drosophila. Fusion failure results in an opening or groove on the body surface. Drosophila pupal thorax closure is an established model to investigate the mechanism of tissue closure. Here, we report the identification of T-box transcription factor genes Dorsocross (Doc) as Decapentaplegic (Dpp) targets in the leading edge cells of the notum in the late third instar larval and early pupal stages. Reduction of Doc in the notum region results in a thorax closure defect, similar to that in dpp loss-of-function flies. Nine genes are identified as potential downstream targets of Doc in regulating thorax closure by molecular and genetic screens. Our results reveal a novel function of Doc in Drosophila development. The candidate target genes provide new clues for unravelling the mechanism of collective cell movement.
Collapse
|
12
|
Abstract
The Drosophila wing imaginal disc is a tissue of undifferentiated cells that are precursors of the wing and most of the notum of the adult fly. The wing disc first forms during embryogenesis from a cluster of ∼30 cells located in the second thoracic segment, which invaginate to form a sac-like structure. They undergo extensive proliferation during larval stages to form a mature larval wing disc of ∼35,000 cells. During this time, distinct cell fates are assigned to different regions, and the wing disc develops a complex morphology. Finally, during pupal stages the wing disc undergoes morphogenetic processes and then differentiates to form the adult wing and notum. While the bulk of the wing disc comprises epithelial cells, it also includes neurons and glia, and is associated with tracheal cells and muscle precursor cells. The relative simplicity and accessibility of the wing disc, combined with the wealth of genetic tools available in Drosophila, have combined to make it a premier system for identifying genes and deciphering systems that play crucial roles in animal development. Studies in wing imaginal discs have made key contributions to many areas of biology, including tissue patterning, signal transduction, growth control, regeneration, planar cell polarity, morphogenesis, and tissue mechanics.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
13
|
Athilingam T, Parihar SS, Bhattacharya R, Rizvi MS, Kumar A, Sinha P. Proximate larval epidermal cell layer generates forces for Pupal thorax closure in Drosophila. Genetics 2022; 221:6528854. [PMID: 35166774 PMCID: PMC9071563 DOI: 10.1093/genetics/iyac030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 11/14/2022] Open
Abstract
During tissue closures, such as embryonic dorsal closure in Drosophila melanogaster, a proximate extra-embryonic layer, amnioserosa, generates forces that drive migration of the flanking lateral embryonic epidermis, thereby zip-shutting the embryo. Arguably, this paradigm of tissue closure is also recapitulated in mammalian wound healing wherein proximate fibroblasts transform into contractile myofibroblasts, develop cell junctions, and form a tissue layer de novo: contraction of the latter then aids in wound closure. Given this parallelism between disparate exemplars, we posit a general principle of tissue closure via proximate cell layer-generated forces. Here, we have tested this hypothesis in pupal thorax closure wherein 2 halves of the presumptive adult thorax of Drosophila, the contralateral heminotal epithelia, migrate over an underlying larval epidermal cell layer. We show that the proximate larval epidermal cell layer promotes thorax closure by its active contraction, orchestrated by its elaborate actomyosin network-driven epithelial cell dynamics, cell delamination, and death-the latter being prefigured by the activation of caspases. Larval epidermal cell dynamics generate contraction forces, which when relayed to the flanking heminota-via their mutual integrin-based adhesions-mediate thorax closure. Compromising any of these contraction force-generating mechanisms in the larval epidermal cell layer slows down heminotal migration, while loss of its relay to the flanking heminota abrogates the thorax closure altogether. Mathematical modeling further reconciles the biophysical underpinning of this emergent mechanism of thorax closure. Revealing mechanism of thorax closure apart, these findings show conservation of an essential principle of a proximate cell layer-driven tissue closure.
Collapse
Affiliation(s)
- Thamarailingam Athilingam
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Saurabh S Parihar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Rachita Bhattacharya
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Mohd S Rizvi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Amit Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Pradip Sinha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India,Corresponding author: Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
| |
Collapse
|
14
|
Mierke CT. Viscoelasticity, Like Forces, Plays a Role in Mechanotransduction. Front Cell Dev Biol 2022; 10:789841. [PMID: 35223831 PMCID: PMC8864183 DOI: 10.3389/fcell.2022.789841] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Viscoelasticity and its alteration in time and space has turned out to act as a key element in fundamental biological processes in living systems, such as morphogenesis and motility. Based on experimental and theoretical findings it can be proposed that viscoelasticity of cells, spheroids and tissues seems to be a collective characteristic that demands macromolecular, intracellular component and intercellular interactions. A major challenge is to couple the alterations in the macroscopic structural or material characteristics of cells, spheroids and tissues, such as cell and tissue phase transitions, to the microscopic interferences of their elements. Therefore, the biophysical technologies need to be improved, advanced and connected to classical biological assays. In this review, the viscoelastic nature of cytoskeletal, extracellular and cellular networks is presented and discussed. Viscoelasticity is conceptualized as a major contributor to cell migration and invasion and it is discussed whether it can serve as a biomarker for the cells' migratory capacity in several biological contexts. It can be hypothesized that the statistical mechanics of intra- and extracellular networks may be applied in the future as a powerful tool to explore quantitatively the biomechanical foundation of viscoelasticity over a broad range of time and length scales. Finally, the importance of the cellular viscoelasticity is illustrated in identifying and characterizing multiple disorders, such as cancer, tissue injuries, acute or chronic inflammations or fibrotic diseases.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
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
|
Montanari MP, Tran NV, Shimmi O. Regulation of spatial distribution of BMP ligands for pattern formation. Dev Dyn 2021; 251:198-212. [PMID: 34241935 DOI: 10.1002/dvdy.397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 12/25/2022] Open
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-ß (TGF-ß) family, have been shown to contribute to embryogenesis and organogenesis during animal development. Relevant studies provide support for the following concepts: (a) BMP signals are evolutionarily highly conserved as a genetic toolkit; (b) spatiotemporal distributions of BMP signals are precisely controlled at the post-translational level; and (c) the BMP signaling network has been co-opted to adapt to diversified animal development. These concepts originated from the historical findings of the Spemann-Mangold organizer and the subsequent studies about how this organizer functions at the molecular level. In this Commentary, we focus on two topics. First, we review how the BMP morphogen gradient is formed to sustain larval wing imaginal disc and early embryo growth and patterning in Drosophila. Second, we discuss how BMP signal is tightly controlled in a context-dependent manner, and how the signal and tissue dynamics are coupled to facilitate complex tissue structure formation. Finally, we argue how these concepts might be developed in the future for further understanding the significance of BMP signaling in animal development.
Collapse
Affiliation(s)
| | - Ngan Vi Tran
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Osamu Shimmi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| |
Collapse
|
17
|
Athilingam T, Tiwari P, Toyama Y, Saunders TE. Mechanics of epidermal morphogenesis in the Drosophila pupa. Semin Cell Dev Biol 2021; 120:171-180. [PMID: 34167884 DOI: 10.1016/j.semcdb.2021.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
Adult epidermal development in Drosophila showcases a striking balance between en masse spreading of the developing adult precursor tissues and retraction of the degenerating larval epidermis. The adult precursor tissues, driven by both intrinsic plasticity and extrinsic mechanical cues, shape the segments of the adult epidermis and appendages. Here, we review the tissue architectural changes that occur during epidermal morphogenesis in the Drosophila pupa, with a particular emphasis on the underlying mechanical principles. We highlight recent developments in our understanding of adult epidermal morphogenesis. We further discuss the forces that drive these morphogenetic events and finally outline open questions and challenges.
Collapse
Affiliation(s)
| | - Prabhat Tiwari
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Yusuke Toyama
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Science, National University of Singapore, Singapore
| | - Timothy E Saunders
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Science, National University of Singapore, Singapore; Institute of Molecular Biology, A⁎Star, Singapore; Warwick Medical School, The University of Warwick, Coventry, United Kingdom.
| |
Collapse
|
18
|
Abstract
AbstractAn important goal in the fight against cancer is to understand how tumors become invasive and metastatic. A crucial early step in metastasis is thought to be the epithelial mesenchymal transition (EMT), the process in which epithelial cells transition into a more migratory and invasive, mesenchymal state. Since the genetic regulatory networks driving EMT in tumors derive from those used in development, analysis of EMTs in genetic model organisms such as the vinegar fly, Drosophila melanogaster, can provide great insight into cancer. In this review I highlight the many ways in which studies in the fly are shedding light on cancer metastasis. The review covers both normal developmental events in which epithelial cells become migratory, as well as induced events, whereby normal epithelial cells become metastatic due to genetic manipulations. The ability to make such precise genetic perturbations in the context of a normal, in vivo environment, complete with a working innate immune system, is making the fly increasingly important in understanding metastasis.
Collapse
Affiliation(s)
- Michael J. Murray
- School of BioSciences, Faculty of Science, University of Melbourne, Victoria 3010, Melbourne, Australia
| |
Collapse
|
19
|
Abstract
Over 50 years after its discovery in early chick embryos, the concept of epithelial-mesenchymal transition (EMT) is now widely applied to morphogenetic studies in both physiological and pathological contexts. Indeed, the EMT field has witnessed exponential growth in recent years, driven primarily by a rapid expansion of cancer-oriented EMT research. This has led to EMT-based therapeutic interventions that bear the prospect of fighting cancer, and has given developmental biologists new impetus to investigate EMT phenomena more closely and to find suitable models to address emerging EMT-related questions. Here, and in the accompanying poster, I provide a brief summary of the current status of EMT research and give an overview of EMT models that have been used in developmental studies. I also highlight dynamic epithelialization and de-epithelialization events that are involved in many developmental processes and that should be considered to provide a broader perspective of EMT. Finally, I put forward a set of criteria to separate morphogenetic phenomena that are EMT-related from those that are not.
Collapse
Affiliation(s)
- Guojun Sheng
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| |
Collapse
|
20
|
DaCrema D, Bhandari R, Karanja F, Yano R, Halme A. Ecdysone regulates the Drosophila imaginal disc epithelial barrier, determining the length of regeneration checkpoint delay. Development 2021; 148:dev.195057. [PMID: 33658221 DOI: 10.1242/dev.195057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 02/15/2021] [Indexed: 02/06/2023]
Abstract
Regeneration of Drosophila imaginal discs, larval precursors to adult tissues, activates a regeneration checkpoint that coordinates regenerative growth with developmental progression. This regeneration checkpoint results from the release of the relaxin-family peptide Dilp8 from regenerating imaginal tissues. Secreted Dilp8 protein is detected within the imaginal disc lumen, in which it is separated from its receptor target Lgr3, which is expressed in the brain and prothoracic gland, by the disc epithelial barrier. Here, we demonstrate that following damage the imaginal disc epithelial barrier limits Dilp8 signaling and the duration of regeneration checkpoint delay. We also find that the barrier becomes increasingly impermeable to the transepithelial diffusion of labeled dextran during the second half of the third instar. This change in barrier permeability is driven by the steroid hormone ecdysone and correlates with changes in localization of Coracle, a component of the septate junctions that is required for the late-larval impermeable epithelial barrier. Based on these observations, we propose that the imaginal disc epithelial barrier regulates the duration of the regenerative checkpoint, providing a mechanism by which tissue function can signal the completion of regeneration.
Collapse
Affiliation(s)
- Danielle DaCrema
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22902, USA
| | - Rajan Bhandari
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22902, USA
| | - Faith Karanja
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22902, USA
| | - Ryunosuke Yano
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22902, USA
| | - Adrian Halme
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22902, USA
| |
Collapse
|
21
|
Chaturvedi V, Murray MJ. Netrins: Evolutionarily Conserved Regulators of Epithelial Fusion and Closure in Development and Wound Healing. Cells Tissues Organs 2021; 211:193-211. [PMID: 33691313 DOI: 10.1159/000513880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/18/2020] [Indexed: 11/19/2022] Open
Abstract
Epithelial remodelling plays a crucial role during development. The ability of epithelial sheets to temporarily lose their integrity as they fuse with other epithelial sheets underpins events such as the closure of the neural tube and palate. During fusion, epithelial cells undergo some degree of epithelial-mesenchymal transition (EMT), whereby cells from opposing sheets dissolve existing cell-cell junctions, degrade the basement membrane, extend motile processes to contact each other, and then re-establish cell-cell junctions as they fuse. Similar events occur when an epithelium is wounded. Cells at the edge of the wound undergo a partial EMT and migrate towards each other to close the gap. In this review, we highlight the emerging role of Netrins in these processes, and provide insights into the possible signalling pathways involved. Netrins are secreted, laminin-like proteins that are evolutionarily conserved throughout the animal kingdom. Although best known as axonal chemotropic guidance molecules, Netrins also regulate epithelial cells. For example, Netrins regulate branching morphogenesis of the lung and mammary gland, and promote EMT during Drosophila wing eversion. Netrins also control epithelial fusion during optic fissure closure and inner ear formation, and are strongly implicated in neural tube closure and secondary palate closure. Netrins are also upregulated in response to organ damage and epithelial wounding, and can protect against ischemia-reperfusion injury and speed wound healing in cornea and skin. Since Netrins also have immunomodulatory properties, and can promote angiogenesis and re-innervation, they hold great promise as potential factors in future wound healing therapies.
Collapse
Affiliation(s)
- Vishal Chaturvedi
- School of BioSciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael J Murray
- School of BioSciences, University of Melbourne, Melbourne, Victoria, Australia,
| |
Collapse
|
22
|
Thompson BJ. From genes to shape during metamorphosis: a history. CURRENT OPINION IN INSECT SCIENCE 2021; 43:1-10. [PMID: 32898719 DOI: 10.1016/j.cois.2020.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
Metamorphosis (Greek for a state of transcending-form or change-in-shape) refers to a dramatic transformation of an animal's body structure that occurs after development of the embryo or larva in many species. The development of a fly (or butterfly) from a crawling larva (or caterpillar) that forms a pupa (or chrysalis) before eclosing as a flying adult is a classic example of metamorphosis that captures the imagination and has been immortalized in children's books. Powerful genetic experiments in the fruit fly Drosophila melanogaster have revealed how genes can instruct the behaviour of individual cells to control patterns of tissue growth, mechanical force, cell-cell adhesion and cell-matrix adhesion drive morphogenetic change in epithelial tissues. Together, the distribution of mass, force and resistance determines cell shape changes, cell-cell rearrangements, and/or the orientation of cell divisions to generate the final form of the tissue. In organising tissue shape, genes harness the power of self-organisation to determine the collective behaviour of molecules and cells, which can often be reproduced in computer simulations of cell polarity and/or tissue mechanics. This review highlights fundamental discoveries in epithelial morphogenesis made by pioneers who were fascinated by metamorphosis, including D'Arcy Thompson, Conrad Waddington, Dianne Fristrom and Antonio Garcia-Bellido.
Collapse
Affiliation(s)
- Barry J Thompson
- John Curtin School of Medical Research, The Australian National University, 131 Garran Rd, Acton, Canberra, Australian Capital Territory (ACT), 2601, Australia.
| |
Collapse
|
23
|
Golenkina S, Manhire-Heath R, Murray MJ. Exploiting Drosophila melanogaster Wing Imaginal Disc Eversion to Screen for New EMT Effectors. Methods Mol Biol 2021; 2179:115-134. [PMID: 32939717 DOI: 10.1007/978-1-0716-0779-4_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the early stages of Drosophila melanogaster (Drosophila) metamorphosis, a partial epithelial-mesenchymal transition (pEMT) takes place in the peripodial epithelium of wing imaginal discs. Blocking this pEMT results in adults with internalized wings and missing thoracic tissue. Using peripodial GAL4 drivers, GAL80ts temporal control, and UAS RNAi transgenes, one can use these phenotypes to screen for genes involved in the pEMT. Dominant modifier tests can then be employed to identify genetic enhancers and suppressors. To analyze a gene's role in the pEMT, one can then visualize peripodial cells in vivo at the time of eversion within the pupal case using live markers, and by dissecting, fixing, and immunostaining the prepupae. Alternatively, one can analyze the pEMT ex vivo by dissecting out wing discs and culturing them in the presence of ecdysone to induce eversion. This can provide a clearer view of the cellular processes involved and permit drug treatments to be easily applied.
Collapse
Affiliation(s)
- Sofia Golenkina
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | | | - Michael J Murray
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
24
|
Maintenance of Cell Fate by the Polycomb Group Gene Sex Combs Extra Enables a Partial Epithelial Mesenchymal Transition in Drosophila. G3-GENES GENOMES GENETICS 2020; 10:4459-4471. [PMID: 33051260 PMCID: PMC7718746 DOI: 10.1534/g3.120.401785] [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] [Indexed: 12/02/2022]
Abstract
Epigenetic silencing by Polycomb group (PcG) complexes can promote epithelial-mesenchymal transition (EMT) and stemness and is associated with malignancy of solid cancers. Here we report a role for Drosophila PcG repression in a partial EMT event that occurs during wing disc eversion, an early event during metamorphosis. In a screen for genes required for eversion we identified the PcG genes Sex combs extra (Sce) and Sex combs midleg (Scm). Depletion of Sce or Scm resulted in internalized wings and thoracic clefts, and loss of Sce inhibited the EMT of the peripodial epithelium and basement membrane breakdown, ex vivo. Targeted DamID (TaDa) using Dam-Pol II showed that Sce knockdown caused a genomic transcriptional response consistent with a shift toward a more stable epithelial fate. Surprisingly only 17 genes were significantly upregulated in Sce-depleted cells, including Abd-B, abd-A, caudal, and nubbin. Each of these loci were enriched for Dam-Pc binding. Of the four genes, only Abd-B was robustly upregulated in cells lacking Sce expression. RNAi knockdown of all four genes could partly suppress the Sce RNAi eversion phenotype, though Abd-B had the strongest effect. Our results suggest that in the absence of continued PcG repression peripodial cells express genes such as Abd-B, which promote epithelial state and thereby disrupt eversion. Our results emphasize the important role that PcG suppression can play in maintaining cell states required for morphogenetic events throughout development and suggest that PcG repression of Hox genes may affect epithelial traits that could contribute to metastasis.
Collapse
|
25
|
Pastor-Pareja JC. Atypical basement membranes and basement membrane diversity - what is normal anyway? J Cell Sci 2020; 133:133/8/jcs241794. [PMID: 32317312 DOI: 10.1242/jcs.241794] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The evolution of basement membranes (BMs) played an essential role in the organization of animal cells into tissues and diversification of body plans. The archetypal BM is a compact extracellular matrix polymer containing laminin, nidogen, collagen IV and perlecan (LNCP matrix) tightly packed into a homogenously thin planar layer. Contrasting this clear-cut morphological and compositional definition, there are numerous examples of LNCP matrices with unusual characteristics that deviate from this planar organization. Furthermore, BM components are found in non-planar matrices that are difficult to categorize as BMs at all. In this Review, I discuss examples of atypical BM organization. First, I highlight atypical BM structures in human tissues before describing the functional dissection of a plethora of BMs and BM-related structures in their tissue contexts in the fruit fly Drosophila melanogaster To conclude, I summarize our incipient understanding of the mechanisms that provide morphological, compositional and functional diversity to BMs. It is becoming increasingly clear that atypical BMs are quite prevalent, and that even typical planar BMs harbor a lot of diversity that we do not yet comprehend.
Collapse
Affiliation(s)
- José C Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing 100084, China .,Peking-Tsinghua Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
26
|
La Marca JE, Richardson HE. Two-Faced: Roles of JNK Signalling During Tumourigenesis in the Drosophila Model. Front Cell Dev Biol 2020; 8:42. [PMID: 32117973 PMCID: PMC7012784 DOI: 10.3389/fcell.2020.00042] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/17/2020] [Indexed: 12/27/2022] Open
Abstract
The highly conserved c-Jun N-terminal Kinase (JNK) signalling pathway has many functions, regulating a diversity of processes: from cell movement during embryogenesis to the stress response of cells after environmental insults. Studies modelling cancer using the vinegar fly, Drosophila melanogaster, have identified both pro- and anti-tumourigenic roles for JNK signalling, depending on context. As a tumour suppressor, JNK signalling commonly is activated by conserved Tumour Necrosis Factor (TNF) signalling, which promotes the caspase-mediated death of tumourigenic cells. JNK pathway activation can also occur via actin cytoskeleton alterations, and after cellular damage inflicted by reactive oxygen species (ROS). Additionally, JNK signalling frequently acts in concert with Salvador-Warts-Hippo (SWH) signalling – either upstream of or parallel to this potent growth-suppressing pathway. As a tumour promoter, JNK signalling is co-opted by cells expressing activated Ras-MAPK signalling (among other pathways), and used to drive cell morphological changes, induce invasive behaviours, block differentiation, and enable persistent cell proliferation. Furthermore, JNK is capable of non-autonomous influences within tumour microenvironments by effecting the transcription of various cell growth- and proliferation-promoting molecules. In this review, we discuss these aspects of JNK signalling in Drosophila tumourigenesis models, and highlight recent publications that have expanded our knowledge of this important and versatile pathway.
Collapse
Affiliation(s)
- John E La Marca
- Richardson Laboratory, Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Helena E Richardson
- Richardson Laboratory, Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Role of Notch Signaling in Leg Development in Drosophila melanogaster. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:103-127. [PMID: 32060874 DOI: 10.1007/978-3-030-34436-8_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Notch pathway plays diverse and fundamental roles during animal development. One of the most relevant, which arises directly from its unique mode of activation, is the specification of cell fates and tissue boundaries. The development of the leg of Drosophila melanogaster is a fine example of this Notch function, as it is required to specify the fate of the cells that will eventually form the leg joints, the flexible structures that separate the different segments of the adult leg. Notch activity is accurately activated and maintained at the distal end of each segment in response to the proximo-distal patterning gene network of the developing leg. Region-specific downstream targets of Notch in turn regulate the formation of the different types of joints. We discuss recent findings that shed light on the molecular and cellular mechanisms that are ultimately governed by Notch to achieve epithelial fold and joint morphogenesis. Finally, we briefly summarize the role that Notch plays in inducing the nonautonomous growth of the leg. Overall, this book chapter aims to highlight leg development as a useful model to study how patterning information is translated into specific cell behaviors that shape the final form of an adult organ.
Collapse
|
28
|
Kumar A, Rizvi MS, Athilingam T, Parihar SS, Sinha P. Heterophilic cell-cell adhesion of atypical cadherins Fat and Dachsous regulate epithelial cell size dynamics during Drosophila thorax morphogenesis. Mol Biol Cell 2019; 31:546-560. [PMID: 31877063 PMCID: PMC7202070 DOI: 10.1091/mbc.e19-08-0468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Spatiotemporal changes in epithelial cell sizes-or epithelial cell size dynamics (ECD)-during morphogenesis entail interplay between two opposing forces: cell contraction via actomyosin cytoskeleton and cell expansion via cell-cell adhesion. Cell-cell adhesion-based ECD, however, has not yet been clearly demonstrated. For instance, changing levels of homophilic E-cadherin-based cell-cell adhesion induce cell sorting, but not ECD. Here we show that cell-expansive forces of heterophilic cell-cell adhesion regulate ECD: higher cell-cell adhesion results in cell size enlargement. Thus, ECD during morphogenesis in the heminotal epithelia of Drosophila pupae leading to thorax closure corresponds with spatiotemporal gradients of two heterophilic atypical cadherins-Fat (Ft) and Dachsous (Ds)-and the levels of Ft-Ds heterodimers formed concomitantly. Our mathematical modeling and genetic tests validate this mechanism of dynamic heterophilic cell-cell adhesion-based regulation of ECD. Conservation of these atypical cadherins suggests a wider prevalence of heterophilic cell-cell adhesion-based ECD regulation during animal morphogenesis.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Mohd Suhail Rizvi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Thamarailingam Athilingam
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Saurabh Singh Parihar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Pradip Sinha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
29
|
Held LI, Sessions SK. Reflections on Bateson's rule: Solving an old riddle about why extra legs are mirror‐symmetric. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2019; 332:219-237. [DOI: 10.1002/jez.b.22910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/18/2019] [Accepted: 09/26/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Lewis I. Held
- Department of Biological SciencesTexas Tech University Lubbock Texas
| | | |
Collapse
|
30
|
Parvy JP, Yu Y, Dostalova A, Kondo S, Kurjan A, Bulet P, Lemaître B, Vidal M, Cordero JB. The antimicrobial peptide defensin cooperates with tumour necrosis factor to drive tumour cell death in Drosophila. eLife 2019; 8:45061. [PMID: 31358113 PMCID: PMC6667213 DOI: 10.7554/elife.45061] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/26/2019] [Indexed: 12/28/2022] Open
Abstract
Antimicrobial peptides (AMPs) are small cationic molecules best known as mediators of the innate defence against microbial infection. While in vitro and ex vivo evidence suggest AMPs’ capacity to kill cancer cells, in vivo demonstration of an anti-tumour role of endogenous AMPs is lacking. Using a Drosophila model of tumourigenesis, we demonstrate a role for the AMP Defensin in the control of tumour progression. Our results reveal that Tumour Necrosis Factor mediates exposure of phosphatidylserine (PS), which makes tumour cells selectively sensitive to the action of Defensin remotely secreted from tracheal and fat tissues. Defensin binds tumour cells in PS-enriched areas, provoking cell death and tumour regression. Altogether, our results provide the first in vivo demonstration for a role of an endogenous AMP as an anti-cancer agent, as well as a mechanism that explains tumour cell sensitivity to the action of AMPs. Animals have a natural defence system – the immune system – that is needed to fight off disease-causing microbes, known as pathogens. One way the immune system attacks pathogens is by producing small microbe-killing molecules called antimicrobial peptides. These antimicrobial peptides carry a positive charge, which allows them to interact with and disrupt the negatively charged cell surfaces of many microbes. Healthy animal cells do not have these negatively charged components on their cell surface, which means they are invisible to antimicrobial peptides. Studies have reported that antimicrobial peptides can attack cancer cells grown in a dish. However, it was unclear whether they could fight cancer cells in a live animal. Parvy et al. have now addressed this issue by studying tumours in the larvae of fruit flies. Flies with tumours made an antimicrobial peptide called Defensin, which normally helps to fight infections. When Parvy et al. deleted the gene coding for Defensin, less tumour cells were dying and the tumours became bigger. This result indicated that Defensin was protecting the fruit flies from tumours. Examining the tumours under the microscope showed that Defensin protein interacted with the membranes of tumour cells. Defensin was not, however, interacting with healthy cells. Further analysis revealed that a negatively charged component of cell membranes called phosphatidylserine, which normally faces the inside of healthy cells, is exposed to the outer surface of tumour cells. This negatively charged molecule renders cancer cells visible to positively charged Defensin. Importantly, the exposure of the phosphatidylserine is mediated by the fly equivalent of a protein called Tumour Necrosis Factor, a key player in cancer biology. Defensin binding to tumour cells leads to their death. These experiments in the fruit fly highlight key molecular mechanisms that allow antimicrobial peptides to fight cancer cells in a living organism. Because human tumour cells can also expose phosphatidylserine, these latest findings may open up the possibility of a new kind of anti-cancer therapy for patients.
Collapse
Affiliation(s)
| | - Yachuan Yu
- CRUK Beatson Institute, Glasgow, United Kingdom
| | - Anna Dostalova
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Shu Kondo
- Invertebrate Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima, Japan
| | - Alina Kurjan
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Philippe Bulet
- Institute for Advanced Biosciences, CR University Grenoble Alpes, Inserm U1209, CNRS UMR5309, Immunologie Analytique des Pathologies Chroniques, Grenoble, France
| | - Bruno Lemaître
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | | | - Julia B Cordero
- CRUK Beatson Institute, Glasgow, United Kingdom.,Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
31
|
Li P, Ma Z, Yu Y, Hu X, Zhou Y, Song H. FER promotes cell migration via regulating JNK activity. Cell Prolif 2019; 52:e12656. [PMID: 31264309 PMCID: PMC6797522 DOI: 10.1111/cpr.12656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
Objectives Cell migration has a key role in cancer metastasis, which contributes to drug resistance and tumour recurrence. Better understanding of the mechanisms involved in this process will potentially reveal new drug targets for cancer therapy. Fer is a non‐receptor protein tyrosine kinase aberrantly expressed in various human cancers, whereas its role in tumour progression remains elusive. Materials and Methods Transgenic flies and epigenetic analysis were employed to investigate the role of Drosophila Fer (FER) in cell migration and underlying mechanisms. Co‐immunoprecipitation assay was used to monitor the interaction between FER and Drosophila JNK (Bsk). The conservation of Fer in regulating JNK signalling was explored in mammalian cancer and non‐cancer cells. Results Overexpression of FER triggered cell migration and activated JNK signalling in the Drosophila wing disc. Upregulation and downregulation in the basal activity of Bsk exacerbated and eliminated FER‐mediated migration, respectively. In addition, loss of FER blocked signal transduction of the JNK pathway. Specifically, FER interacted with and promoted the activity of Bsk, which required both the kinase domain and the C‐terminal of Bsk. Lastly, Fer regulated JNK activities in mammalian cells. Conclusions Our study reveals FER as a positive regulator of JNK‐mediated cell migration and suggests its potential role as a therapeutic target for cancer metastasis.
Collapse
Affiliation(s)
- Ping Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhiwei Ma
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yun Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xingjie Hu
- School of Public Health, Guangzhou Medical University, Guangdong, China
| | - Yanfeng Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyun Song
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
32
|
Proag A, Monier B, Suzanne M. Physical and functional cell-matrix uncoupling in a developing tissue under tension. Development 2019; 146:dev.172577. [PMID: 31064785 PMCID: PMC6589077 DOI: 10.1242/dev.172577] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 04/26/2019] [Indexed: 01/02/2023]
Abstract
Tissue mechanics play a crucial role in organ development. They rely on the properties of cells and the extracellular matrix (ECM), but the relative physical contribution of cells and ECM to morphogenesis is poorly understood. Here, we have analyzed the behavior of the peripodial epithelium (PE) of the Drosophila leg disc in the light of the dynamics of its cellular and ECM components. The PE undergoes successive changes during leg development, including elongation, opening and removal to free the leg. During elongation, we found that the ECM and cell layer are progressively uncoupled. Concomitantly, the tension, mainly borne by the ECM at first, builds up in the cell monolayer. Then, each layer of the peripodial epithelium is removed by an independent mechanism: while the ECM layer withdraws following local proteolysis, cellular monolayer withdrawal is independent of ECM degradation and is driven by myosin II-dependent contraction. These results reveal a surprising physical and functional cell-matrix uncoupling in a monolayer epithelium under tension during development. This article has an associated ‘The people behind the papers’ interview. Highlighted Article: The independent behavior of extracellular matrix and cell monolayer of an epithelium under tension during Drosophila leg development highlights the interplay between tissue mechanics and cell-matrix coupling.
Collapse
Affiliation(s)
- Amsha Proag
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Bruno Monier
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| | - Magali Suzanne
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31062, France
| |
Collapse
|
33
|
Coupling between dynamic 3D tissue architecture and BMP morphogen signaling during Drosophila wing morphogenesis. Proc Natl Acad Sci U S A 2019; 116:4352-4361. [PMID: 30760594 PMCID: PMC6410814 DOI: 10.1073/pnas.1815427116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tissue morphogenesis is a dynamic process often accompanied by cell patterning and differentiation. Although how conserved growth factor signaling affects cell and tissue shapes has been actively studied, much less is known about how signaling and dynamic morphogenesis are mutually coordinated. Our study shows that BMP signaling and 3D morphogenesis of the Drosophila pupal wing are tightly coupled. These findings are highlighted by the fact that the directionality of BMP signal is changed from lateral planar during the inflation stage to interplanar after re-apposition of the dorsal and ventral wing epithelia. We suspect that the dynamic interplay between planar and interplanar signaling linked to tissue shape changes is likely to be used across species in many developing organs. At the level of organ formation, tissue morphogenesis drives developmental processes in animals, often involving the rearrangement of two-dimensional (2D) structures into more complex three-dimensional (3D) tissues. These processes can be directed by growth factor signaling pathways. However, little is known about how such morphological changes affect the spatiotemporal distribution of growth factor signaling. Here, using the Drosophila pupal wing, we address how decapentaplegic (Dpp)/bone morphogenetic protein (BMP) signaling and 3D wing morphogenesis are coordinated. Dpp, expressed in the longitudinal veins (LVs) of the pupal wing, initially diffuses laterally within both dorsal and ventral wing epithelia during the inflation stage to regulate cell proliferation. Dpp localization is then refined to the LVs within each epithelial plane, but with active interplanar signaling for vein patterning/differentiation, as the two epithelia appose. Our data further suggest that the 3D architecture of the wing epithelia and the spatial distribution of BMP signaling are tightly coupled, revealing that 3D morphogenesis is an emergent property of the interactions between extracellular signaling and tissue shape changes.
Collapse
|
34
|
Cosolo A, Jaiswal J, Csordás G, Grass I, Uhlirova M, Classen AK. JNK-dependent cell cycle stalling in G2 promotes survival and senescence-like phenotypes in tissue stress. eLife 2019; 8:41036. [PMID: 30735120 PMCID: PMC6389326 DOI: 10.7554/elife.41036] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/06/2019] [Indexed: 01/10/2023] Open
Abstract
The restoration of homeostasis after tissue damage relies on proper spatial-temporal control of damage-induced apoptosis and compensatory proliferation. In Drosophila imaginal discs these processes are coordinated by the stress response pathway JNK. We demonstrate that JNK signaling induces a dose-dependent extension of G2 in tissue damage and tumors, resulting in either transient stalling or a prolonged but reversible cell cycle arrest. G2-stalling is mediated by downregulation of the G2/M-specific phosphatase String(Stg)/Cdc25. Ectopic expression of stg is sufficient to suppress G2-stalling and reveals roles for stalling in survival, proliferation and paracrine signaling. G2-stalling protects cells from JNK-induced apoptosis, but under chronic conditions, reduces proliferative potential of JNK-signaling cells while promoting non-autonomous proliferation. Thus, transient cell cycle stalling in G2 has key roles in wound healing but becomes detrimental upon chronic JNK overstimulation, with important implications for chronic wound healing pathologies or tumorigenic transformation.
Collapse
Affiliation(s)
- Andrea Cosolo
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany.,Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Janhvi Jaiswal
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Gábor Csordás
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Isabelle Grass
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany.,Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Mirka Uhlirova
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Anne-Kathrin Classen
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany.,Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| |
Collapse
|
35
|
Boix-Fabrés J, Karkali K, Martín-Blanco E, Rebollo E. Automated Macro Approach to Remove Vitelline Membrane Autofluorescence in Drosophila Embryo 4D Movies. Methods Mol Biol 2019; 2040:155-175. [PMID: 31432480 DOI: 10.1007/978-1-4939-9686-5_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This chapter provides an ImageJ/Fiji automated macro approach to remove the vitelline membrane autofluorescence in live Drosophila embryo movies acquired in a 4D (3D plus time) fashion. The procedure consists in a segmentation pipeline that can cope with different relative intensities of the vitelline membrane autofluorescence, followed by a developed algorithm that adjusts the extracted outline selection to the shape deformations that naturally occur during Drosophila embryo development. Finally, the fitted selection is used to clear the external glowing halo that, otherwise, would obscure the visualization of the internal embryo labeling upon projection or 3D rendering.
Collapse
Affiliation(s)
- Jaume Boix-Fabrés
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona, Spanish National Research Council (CSIC), Barcelona, Spain
| | - Katerina Karkali
- Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | | | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (CSIC), Barcelona, Spain.
| |
Collapse
|
36
|
Keeley DP, Sherwood DR. Tissue linkage through adjoining basement membranes: The long and the short term of it. Matrix Biol 2019; 75-76:58-71. [PMID: 29803937 PMCID: PMC6252152 DOI: 10.1016/j.matbio.2018.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 01/01/2023]
Abstract
Basement membranes (BMs) are thin dense sheets of extracellular matrix that surround most tissues. When the BMs of neighboring tissues come into contact, they usually slide along one another and act to separate tissues and organs into distinct compartments. However, in certain specialized regions, the BMs of neighboring tissues link, helping to bring tissues together. These BM connections can be transient, such as during tissue fusion events in development, or long-term, as with adult tissues involved with filtration, including the blood brain barrier and kidney glomerulus. The transitory nature of these connections in development and the complexity of tissue filtration systems in adults have hindered the understanding of how juxtaposed BMs fasten together. The recent identification of a BM-BM adhesion system in C. elegans, termed B-LINK (BM linkage), however, is revealing cellular and extracellular matrix components of a nascent tissue adhesion system. We discuss insights gained from studying the B-LINK tissue adhesion system in C. elegans, compare this adhesion with other BM-BM connections in Drosophila and vertebrates, and outline important future directions towards elucidating this fascinating and poorly understood mode of adhesion that joins neighboring tissues.
Collapse
Affiliation(s)
- Daniel P Keeley
- Department of Biology, Regeneration Next, Duke University, Box 90338, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
37
|
Andjelković A, Mordas A, Bruinsma L, Ketola A, Cannino G, Giordano L, Dhandapani PK, Szibor M, Dufour E, Jacobs HT. Expression of the Alternative Oxidase Influences Jun N-Terminal Kinase Signaling and Cell Migration. Mol Cell Biol 2018; 38:e00110-18. [PMID: 30224521 PMCID: PMC6275184 DOI: 10.1128/mcb.00110-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/11/2018] [Accepted: 09/11/2018] [Indexed: 12/25/2022] Open
Abstract
Downregulation of Jun N-terminal kinase (JNK) signaling inhibits cell migration in diverse model systems. In Drosophila pupal development, attenuated JNK signaling in the thoracic dorsal epithelium leads to defective midline closure, resulting in cleft thorax. Here we report that concomitant expression of the Ciona intestinalis alternative oxidase (AOX) was able to compensate for JNK pathway downregulation, substantially correcting the cleft thorax phenotype. AOX expression also promoted wound-healing behavior and single-cell migration in immortalized mouse embryonic fibroblasts (iMEFs), counteracting the effect of JNK pathway inhibition. However, AOX was not able to rescue developmental phenotypes resulting from knockdown of the AP-1 transcription factor, the canonical target of JNK, nor its targets and had no effect on AP-1-dependent transcription. The migration of AOX-expressing iMEFs in the wound-healing assay was differentially stimulated by antimycin A, which redirects respiratory electron flow through AOX, altering the balance between mitochondrial ATP and heat production. Since other treatments affecting mitochondrial ATP did not stimulate wound healing, we propose increased mitochondrial heat production as the most likely primary mechanism of action of AOX in promoting cell migration in these various contexts.
Collapse
Affiliation(s)
- Ana Andjelković
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Amelia Mordas
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Lyon Bruinsma
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Annika Ketola
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Giuseppe Cannino
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Luca Giordano
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Praveen K Dhandapani
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Marten Szibor
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eric Dufour
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Howard T Jacobs
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- BioMediTech Institute, University of Tampere, Tampere, Finland
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
38
|
Kosakamoto H, Fujisawa Y, Obata F, Miura M. High expression of A-type lamin in the leading front is required for Drosophila thorax closure. Biochem Biophys Res Commun 2018; 499:209-214. [PMID: 29559239 DOI: 10.1016/j.bbrc.2018.03.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/16/2018] [Indexed: 10/17/2022]
Abstract
Tissue closure involves the coordinated unidirectional movement of a group of cells without loss of cell-cell contact. However, the molecular mechanisms controlling the tissue closure are not fully understood. Here, we demonstrate that Lamin C, the sole A-type lamin in Drosophila, contributes to the process of thorax closure in pupa. High expression of Lamin C was observed at the leading front of the migrating wing imaginal discs. Live imaging analysis revealed that knockdown of Lamin C in the thorax region affected the coordinated movement of the leading front, resulting in incomplete tissue fusion required for formation of the adult thorax. The closure defect due to knockdown of Lamin C correlated with insufficient accumulation of F-actin at the front. Our study indicates a link between A-type lamin and the cell migration behavior during tissue closure.
Collapse
Affiliation(s)
- Hina Kosakamoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Yuya Fujisawa
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Fumiaki Obata
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan.
| |
Collapse
|
39
|
Zhai Z, Boquete JP, Lemaitre B. Cell-Specific Imd-NF-κB Responses Enable Simultaneous Antibacterial Immunity and Intestinal Epithelial Cell Shedding upon Bacterial Infection. Immunity 2018; 48:897-910.e7. [DOI: 10.1016/j.immuni.2018.04.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/31/2017] [Accepted: 04/10/2018] [Indexed: 12/13/2022]
|
40
|
Calleja M, Morata G, Casanova J. Tumorigenic Properties of Drosophila Epithelial Cells Mutant for lethal giant larvae. Dev Dyn 2018; 245:834-43. [PMID: 27239786 DOI: 10.1002/dvdy.24420] [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: 03/30/2015] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mutations in Drosophila tumor suppressor genes (TSGs) lead to the formation of invasive tumors in the brain and imaginal discs. RESULTS Here we studied the tumorigenic properties of imaginal discs mutant for the TSG gene lethal giant larvae (lgl). lgl mutant cells display the characteristic features of mammalian tumor cells: they can proliferate indefinitely, induce additional tracheogenesis (an insect counterpart of vasculogenesis) and invade neighboring tissues. Lgl mutant tissues exhibit high apoptotic levels, which lead to the activation of the Jun-N-Terminal Kinase (JNK) pathway. We propose that JNK is a key factor in the acquisition of these tumorigenic properties; it promotes cell proliferation and induces high levels of Mmp1 and confers tumor cells capacity to invade wild-type tissue. Noteworthy, lgl RNAi-mediated down-regulation does not produce similar transformations in the central nervous system (CNS), thereby indicating a fundamental difference between the cells of developing imaginal discs and those of differentiated organs. We discuss these results in the light of the "single big-hit origin" of some human pediatric or developmental cancers. CONCLUSIONS Down-regulation of lgl in imaginal discs is sufficient to enhance tracheogenesis and to promote invasion and colonization of other larval structures including the CNS. Developmental Dynamics 245:834-843, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Ginés Morata
- Centro de Biología Molecular, CSIC-UAM, Madrid, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC) Barcelona, Catalonia, Spain.,Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| |
Collapse
|
41
|
Medwig TN, Matus DQ. Breaking down barriers: the evolution of cell invasion. Curr Opin Genet Dev 2017; 47:33-40. [PMID: 28881331 PMCID: PMC5716887 DOI: 10.1016/j.gde.2017.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/02/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022]
Abstract
Cell invasion is a specialized cell behavior that likely co-evolved with the emergence of basement membranes in metazoans as a mechanism to break down the barriers that separate tissues. A variety of conserved and lineage-specific biological processes that occur during development and homeostasis rely on cell invasive behavior. Recent innovations in genome editing and live-cell imaging have shed some light on the programs that mediate acquisition of an invasive phenotype; however, comparative approaches among species are necessary to understand how this cell behavior evolved. Here, we discuss the contexts of cell invasion, highlighting both established and emerging model systems, and underscore gaps in our understanding of the evolution of this key cellular behavior.
Collapse
Affiliation(s)
- Taylor N Medwig
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - David Q Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA.
| |
Collapse
|
42
|
Osterfield M, Berg CA, Shvartsman SY. Epithelial Patterning, Morphogenesis, and Evolution: Drosophila Eggshell as a Model. Dev Cell 2017; 41:337-348. [PMID: 28535370 DOI: 10.1016/j.devcel.2017.02.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 02/06/2017] [Accepted: 02/24/2017] [Indexed: 11/30/2022]
Abstract
Understanding the mechanisms driving tissue and organ formation requires knowledge across scales. How do signaling pathways specify distinct tissue types? How does the patterning system control morphogenesis? How do these processes evolve? The Drosophila egg chamber, where EGF and BMP signaling intersect to specify unique cell types that construct epithelial tubes for specialized eggshell structures, has provided a tractable system to ask these questions. Work there has elucidated connections between scales of development, including across evolutionary scales, and fostered the development of quantitative modeling tools. These tools and general principles can be applied to the understanding of other developmental processes across organisms.
Collapse
Affiliation(s)
- Miriam Osterfield
- Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Celeste A Berg
- Molecular and Cellular Biology Program and Department of Genome Sciences, University of Washington, Seattle, WA 98195-5065, USA
| | - Stanislav Y Shvartsman
- Department of Chemical and Biological Engineering and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
43
|
Live-cell confocal microscopy and quantitative 4D image analysis of anchor-cell invasion through the basement membrane in Caenorhabditis elegans. Nat Protoc 2017; 12:2081-2096. [PMID: 28880279 DOI: 10.1038/nprot.2017.093] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cell invasion through basement membrane (BM) barriers is crucial in development, leukocyte trafficking and the spread of cancer. The mechanisms that direct invasion, despite their importance in normal and disease states, are poorly understood, largely because of the inability to visualize dynamic cell-BM interactions in vivo. This protocol describes multichannel time-lapse confocal imaging of anchor-cell invasion in live Caenorhabditis elegans. Methods presented include outline-slide preparation and worm growth synchronization (15 min), mounting (20 min), image acquisition (20-180 min), image processing (20 min) and quantitative analysis (variable timing). The acquired images enable direct measurement of invasive dynamics including formation of invadopodia and cell-membrane protrusions, and removal of BM. This protocol can be combined with genetic analysis, molecular-activity probes and optogenetic approaches to uncover the molecular mechanisms underlying cell invasion. These methods can also be readily adapted by any worm laboratory for real-time analysis of cell migration, BM turnover and cell-membrane dynamics.
Collapse
|
44
|
|
45
|
Campbell K, Casanova J. A common framework for EMT and collective cell migration. Development 2016; 143:4291-4300. [DOI: 10.1242/dev.139071] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During development, cells often switch between static and migratory behaviours. Such transitions are fundamental events in development and are linked to harmful consequences in pathology. It has long been considered that epithelial cells either migrate collectively as epithelial cells, or undergo an epithelial-to-mesenchymal transition and migrate as individual mesenchymal cells. Here, we assess what is currently known about in vivo cell migratory phenomena and hypothesise that such migratory behaviours do not fit into alternative and mutually exclusive categories. Rather, we propose that these categories can be viewed as the most extreme cases of a general continuum of morphological variety, with cells harbouring different degrees or combinations of epithelial and mesenchymal features and displaying an array of migratory behaviours.
Collapse
Affiliation(s)
- Kyra Campbell
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
- Institut de Recerca Biomèdica de Barcelona, C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
- Institut de Recerca Biomèdica de Barcelona, C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
| |
Collapse
|
46
|
Abstract
decapentaplegic (dpp), the Drosophila ortholog of BMP 2/4, directs ventral adult head morphogenesis through expression in the peripodial epithelium of the eye-antennal disc. This dpp expressing domain exerts effects both on the peripodial epithelium, and the underlying disc proper epithelium. We have uncovered a role for the Jun N-terminal kinase (JNK) pathway in dpp-mediated ventral head development. JNK activity is required for dpp's action on the disc proper, but in the absence of dpp expression, excessive JNK activity is produced, leading to specific loss of maxillary palps. In this review we outline our hypotheses on how dpp acts by both short range and longer range mechanisms to direct head morphogenesis and speculate on the dual role of JNK signaling in this process. Finally, we describe the regulatory control of dpp expression in the eye-antennal disc, and pose the problem of how the various expression domains of a secreted protein can be targeted to their specific functions.
Collapse
Affiliation(s)
- Deborah A Hursh
- a Division of Cell and Gene Therapies , Center for Biologics Evaluation and Research, Food and Drug Administration , Silver Spring , MD , USA
| | - Brian G Stultz
- a Division of Cell and Gene Therapies , Center for Biologics Evaluation and Research, Food and Drug Administration , Silver Spring , MD , USA
| | - Sung Yeon Park
- b Ischemic/Hypoxic Disease Institute , Department of Physiology , Seoul National University College of Medicine , Seoul , Republic of Korea
| |
Collapse
|
47
|
miR-8 modulates cytoskeletal regulators to influence cell survival and epithelial organization in Drosophila wings. Dev Biol 2016; 412:83-98. [PMID: 26902111 DOI: 10.1016/j.ydbio.2016.01.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 01/28/2016] [Accepted: 01/31/2016] [Indexed: 02/05/2023]
Abstract
The miR-200 microRNA family plays important tumor suppressive roles. The sole Drosophila miR-200 ortholog, miR-8 plays conserved roles in Wingless, Notch and Insulin signaling - pathways linked to tumorigenesis, yet homozygous null animals are viable and often appear morphologically normal. We observed that wing tissues mosaic for miR-8 levels by genetic loss or gain of function exhibited patterns of cell death consistent with a role for miR-8 in modulating cell survival in vivo. Here we show that miR-8 levels impact several actin cytoskeletal regulators that can affect cell survival and epithelial organization. We show that loss of miR-8 can confer resistance to apoptosis independent of an epithelial to mesenchymal transition while the persistence of cells expressing high levels of miR-8 in the wing epithelium leads to increased JNK signaling, aberrant expression of extracellular matrix remodeling proteins and disruption of proper wing epithelial organization. Altogether our results suggest that very low as well as very high levels of miR-8 can contribute to hallmarks associated with cancer, suggesting approaches to increase miR-200 microRNAs in cancer treatment should be moderate.
Collapse
|
48
|
Accumulation of differentiating intestinal stem cell progenies drives tumorigenesis. Nat Commun 2015; 6:10219. [PMID: 26690827 PMCID: PMC4703904 DOI: 10.1038/ncomms10219] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/13/2015] [Indexed: 12/31/2022] Open
Abstract
Stem cell self-renewal and differentiation are coordinated to maintain tissue homeostasis and prevent cancer. Mutations causing stem cell proliferation are traditionally the focus of cancer studies. However, the contribution of the differentiating stem cell progenies in tumorigenesis is poorly characterized. Here we report that loss of the SOX transcription factor, Sox21a, blocks the differentiation programme of enteroblast (EB), the intestinal stem cell progeny in the adult Drosophila midgut. This results in EB accumulation and formation of tumours. Sox21a tumour initiation and growth involve stem cell proliferation induced by the unpaired 2 mitogen released from accumulating EBs generating a feed-forward loop. EBs found in the tumours are heterogeneous and grow towards the intestinal lumen. Sox21a tumours modulate their environment by secreting matrix metalloproteinase and reactive oxygen species. Enterocytes surrounding the tumours are eliminated through delamination allowing tumour progression, a process requiring JNK activation. Our data highlight the tumorigenic properties of transit differentiating cells.
Collapse
|
49
|
Santabárbara-Ruiz P, López-Santillán M, Martínez-Rodríguez I, Binagui-Casas A, Pérez L, Milán M, Corominas M, Serras F. ROS-Induced JNK and p38 Signaling Is Required for Unpaired Cytokine Activation during Drosophila Regeneration. PLoS Genet 2015; 11:e1005595. [PMID: 26496642 PMCID: PMC4619769 DOI: 10.1371/journal.pgen.1005595] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 09/18/2015] [Indexed: 12/26/2022] Open
Abstract
Upon apoptotic stimuli, epithelial cells compensate the gaps left by dead cells by activating proliferation. This has led to the proposal that dying cells signal to surrounding living cells to maintain homeostasis. Although the nature of these signals is not clear, reactive oxygen species (ROS) could act as a signaling mechanism as they can trigger pro-inflammatory responses to protect epithelia from environmental insults. Whether ROS emerge from dead cells and what is the genetic response triggered by ROS is pivotal to understand regeneration of Drosophila imaginal discs. We genetically induced cell death in wing imaginal discs, monitored the production of ROS and analyzed the signals required for repair. We found that cell death generates a burst of ROS that propagate to the nearby surviving cells. Propagated ROS activate p38 and induce tolerable levels of JNK. The activation of JNK and p38 results in the expression of the cytokines Unpaired (Upd), which triggers the JAK/STAT signaling pathway required for regeneration. Our findings demonstrate that this ROS/JNK/p38/Upd stress responsive module restores tissue homeostasis. This module is not only activated after cell death induction but also after physical damage and reveals one of the earliest responses for imaginal disc regeneration. Regenerative biology pursues to unveil the genetic networks triggered by tissue damage. Regeneration can occur after damage by cell death or by injury. We used the imaginal disc of Drosophila in which we genetically activated apoptosis or physically removed some parts and monitored the capacity to repair the damage. We found that dying cells generate a burst of reactive oxygen species (ROS) necessary to activate JNK and p38 signaling pathways in the surrounding living cells. The action of these pathways is necessary for the activation of the cytokines Unpaired (Upd). Eventually, Upd will turn on the JAK/STAT signaling pathway to induce regenerative growth. Thus, we present here a module of signals that depends on oxidative stress and that, through the p38-JNK interplay, will activate cytokine-dependent regeneration.
Collapse
Affiliation(s)
- Paula Santabárbara-Ruiz
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Mireya López-Santillán
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Irene Martínez-Rodríguez
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Anahí Binagui-Casas
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Lídia Pérez
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Marco Milán
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain
| | - Montserrat Corominas
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Florenci Serras
- Departament de Genètica, Facultat de Biologia and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
50
|
Shen J, Lu J, Sui L, Wang D, Yin M, Hoffmann I, Legler A, Pflugfelder GO. The orthologous Tbx transcription factors Omb and TBX2 induce epithelial cell migration and extrusion in vivo without involvement of matrix metalloproteinases. Oncotarget 2015; 5:11998-2015. [PMID: 25344916 PMCID: PMC4322970 DOI: 10.18632/oncotarget.2426] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/02/2014] [Indexed: 01/06/2023] Open
Abstract
The transcription factors TBX2 and TBX3 are overexpressed in various human cancers. Here, we investigated the effect of overexpressing the orthologous Tbx genes Drosophila optomotor-blind (omb) and human TBX2 in the epithelium of the Drosophila wing imaginal disc and observed two types of cell motility. Omb/TBX2 overexpressing cells could move within the plane of the epithelium. Invasive cells migrated long-distance as single cells retaining or regaining normal cell shape and apico-basal polarity in spite of attenuated apical DE-cadherin concentration. Inappropriate levels of DE-cadherin were sufficient to drive cell migration in the wing disc epithelium. Omb/TBX2 overexpression and reduced DE-cadherin-dependent adhesion caused the formation of actin-rich lateral cell protrusions. Omb/TBX2 overexpressing cells could also delaminate basally, penetratingthe basal lamina, however, without degradation of extracellular matrix. Expression of Timp, an inhibitor of matrix metalloproteases, blocked neither intraepithelial motility nor basal extrusion. Our results reveal an MMP-independent mechanism of cell invasion and suggest a conserved role of Tbx2-related proteins in cell invasion and metastasis-related processes.
Collapse
Affiliation(s)
- Jie Shen
- Department of Entomology, China Agricultural University, Beijing, China
| | - Juan Lu
- Department of Entomology, China Agricultural University, Beijing, China
| | - Liyuan Sui
- Department of Entomology, China Agricultural University, Beijing, China
| | - Dan Wang
- Department of Entomology, China Agricultural University, Beijing, China
| | - Meizhen Yin
- Key Laboratory of Carbon Fiber and Functional Polymers, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Inka Hoffmann
- Institute of Genetics, Johannes Gutenberg-University, Mainz, Germany
| | - Anne Legler
- Institute of Genetics, Johannes Gutenberg-University, Mainz, Germany
| | | |
Collapse
|