1
|
Raval K, Jamshidi N, Seyran B, Salwinski L, Pillai R, Yang L, Ma F, Pellegrini M, Shin J, Yang X, Tudzarova S. Dysfunctional β-cell longevity in diabetes relies on energy conservation and positive epistasis. Life Sci Alliance 2024; 7:e202402743. [PMID: 39313296 PMCID: PMC11420665 DOI: 10.26508/lsa.202402743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
Long-lived PFKFB3-expressing β-cells are dysfunctional partly because of prevailing glycolysis that compromises metabolic coupling of insulin secretion. Their accumulation in type 2 diabetes (T2D) appears to be related to the loss of apoptotic competency of cell fitness competition that maintains islet function by favoring constant selection of healthy "winner" cells. To investigate how PFKFB3 can disguise the competitive traits of dysfunctional "loser" β-cells, we analyzed the overlap between human β-cells with bona fide "loser signature" across diabetes pathologies using the HPAP scRNA-seq and spatial transcriptomics of PFKFB3-positive β-cells from nPOD T2D pancreata. The overlapping transcriptional profile of "loser" β-cells was represented by down-regulated ribosomal biosynthesis and genes encoding for mitochondrial respiration. PFKFB3-positive "loser" β-cells had the reduced expression of HLA class I and II genes. Gene-gene interaction analysis revealed that PFKFB3 rs1983890 can interact with the anti-apoptotic gene MAIP1 implicating positive epistasis as a mechanism for prolonged survival of "loser" β-cells in T2D. Inhibition of PFKFB3 resulted in the clearance of dysfunctional "loser" β-cells leading to restored glucose tolerance in the mouse model of T2D.
Collapse
Affiliation(s)
- Kavit Raval
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Neema Jamshidi
- Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Berfin Seyran
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Lukasz Salwinski
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Raju Pillai
- Department of Pathology, City-of-Hope, Duarte, CA, USA
| | - Lixin Yang
- Department of Pathology, City-of-Hope, Duarte, CA, USA
| | - Feiyang Ma
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Juliana Shin
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Slavica Tudzarova
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
2
|
Fischer F, Ernst L, Frey A, Holstein K, Prasad D, Weichselberger V, Balaji R, Classen AK. A mismatch in the expression of cell surface molecules induces tissue-intrinsic defense against aberrant cells. Curr Biol 2024; 34:980-996.e6. [PMID: 38350446 DOI: 10.1016/j.cub.2024.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 09/29/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
Tissue-intrinsic error correction enables epithelial cells to detect abnormal neighboring cells and facilitate their removal from the tissue. One of these pathways, "interface surveillance," is triggered by cells with aberrant developmental and cell-fate-patterning pathways. It remains unknown which molecular mechanisms provide cells with the ability to compare fate between neighboring cells. We demonstrate that Drosophila imaginal discs express an array of cell surface molecules previously implicated in neuronal axon guidance processes. They include members of the Robo, Teneurin, Ephrin, Toll-like, or atypical cadherin families. Importantly, a mismatch in expression levels of these cell surface molecules between adjacent cells is sufficient to induce interface surveillance, indicating that differences in expression levels between neighboring cells, rather than their absolute expression levels, are crucial. Specifically, a mismatch in Robo2 and Robo3, but not Robo1, induces enrichment of actin, myosin II, and Ena/Vasp, as well as activation of JNK and apoptosis at clonal interfaces. Moreover, Robo2 can induce interface surveillance independently of its cytosolic domain and without the need for the Robo-ligand Slit. The expression of Robo2 and other cell surface molecules, such as Teneurins or the Ephrin receptor is regulated by fate-patterning pathways intrinsic and extrinsic to the wing disc, as well as by expression of oncogenic RasV12. Combined, we demonstrate that neighboring cells respond to a mismatch in surface code patterns mediated by specific transmembrane proteins and reveal a novel function for these cell surface proteins in cell fate recognition and removal of aberrant cells during development and homeostasis of epithelial tissues.
Collapse
Affiliation(s)
- Friedericke Fischer
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Laurin Ernst
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; International Max Planck Research School for Immunobiology, Epigenetics, and Metabolism, 79108 Freiburg, Germany
| | - Anna Frey
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Katrin Holstein
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Deepti Prasad
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany
| | - Vanessa Weichselberger
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Aix Marseille University, CNRS, UMR 7288, IBDM, 13288 Marseille, France
| | - Ramya Balaji
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
3
|
Mattson MP, Leak RK. The hormesis principle of neuroplasticity and neuroprotection. Cell Metab 2024; 36:315-337. [PMID: 38211591 DOI: 10.1016/j.cmet.2023.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/06/2023] [Accepted: 12/18/2023] [Indexed: 01/13/2024]
Abstract
Animals live in habitats fraught with a range of environmental challenges to their bodies and brains. Accordingly, cells and organ systems have evolved stress-responsive signaling pathways that enable them to not only withstand environmental challenges but also to prepare for future challenges and function more efficiently. These phylogenetically conserved processes are the foundation of the hormesis principle, in which single or repeated exposures to low levels of environmental challenges improve cellular and organismal fitness and raise the probability of survival. Hormetic principles have been most intensively studied in physical exercise but apply to numerous other challenges known to improve human health (e.g., intermittent fasting, cognitive stimulation, and dietary phytochemicals). Here we review the physiological mechanisms underlying hormesis-based neuroplasticity and neuroprotection. Approaching natural resilience from the lens of hormesis may reveal novel methods for optimizing brain function and lowering the burden of neurological disorders.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Matamoro-Vidal A, Cumming T, Davidović A, Levillayer F, Levayer R. Patterned apoptosis has an instructive role for local growth and tissue shape regulation in a fast-growing epithelium. Curr Biol 2024; 34:376-388.e7. [PMID: 38215743 PMCID: PMC10808510 DOI: 10.1016/j.cub.2023.12.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/13/2023] [Accepted: 12/11/2023] [Indexed: 01/14/2024]
Abstract
What regulates organ size and shape remains one fundamental mystery of modern biology. Research in this area has primarily focused on deciphering the regulation in time and space of growth and cell division, while the contribution of cell death has been overall neglected. This includes studies of the Drosophila wing, one of the best-characterized systems for the study of growth and patterning, undergoing massive growth during larval stage and important morphogenetic remodeling during pupal stage. So far, it has been assumed that cell death was relatively neglectable in this tissue both during larval stage and pupal stage, and as a result, the pattern of growth was usually attributed to the distribution of cell division. Here, using systematic mapping and registration combined with quantitative assessment of clone size and disappearance as well as live imaging, we outline a persistent pattern of cell death and clone elimination emerging in the larval wing disc and persisting during pupal wing morphogenesis. Local variation of cell death is associated with local variation of clone size, pointing to an impact of cell death on local growth that is not fully compensated by proliferation. Using morphometric analyses of adult wing shape and genetic perturbations, we provide evidence that patterned death locally and globally affects adult wing shape and size. This study describes a roadmap for precise assessment of the contribution of cell death to tissue shape and outlines an important instructive role of cell death in modulating quantitatively local growth and morphogenesis of a fast-growing tissue.
Collapse
Affiliation(s)
- Alexis Matamoro-Vidal
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France
| | - Tom Cumming
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France; PPU program Institut Pasteur, Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Anđela Davidović
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, 75015 Paris, France
| | - Florence Levillayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France.
| |
Collapse
|
5
|
Matsuda S, Affolter M. Is Drosophila Dpp/BMP morphogen spreading required for wing patterning and growth? Bioessays 2023; 45:e2200218. [PMID: 37452394 DOI: 10.1002/bies.202200218] [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: 11/09/2022] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023]
Abstract
Secreted signaling molecules act as morphogens to control patterning and growth in many developing tissues. Since locally produced morphogens spread to form a concentration gradient in the surrounding tissue, spreading is generally thought to be the key step in the non-autonomous actions. Here, we review recent advances in tool development to investigate morphogen function using the role of decapentaplegic (Dpp)/bone morphogenetic protein (BMP)-type ligand in the Drosophila wing disc as an example. By applying protein binder tools to distinguish between the roles of Dpp spreading and local Dpp signaling, we found that Dpp signaling in the source cells is important for wing patterning and growth but Dpp spreading from this source cells is not as strictly required as previously thought. Given recent studies showing unexpected requirements of long-range action of different morphogens, manipulating endogenous morphogen gradients by synthetic protein binder tools could shed more light on how morphogens act in developing tissues.
Collapse
Affiliation(s)
- Shinya Matsuda
- Growth & Development, Biozentrum, University of Basel, Basel, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
6
|
Ramezani A, Britton S, Zandi R, Alber M, Nematbakhsh A, Chen W. A multiscale chemical-mechanical model predicts impact of morphogen spreading on tissue growth. NPJ Syst Biol Appl 2023; 9:16. [PMID: 37210381 DOI: 10.1038/s41540-023-00278-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 05/03/2023] [Indexed: 05/22/2023] Open
Abstract
The exact mechanism controlling cell growth remains a grand challenge in developmental biology and regenerative medicine. The Drosophila wing disc tissue serves as an ideal biological model to study mechanisms involved in growth regulation. Most existing computational models for studying tissue growth focus specifically on either chemical signals or mechanical forces. Here we developed a multiscale chemical-mechanical model to investigate the growth regulation mechanism based on the dynamics of a morphogen gradient. By comparing the spatial distribution of dividing cells and the overall tissue shape obtained in model simulations with experimental data of the wing disc, it is shown that the size of the domain of the Dpp morphogen is critical in determining tissue size and shape. A larger tissue size with a faster growth rate and more symmetric shape can be achieved if the Dpp gradient spreads in a larger domain. Together with Dpp absorbance at the peripheral zone, the feedback regulation that downregulates Dpp receptors on the cell membrane allows for further spreading of the morphogen away from its source region, resulting in prolonged tissue growth at a more spatially homogeneous growth rate.
Collapse
Affiliation(s)
- Alireza Ramezani
- Department of Physics and Astronomy, University of California, Riverside, CA, 92521, USA
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
| | - Samuel Britton
- Department of Mathematics, University of California, Riverside, CA, 92521, USA
| | - Roya Zandi
- Department of Physics and Astronomy, University of California, Riverside, CA, 92521, USA
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
| | - Mark Alber
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
- Department of Mathematics, University of California, Riverside, CA, 92521, USA
| | - Ali Nematbakhsh
- Department of Mathematics, University of California, Riverside, CA, 92521, USA.
| | - Weitao Chen
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA.
- Department of Mathematics, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|
7
|
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] [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
|
8
|
Prasad D, Illek K, Fischer F, Holstein K, Classen AK. Bilateral JNK activation is a hallmark of interface surveillance and promotes elimination of aberrant cells. eLife 2023; 12:e80809. [PMID: 36744859 PMCID: PMC9917460 DOI: 10.7554/elife.80809] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
Tissue-intrinsic defense mechanisms eliminate aberrant cells from epithelia and thereby maintain the health of developing tissues or adult organisms. 'Interface surveillance' comprises one such distinct mechanism that specifically guards against aberrant cells which undergo inappropriate cell fate and differentiation programs. The cellular mechanisms which facilitate detection and elimination of these aberrant cells are currently unknown. We find that in Drosophila imaginal discs, clones of cells with inappropriate activation of cell fate programs induce bilateral JNK activation at clonal interfaces, where wild type and aberrant cells make contact. JNK activation is required to drive apoptotic elimination of interface cells. Importantly, JNK activity and apoptosis are highest in interface cells within small aberrant clones, which likely supports the successful elimination of aberrant cells when they arise. Our findings are consistent with a model where clone size affects the topology of interface contacts and thereby the strength of JNK activation in wild type and aberrant interface cells. Bilateral JNK activation is unique to 'interface surveillance' and is not observed in other tissue-intrinsic defense mechanisms, such as classical 'cell-cell competition'. Thus, bilateral JNK interface signaling provides an independent tissue-level mechanism to eliminate cells with inappropriate developmental fate but normal cellular fitness. Finally, oncogenic Ras-expressing clones activate 'interface surveillance' but evade elimination by bilateral JNK activation. Combined, our work establishes bilateral JNK interface signaling and interface apoptosis as a new hallmark of interface surveillance and highlights how oncogenic mutations evade tumor suppressor function encoded by this tissue-intrinsic surveillance system.
Collapse
Affiliation(s)
- Deepti Prasad
- Hilde-Mangold-Haus, University of FreiburgFreiburgGermany
- Spemann Graduate School of Biology and Medicine (SGBM), University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
| | | | - Friedericke Fischer
- Hilde-Mangold-Haus, University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
- International Max Planck Research School for Immunobiology, Epigenetics, and MetabolismFreiburgGermany
| | | | - Anne-Kathrin Classen
- Hilde-Mangold-Haus, University of FreiburgFreiburgGermany
- Faculty of Biology, University of FreiburgFreiburgGermany
- CIBSS Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
- BIOSS Centre for Biological Signalling Studies, University of FreiburgFreiburgGermany
| |
Collapse
|
9
|
Cell polarity and extrusion: How to polarize extrusion and extrude misspolarized cells? Curr Top Dev Biol 2023; 154:131-167. [PMID: 37100516 DOI: 10.1016/bs.ctdb.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The barrier function of epithelia is one of the cornerstones of the body plan organization of metazoans. It relies on the polarity of epithelial cells which organizes along the apico-basal axis the mechanical properties, signaling as well as transport. This barrier function is however constantly challenged by the fast turnover of epithelia occurring during morphogenesis or adult tissue homeostasis. Yet, the sealing property of the tissue can be maintained thanks to cell extrusion: a series of remodeling steps involving the dying cell and its neighbors leading to seamless cell expulsion. Alternatively, the tissue architecture can also be challenged by local damages or the emergence of mutant cells that may alter its organization. This includes mutants of the polarity complexes which can generate neoplastic overgrowths or be eliminated by cell competition when surrounded by wild type cells. In this review, we will provide an overview of the regulation of cell extrusion in various tissues focusing on the relationship between cell polarity, cell organization and the direction of cell expulsion. We will then describe how local perturbations of polarity can also trigger cell elimination either by apoptosis or by cell exclusion, focusing specifically on how polarity defects can be directly causal to cell elimination. Overall, we propose a general framework connecting the influence of polarity on cell extrusion and its contribution to aberrant cell elimination.
Collapse
|
10
|
Macabenta F, Sun HT, Stathopoulos A. BMP-gated cell-cycle progression drives anoikis during mesenchymal collective migration. Dev Cell 2022; 57:1683-1693.e3. [PMID: 35709766 PMCID: PMC9339487 DOI: 10.1016/j.devcel.2022.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/02/2022] [Accepted: 05/20/2022] [Indexed: 11/03/2022]
Abstract
Tissue homeostasis involves the elimination of abnormal cells to avoid compromised patterning and function. Although quality control through cell competition is well studied in epithelial tissues, it is unknown if and how homeostasis is regulated in mesenchymal collectives. Here, we demonstrate that collectively migrating Drosophila muscle precursors utilize both fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) signaling to promote homeostasis via anoikis, a form of cell death in response to substrate de-adhesion. Cell-cycle-regulated expression of the cell death gene head involution defective is responsible for caudal visceral mesoderm (CVM) anoikis. The secreted BMP ligand drives cell-cycle progression via a visceral mesoderm-specific cdc25/string enhancer to synchronize collective proliferation, as well as apoptosis of cells that have lost access to substrate-derived FGF. Perturbation of BMP-dependent cell-cycle progression is sufficient to confer anoikis resistance to mismigrating cells and thus facilitate invasion of other tissues. This BMP-gated cell-cycle checkpoint defines a quality control mechanism during mesenchymal collective migration.
Collapse
Affiliation(s)
- Frank Macabenta
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Hsuan-Te Sun
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- California Institute of Technology, Division of Biology and Biological Engineering, 1200 East California Boulevard, Pasadena, CA 91125, USA.
| |
Collapse
|
11
|
Reibring CG, El Shahawy M, Hallberg K, Harfe BD, Linde A, Gritli-Linde A. Loss of BMP2 and BMP4 Signaling in the Dental Epithelium Causes Defective Enamel Maturation and Aberrant Development of Ameloblasts. Int J Mol Sci 2022; 23:6095. [PMID: 35682776 PMCID: PMC9180982 DOI: 10.3390/ijms23116095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/18/2022] [Accepted: 05/25/2022] [Indexed: 12/10/2022] Open
Abstract
BMP signaling is crucial for differentiation of secretory ameloblasts, the cells that secrete enamel matrix. However, whether BMP signaling is required for differentiation of maturation-stage ameloblasts (MA), which are instrumental for enamel maturation into hard tissue, is hitherto unknown. To address this, we used an in vivo genetic approach which revealed that combined deactivation of the Bmp2 and Bmp4 genes in the murine dental epithelium causes development of dysmorphic and dysfunctional MA. These fail to exhibit a ruffled apical plasma membrane and to reabsorb enamel matrix proteins, leading to enamel defects mimicking hypomaturation amelogenesis imperfecta. Furthermore, subsets of mutant MA underwent pathological single or collective cell migration away from the ameloblast layer, forming cysts and/or exuberant tumor-like and gland-like structures. Massive apoptosis in the adjacent stratum intermedium and the abnormal cell-cell contacts and cell-matrix adhesion of MA may contribute to this aberrant behavior. The mutant MA also exhibited severely diminished tissue non-specific alkaline phosphatase activity, revealing that this enzyme's activity in MA crucially depends on BMP2 and BMP4 inputs. Our findings show that combined BMP2 and BMP4 signaling is crucial for survival of the stratum intermedium and for proper development and function of MA to ensure normal enamel maturation.
Collapse
Affiliation(s)
- Claes-Göran Reibring
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Maha El Shahawy
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
- Department of Oral Biology, Faculty of Dentistry, Minia University, Minia 61511, Egypt
| | - Kristina Hallberg
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Brian D. Harfe
- Department of Molecular Genetics and Microbiology Genetics Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Anders Linde
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| | - Amel Gritli-Linde
- Department of Oral Biochemistry, Institute of Odontology, Sahlgrenska Academy at the University of Gothenburg, SE-40530 Göteborg, Sweden; (C.-G.R.); (M.E.S.); (K.H.); (A.L.)
| |
Collapse
|
12
|
Yang S, Wu X, Daoutidou EI, Zhang Y, Shimell M, Chuang KH, Peterson AJ, O'Connor MB, Zheng X. The NDNF-like factor Nord is a Hedgehog-induced extracellular BMP modulator that regulates Drosophila wing patterning and growth. eLife 2022; 11:e73357. [PMID: 35037619 PMCID: PMC8856659 DOI: 10.7554/elife.73357] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/15/2022] [Indexed: 11/23/2022] Open
Abstract
Hedgehog (Hh) and Bone Morphogenetic Proteins (BMPs) pattern the developing Drosophila wing by functioning as short- and long-range morphogens, respectively. Here, we show that a previously unknown Hh-dependent mechanism fine-tunes the activity of BMPs. Through genome-wide expression profiling of the Drosophila wing imaginal discs, we identify nord as a novel target gene of the Hh signaling pathway. Nord is related to the vertebrate Neuron-Derived Neurotrophic Factor (NDNF) involved in congenital hypogonadotropic hypogonadism and several types of cancer. Loss- and gain-of-function analyses implicate Nord in the regulation of wing growth and proper crossvein patterning. At the molecular level, we present biochemical evidence that Nord is a secreted BMP-binding protein and localizes to the extracellular matrix. Nord binds to Decapentaplegic (Dpp) or the heterodimer Dpp-Glass-bottom boat (Gbb) to modulate their release and activity. Furthermore, we demonstrate that Nord is a dosage-dependent BMP modulator, where low levels of Nord promote and high levels inhibit BMP signaling. Taken together, we propose that Hh-induced Nord expression fine-tunes both the range and strength of BMP signaling in the developing Drosophila wing.
Collapse
Affiliation(s)
- Shu Yang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Xuefeng Wu
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Euphrosyne I Daoutidou
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Ya Zhang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - MaryJane Shimell
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Kun-Han Chuang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Aidan J Peterson
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Michael B O'Connor
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| |
Collapse
|
13
|
Vasudevan D, Katow H, Huang HW, Tang G, Ryoo HD. A Protein-trap allele reveals roles for Drosophila ATF4 in photoreceptor degeneration, oogenesis and wing development. Dis Model Mech 2021; 15:273766. [PMID: 34919148 PMCID: PMC8938396 DOI: 10.1242/dmm.049119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/08/2021] [Indexed: 11/20/2022] Open
Abstract
Metazoans have evolved various quality control mechanisms to cope with cellular stress inflicted by external and physiological conditions. ATF4 is a major effector of the Integrated Stress Response (ISR), an evolutionarily conserved pathway that mediates adaptation to various cellular stressors. Loss of function of Drosophila ATF4, encoded by the gene cryptocephal (crc), results in lethality during pupal development. The roles of crc in Drosophila disease models and in adult tissue homeostasis thus remain poorly understood. Here, we report that a protein-trap MiMIC insertion in the crc locus generates a crc-GFP fusion protein that allows visualization of crc activity in vivo. This allele also acts as a hypomorphic mutant that uncovers previously unknown roles for crc. Specifically, the crc protein-trap line shows crc-GFP induction in a Drosophila model for Retinitis Pigmentosa (RP). This crc allele renders flies more vulnerable to amino acid deprivation and age-dependent retinal degeneration. These mutants also show defects in wing veins and oocyte maturation. Together, our data reveal previously unknown roles for crc in development, cellular homeostasis and photoreceptor survival.
Collapse
Affiliation(s)
- Deepika Vasudevan
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA.,Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hidetaka Katow
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | | | - Grace Tang
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
14
|
Phenotypical and genetical characterization of the Mad 1-2 allele during Drosophila wing development. Cells Dev 2021; 169:203761. [PMID: 34875394 DOI: 10.1016/j.cdev.2021.203761] [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: 05/17/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/23/2022]
Abstract
Growth and patterning of Drosophila wing depends upon the sequential organizing activities of Hedgehog (Hh) and Decapentaplegic (Dpp) signaling pathways. The Hh signaling directly activates the expression of dpp through the transcription factor cubitus interruptus (Ci). Dpp itself functions as a long-range morphogen to promote cell proliferation and differentiation through an essential transcription factor encoded by Mad. Here we report that the Mad1-2 allele exhibits phenotypes distinct from classical Dpp pathway mutants in the developing wing. The activity of Dpp signaling is attenuated in Mad1-2 mutant cells. However, activation of Dpp signaling is found in a subset of cells surrounding homozygous Mad1-2 clones when the clones are located at the anterior compartment of wing disc. Further analysis reveals that Mad1-2 mutant cells display high level of Hh signaling activity and accumulate significant amount of Ci. Unexpectedly, whole genome resequencing identifies multiple mutations in the 3'UTR region of Pka-C1 genomic loci in the Mad1-2 stock. We provide genetic and molecular evidence that the Pka-C1 mutations carried by Mad1-2 likely underlies the observed Hh signaling defects. Therefore, the contribution of Pka-C1 mutation should be taken in consideration when analyzing Mad1-2 phenotypes. The isolation of independent Mad and Pka-C1 alleles from the Mad1-2 stock further supports our conclusions.
Collapse
|
15
|
Matsuda S, Schaefer JV, Mii Y, Hori Y, Bieli D, Taira M, Plückthun A, Affolter M. Asymmetric requirement of Dpp/BMP morphogen dispersal in the Drosophila wing disc. Nat Commun 2021; 12:6435. [PMID: 34750371 PMCID: PMC8576045 DOI: 10.1038/s41467-021-26726-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 10/20/2021] [Indexed: 11/26/2022] Open
Abstract
How morphogen gradients control patterning and growth in developing tissues remains largely unknown due to lack of tools manipulating morphogen gradients. Here, we generate two membrane-tethered protein binders that manipulate different aspects of Decapentaplegic (Dpp), a morphogen required for overall patterning and growth of the Drosophila wing. One is "HA trap" based on a single-chain variable fragment (scFv) against the HA tag that traps HA-Dpp to mainly block its dispersal, the other is "Dpp trap" based on a Designed Ankyrin Repeat Protein (DARPin) against Dpp that traps Dpp to block both its dispersal and signaling. Using these tools, we found that, while posterior patterning and growth require Dpp dispersal, anterior patterning and growth largely proceed without Dpp dispersal. We show that dpp transcriptional refinement from an initially uniform to a localized expression and persistent signaling in transient dpp source cells render the anterior compartment robust against the absence of Dpp dispersal. Furthermore, despite a critical requirement of dpp for the overall wing growth, neither Dpp dispersal nor direct signaling is critical for lateral wing growth after wing pouch specification. These results challenge the long-standing dogma that Dpp dispersal is strictly required to control and coordinate overall wing patterning and growth.
Collapse
Affiliation(s)
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Yusuke Mii
- National Institute for Basic Biology and Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- JST PRESTO, Kawaguchi, Saitama, Japan
| | - Yutaro Hori
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | | | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Department of Biological Sciences, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
16
|
Costa-Rodrigues C, Couceiro J, Moreno E. Cell competition from development to neurodegeneration. Dis Model Mech 2021; 14:269331. [PMID: 34190316 PMCID: PMC8277968 DOI: 10.1242/dmm.048926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell competition is a process by which suboptimal cells are eliminated to the benefit of cells with higher fitness. It is a surveillance mechanism that senses differences in the fitness status by several modes, such as expression of fitness fingerprints, survival factor uptake rate and resistance to mechanical stress. Fitness fingerprints-mediated cell competition recognizes isoforms of the transmembrane protein Flower, and translates the relative fitness of cells into distinct fates through the Flower code. Impairments in cell competition potentiate the development of diseases like cancer and ageing-related pathologies. In cancer, malignant cells acquire a supercompetitor behaviour, killing the neighbouring cells and overtaking the tissue, thus avoiding elimination. Neurodegenerative disorders affect millions of people and are characterized by cognitive decline and locomotor deficits. Alzheimer's disease is the most common form of dementia, and one of the largely studied diseases. However, the cellular processes taking place remain unclear. Drosophila melanogaster is an emerging neurodegeneration model due to its versatility as a tool for genetic studies. Research in a Drosophila Alzheimer's disease model detected fitness markers in the suboptimal and hyperactive neurons, thus establishing a link between cell competition and Alzheimer's disease. In this Review, we overview cell competition and the new insights related to neurodegenerative disorders, and discuss how research in the field might contribute to the development of new therapeutic targets for these diseases.
Collapse
Affiliation(s)
| | - Joana Couceiro
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Eduardo Moreno
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
17
|
Novel function of N-acetyltransferase for microtubule stability and JNK signaling in Drosophila organ development. Proc Natl Acad Sci U S A 2021; 118:2010140118. [PMID: 33479178 DOI: 10.1073/pnas.2010140118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Regulation of microtubule stability is crucial for the maintenance of cell structure and function. While the acetylation of α-tubulin lysine 40 by acetylase has been implicated in the regulation of microtubule stability, the in vivo functions of N-terminal acetyltransferases (NATs) involved in the acetylation of N-terminal amino acids are not well known. Here, we identify an N-terminal acetyltransferase, Mnat9, that regulates cell signaling and microtubule stability in Drosophila Loss of Mnat9 causes severe developmental defects in multiple tissues. In the wing imaginal disc, Mnat9 RNAi leads to the ectopic activation of c-Jun N-terminal kinase (JNK) signaling and apoptotic cell death. These defects are suppressed by reducing the level of JNK signaling. Overexpression of Mnat9 can also inhibit JNK signaling. Mnat9 colocalizes with mitotic spindles, and its loss results in various spindle defects during mitosis in the syncytial embryo. Furthermore, overexpression of Mnat9 enhances microtubule stability. Mnat9 is physically associated with microtubules and shows a catalytic activity in acetylating N-terminal peptides of α- and β-tubulin in vitro. Cell death and tissue loss in Mnat9-depleted wing discs are restored by reducing the severing protein Spastin, suggesting that Mnat9 protects microtubules from its severing activity. Remarkably, Mnat9 mutated in the acetyl-CoA binding site is as functional as its wild-type form. We also find that human NAT9 can rescue Mnat9 RNAi phenotypes in flies, indicating their functional conservation. Taken together, we propose that Mnat9 is required for microtubule stability and regulation of JNK signaling to promote cell survival in developing Drosophila organs.
Collapse
|
18
|
Fan WM, Luo D, Zhang JZ, Wang D, Shen J. Vestigial suppresses apoptosis and cell migration in a manner dependent on the level of JNK-Caspase signaling in the Drosophila wing disc. INSECT SCIENCE 2021; 28:63-76. [PMID: 32037698 DOI: 10.1111/1744-7917.12762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/02/2020] [Accepted: 01/24/2020] [Indexed: 06/10/2023]
Abstract
The Decapentaplegic (Dpp) and Wingless (Wg) signal pathways play important roles in numerous biological processes in Drosophila. The Drosophila vestigial (vg) gene is selectively required for wing imaginal disc cell proliferation, which is essential for the formation of the adult wing and halter structures, and is regulated by Dpp and Wg signaling. Using a Drosophila invasion model of wing epithelium, we showed herein that inhibition of Dpp or Wg signaling promoted cells to migrate across the cell lineage restrictive anterior/posterior (A/P) compartment boundary. Being downstream of both Dpp and Wg signaling, vg can block cell migration induced by loss of either pathway. In addition, suppression of vg is sufficient to induce cell migration across the A/P boundary. Transcriptomic analysis revealed potential downstream genes involved in the cell migration after suppressing vg in the wing disc. We further demonstrated that the c-Jun N-terminal kinase (JNK) signaling promoted cell migration induced by vg suppression by upregulating Caspase activity. Taken together, our results revealed the requirement of Vg for suppressing cell migration and clarified how developmental signals collaborate to stabilize cells along the compartment boundary.
Collapse
Affiliation(s)
- Wen-Min Fan
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Dan Luo
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Jun-Zheng Zhang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Dan Wang
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| | - Jie Shen
- Department of Entomology and MOA Lab for Pest Monitoring and Green Control, China Agricultural University, Beijing, China
| |
Collapse
|
19
|
Santacreu BJ, Romero DJ, Pescio LG, Tarallo E, Sterin-Speziale NB, Favale NO. Apoptotic cell extrusion depends on single-cell synthesis of sphingosine-1-phosphate by sphingosine kinase 2. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158888. [PMID: 33454434 DOI: 10.1016/j.bbalip.2021.158888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/19/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022]
Abstract
Collecting duct cells are physiologically subject to the hypertonic environment of the kidney. This condition is necessary for kidney maturation and function but represents a stress condition that requires active strategies to ensure epithelial integrity. Madin-Darby Canine Kidney (MDCK) cells develop the differentiated phenotype of collecting duct cells when subject to hypertonicity, serving as a model to study epithelial preservation and homeostasis in this particular environment. The integrity of epithelia is essential to achieve the required functional barrier. One of the mechanisms that ensure integrity is cell extrusion, a process initiated by sphingosine-1-phosphate (S1P) to remove dying or surplus cells while maintaining the epithelium barrier. Both types start with the activation of S1P receptor type 2, located in neighboring cells. In this work, we studied the effect of cell differentiation induced by hypertonicity on cell extrusion in MDCK cells, and we provide new insights into the associated molecular mechanism. We found that the different stages of differentiation influence the rate of apoptotic cell extrusion. Besides, we used a novel methodology to demonstrate that S1P increase in extruding cells of differentiated monolayers. These results show for first time that cell extrusion is triggered by the single-cell synthesis of S1P by sphingosine kinase 2 (SphK2), but not SphK1, of the extruding cell itself. Moreover, the inhibition or knockdown of SphK2 prevents cell extrusion and cell-cell junction protein degradation, but not apoptotic nuclear fragmentation. Thus, we propose SphK2 as the biochemical key to ensure the preservation of the epithelial barrier under hypertonic stress.
Collapse
Affiliation(s)
- Bruno Jaime Santacreu
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Profesor Dr. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Daniela Judith Romero
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Profesor Dr. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Lucila Gisele Pescio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Profesor Dr. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina
| | - Estefanía Tarallo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina
| | - Norma Beatriz Sterin-Speziale
- CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Profesor Dr. Alejandro C. Paladini" (IQUIFIB), Laboratorio Nacional de Investigación y Servicios de Péptidos y Proteínas - Espectrometría de Masa (LANAIS PROEM), Buenos Aires, Argentina
| | - Nicolás Octavio Favale
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas "Profesor Dr. Alejandro C. Paladini" (IQUIFIB), Buenos Aires, Argentina.
| |
Collapse
|
20
|
Scaling a Dpp Morphogen Gradient through Feedback Control of Receptors and Co-receptors. Dev Cell 2021; 53:724-739.e14. [PMID: 32574592 DOI: 10.1016/j.devcel.2020.05.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/23/2020] [Accepted: 05/26/2020] [Indexed: 11/21/2022]
Abstract
Gradients of decapentaplegic (Dpp) pattern Drosophila wing imaginal discs, establishing gene expression boundaries at specific locations. As discs grow, Dpp gradients expand, keeping relative boundary positions approximately stationary. Such scaling fails in mutants for Pentagone (pent), a gene repressed by Dpp that encodes a diffusible protein that expands Dpp gradients. Although these properties fit a recent mathematical model of automatic gradient scaling, that model requires an expander that spreads with minimal loss throughout a morphogen field. Here, we show that Pent's actions are confined to within just a few cell diameters of its site of synthesis and can be phenocopied by manipulating non-diffusible Pent targets strictly within the Pent expression domain. Using genetics and mathematical modeling, we develop an alternative model of scaling driven by feedback downregulation of Dpp receptors and co-receptors. Among the model's predictions is a size beyond which scaling fails-something we observe directly in wing discs.
Collapse
|
21
|
Analysis of Epithelial Architecture and Planar Spindle Orientation in the Drosophila Wing Disc. Methods Mol Biol 2020. [PMID: 33280064 DOI: 10.1007/7651_2020_340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The Drosophila melanogaster wing imaginal disc is an epithelial sac that exhibits dramatic tissue growth during the larval stage. With its simple morphology and accessibility of genetic tools, studies using the wing disc have contributed to the understanding of the mechanisms of epithelial homeostasis including the control of mitotic spindle orientation. This chapter describes a detailed protocol for analyzing epithelial architecture and planar orientation of the mitotic spindle in the wing disc epithelium. The rapid dissection method, effective immunostaining, and mounting tips described here facilitate genetic and cell biological studies of the wing disc and can be applied to a wide array of studies using various Drosophila tissues.
Collapse
|
22
|
Bairzin JCD, Emmons-Bell M, Hariharan IK. The Hippo pathway coactivator Yorkie can reprogram cell fates and create compartment-boundary-like interactions at clone margins. SCIENCE ADVANCES 2020; 6:6/50/eabe8159. [PMID: 33298454 PMCID: PMC7725458 DOI: 10.1126/sciadv.abe8159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/22/2020] [Indexed: 06/12/2023]
Abstract
During development, tissue-specific patterns of gene expression are established by transcription factors and then stably maintained via epigenetic mechanisms. Cancer cells often express genes that are inappropriate for that tissue or developmental stage. Here, we show that high activity levels of Yki, the Hippo pathway coactivator that causes overgrowth in Drosophila imaginal discs, can also disrupt cell fates by altering expression of selector genes like engrailed (en) and Ultrabithorax (Ubx). Posterior clones expressing activated Yki can down-regulate en and express an anterior selector gene, cubitus interruptus (ci). The microRNA bantam and the chromatin regulator Taranis both function downstream of Yki in promoting ci expression. The boundary between Yki-expressing posterior clones and surrounding wild-type cells acquires properties reminiscent of the anteroposterior compartment boundary; Hedgehog signaling pathway activation results in production of Dpp. Thus, at least in principle, heterotypic interactions between Yki-expressing cells and their neighbors could activate boundary-specific signaling mechanisms.
Collapse
Affiliation(s)
- Joanna C D Bairzin
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Maya Emmons-Bell
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
23
|
Michel M, Dahmann C. Tissue mechanical properties modulate cell extrusion in the Drosophila abdominal epidermis. Development 2020; 147:147/5/dev179606. [DOI: 10.1242/dev.179606] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 01/28/2020] [Indexed: 01/22/2023]
Abstract
ABSTRACT
The replacement of cells is a common strategy during animal development. In the Drosophila pupal abdomen, larval epidermal cells (LECs) are replaced by adult progenitor cells (histoblasts). Previous work showed that interactions between histoblasts and LECs result in apoptotic extrusion of LECs during early pupal development. Extrusion of cells is closely preceded by caspase activation and is executed by contraction of a cortical actomyosin cable. Here, we identify a population of LECs that extrudes independently of the presence of histoblasts during late pupal development. Extrusion of these LECs is not closely preceded by caspase activation, involves a pulsatile medial actomyosin network, and correlates with a developmental time period when mechanical tension and E-cadherin turnover at adherens junctions is particularly high. Our work reveals a developmental switch in the cell extrusion mechanism that correlates with changes in tissue mechanical properties.
Collapse
Affiliation(s)
- Marcus Michel
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany
| |
Collapse
|
24
|
Bajpai A, Sinha P. Hh signaling from de novo organizers drive lgl neoplasia in Drosophila epithelium. Dev Biol 2020; 457:1-8. [PMID: 31557471 PMCID: PMC7116463 DOI: 10.1016/j.ydbio.2019.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/22/2019] [Accepted: 09/20/2019] [Indexed: 01/20/2023]
Abstract
The Hedgehog (Hh) morphogen regulates growth and patterning. Since Hh signaling is also implicated in carcinogenesis, it is conceivable that de novo Hh-secreting organizers, if formed in association with oncogenic hit could be tumor-cooperative. Here we validate this hypothesis using the Drosophila model of cooperative epithelial carcinogenesis. We generate somatic clones with simultaneous loss of tumor suppressor, Lgl, and gain of the posterior compartment selector, Engrailed (En), known to induce synthesis of Hh. We show that lgl UAS-en clones in the anterior wing compartment trigger Hh signaling cascade via cross-talk with their Ci-expressing wild type cell neighbors. Hh-Dpp signaling from clone boundaries of such ectopically formed de novo organizers in turn drive lgl carcinogenesis. By contrast, Ci-expressing lgl clones transform by autocrine and/or juxtracine activation of Hh signaling in only the posterior compartment. We further show that sequestration of the Hh ligand or loss of Dpp receptor, Tkv, in these Hh-sending or -receiving lgl clones arrested their carcinogenesis. Our results therefore reveal a hitherto unrecognized mechanism of tumor cooperation by developmental organizers, which are induced fortuitously by oncogenic hits.
Collapse
Affiliation(s)
- Anjali Bajpai
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208 016, India.
| | - Pradip Sinha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208 016, India.
| |
Collapse
|
25
|
Klipa O, Hamaratoglu F. Cell elimination strategies upon identity switch via modulation of apterous in Drosophila wing disc. PLoS Genet 2019; 15:e1008573. [PMID: 31877129 PMCID: PMC6952109 DOI: 10.1371/journal.pgen.1008573] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/09/2020] [Accepted: 12/17/2019] [Indexed: 12/04/2022] Open
Abstract
The ability to establish spatial organization is an essential feature of any developing tissue and is achieved through well-defined rules of cell-cell communication. Maintenance of this organization requires elimination of cells with inappropriate positional identity, a poorly understood phenomenon. Here we studied mechanisms regulating cell elimination in the context of a growing tissue, the Drosophila wing disc and its dorsal determinant Apterous. Systematic analysis of apterous mutant clones along with their twin spots shows that they are eliminated from the dorsal compartment via three different mechanisms: relocation to the ventral compartment, basal extrusion, and death, depending on the position of the clone in the wing disc. We find that basal extrusion is the main elimination mechanism in the hinge, whereas apoptosis dominates in the pouch and in the notum. In the absence of apoptosis, extrusion takes over to ensure clearance in all regions. Notably, clones in the hinge grow larger than those in the pouch, emphasizing spatial differences. Mechanistically, we find that limiting cell division within the clones does not prevent their extrusion. Indeed, even clones of one or two cells can be extruded basally, demonstrating that the clone size is not the main determinant of the elimination mechanism to be used. Overall, we revealed three elimination mechanisms and their spatial biases for preserving pattern in a growing organ. As development proceeds, cells become more specialized and the compartmentalization ensures spatial separation of the specialized cells. This process of pattern formation is rather well understood. How the pattern is maintained afterwards though is largely unknown. Using the Drosophila wing disc as a model organ, we examined what happens to dorsal cells if they lose their dorsal identity. Formerly, it was shown that these cells are eliminated from the dorsal compartment via apoptosis or through relocation to the ventral compartment. Here we show that a third mode of elimination, basal extrusion, also contributes to their clearing. We quantified, for the first time, contributions of each mechanism and discovered a regional bias in their operation. Importantly, if apoptosis is blocked, basal extrusion takes over to ensure clearance from all regions. Recent modeling approaches suggested that there is a lower limit to the clone size for extrusion. Therefore, we tested the hypothesis that the choice of elimination mechanism may be dictated by the clone size. We prevented cell divisions within the clones to be eliminated and found that even 1–2 cell clones readily underwent basal extrusion, demonstrating that there is no lower limit to the clone size for extrusion.
Collapse
Affiliation(s)
- Olga Klipa
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Fisun Hamaratoglu
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Coelho DS, Schwartz S, Merino MM, Hauert B, Topfel B, Tieche C, Rhiner C, Moreno E. Culling Less Fit Neurons Protects against Amyloid-β-Induced Brain Damage and Cognitive and Motor Decline. Cell Rep 2019; 25:3661-3673.e3. [PMID: 30590040 PMCID: PMC6315112 DOI: 10.1016/j.celrep.2018.11.098] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/05/2018] [Accepted: 11/28/2018] [Indexed: 01/25/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia, impairing cognitive and motor functions. One of the pathological hallmarks of AD is neuronal loss, which is not reflected in mouse models of AD. Therefore, the role of neuronal death is still uncertain. Here, we used a Drosophila AD model expressing a secreted form of human amyloid-β42 peptide and showed that it recapitulates key aspects of AD pathology, including neuronal death and impaired long-term memory. We found that neuronal apoptosis is mediated by cell fitness-driven neuronal culling, which selectively eliminates impaired neurons from brain circuits. We demonstrated that removal of less fit neurons delays β-amyloid-induced brain damage and protects against cognitive and motor decline, suggesting that contrary to common knowledge, neuronal death may have a beneficial effect in AD. Peptides linked to neurodegenerative diseases reduce neuronal fitness in Drosophila β-amyloid-induced neuronal death is mediated by fitness regulators flower and azot Suppression of fitness-based neuronal culling aggravates cognitive and motor decline Neuronal death related to fitness-based selection has a beneficial net effect
Collapse
Affiliation(s)
- Dina S Coelho
- Cell Fitness Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Silvia Schwartz
- Stem Cells and Regeneration Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal
| | - Marisa M Merino
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland; Department of Biochemistry, University of Geneva, Quai Ernest-Ansermet 30, 1211 Geneva 4, Switzerland
| | - Barbara Hauert
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Barbara Topfel
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Colin Tieche
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Christa Rhiner
- Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland; Stem Cells and Regeneration Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal.
| | - Eduardo Moreno
- Cell Fitness Lab, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal; Institute for Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland.
| |
Collapse
|
27
|
Matamoro-Vidal A, Levayer R. Multiple Influences of Mechanical Forces on Cell Competition. Curr Biol 2019; 29:R762-R774. [DOI: 10.1016/j.cub.2019.06.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
28
|
Andrés-Delgado L, Ernst A, Galardi-Castilla M, Bazaga D, Peralta M, Münch J, González-Rosa JM, Marques I, Tessadori F, de la Pompa JL, Vermot J, Mercader N. Actin dynamics and the Bmp pathway drive apical extrusion of proepicardial cells. Development 2019; 146:dev.174961. [PMID: 31175121 PMCID: PMC6633599 DOI: 10.1242/dev.174961] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 05/24/2019] [Indexed: 12/30/2022]
Abstract
The epicardium, the outer mesothelial layer enclosing the myocardium, plays key roles in heart development and regeneration. During embryogenesis, the epicardium arises from the proepicardium (PE), a cell cluster that appears in the dorsal pericardium (DP) close to the venous pole of the heart. Little is known about how the PE emerges from the pericardial mesothelium. Using a zebrafish model and a combination of genetic tools, pharmacological agents and quantitative in vivo imaging, we reveal that a coordinated collective movement of DP cells drives PE formation. We found that Bmp signaling and the actomyosin cytoskeleton promote constriction of the DP, which enables PE cells to extrude apically. We provide evidence that cell extrusion, which has been described in the elimination of unfit cells from epithelia and the emergence of hematopoietic stem cells, is also a mechanism for PE cells to exit an organized mesothelium and fulfil their developmental fate to form a new tissue layer, the epicardium. Summary: Proepicardial cells emerge from the pericardial mesothelium through apical extrusion, a process that depends on BMP signaling and actomyosin rearrangements.
Collapse
Affiliation(s)
- Laura Andrés-Delgado
- Development of the Epicardium and its Role During Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.,Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Alexander Ernst
- Institute of Anatomy, University of Bern, 3000 Bern 9, Switzerland
| | - María Galardi-Castilla
- Development of the Epicardium and its Role During Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - David Bazaga
- Development of the Epicardium and its Role During Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Marina Peralta
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France.,Université de Strasbourg, 67411 Illkirch, France
| | - Juliane Münch
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.,Ciber CV, 28029 Madrid, Spain
| | - Juan M González-Rosa
- Development of the Epicardium and its Role During Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Inês Marques
- Institute of Anatomy, University of Bern, 3000 Bern 9, Switzerland
| | - Federico Tessadori
- Hubrecht Institute-KNAW and UMC Utrecht, Uppsalalaan 8, 3584CT Utrecht, The Netherlands
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain.,Ciber CV, 28029 Madrid, Spain
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France.,Université de Strasbourg, 67411 Illkirch, France
| | - Nadia Mercader
- Development of the Epicardium and its Role During Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain .,Institute of Anatomy, University of Bern, 3000 Bern 9, Switzerland
| |
Collapse
|
29
|
Levayer R. Solid stress, competition for space and cancer: The opposing roles of mechanical cell competition in tumour initiation and growth. Semin Cancer Biol 2019; 63:69-80. [PMID: 31077845 PMCID: PMC7221353 DOI: 10.1016/j.semcancer.2019.05.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/23/2019] [Accepted: 05/07/2019] [Indexed: 12/24/2022]
Abstract
The regulation of cell growth, cell proliferation and cell death is at the basis of the homeostasis of tissues. While they can be regulated by intrinsic and genetic factors, their response to external signals emanating from the local environment is also essential for tissue homeostasis. Tumour initiation and progression is based on the misregulation of growth, proliferation and death mostly through the accumulation of genetic mutations. Yet, there is an increasing body of evidences showing that tumour microenvironment also has a strong impact on cancer initiation and progression. This includes the mechanical constrains and the compressive forces generated by the resistance of the surrounding tissue/matrix to tumour expansion. Recently, mechanical stress has been proposed to promote competitive interactions between cells through a process called mechanical cell competition. Cell population with a high proliferative rate can compact and eliminate the neighbouring cells which are more sensitive to compaction. While this emerging concept has been recently validated in vivo, the relevance of this process during tumour progression has never been discussed extensively. In this review, I will first describe the phenomenology of mechanical cell competition focusing on the main parameters and the pathways regulating cell elimination. I will then discuss the relevance of mechanical cell competition in tumour initiation and expansion while emphasizing its potential opposing contributions to tumourogenesis.
Collapse
Affiliation(s)
- Romain Levayer
- Institut Pasteur, Department of Developmental and Stem Cell Biology, 25 rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
30
|
Valon L, Levayer R. Dying under pressure: cellular characterisation and in vivo functions of cell death induced by compaction. Biol Cell 2019; 111:51-66. [PMID: 30609052 DOI: 10.1111/boc.201800075] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/05/2018] [Indexed: 12/16/2022]
Abstract
Cells and tissues are exposed to multiple mechanical stresses during development, tissue homoeostasis and diseases. While we start to have an extensive understanding of the influence of mechanics on cell differentiation and proliferation, how excessive mechanical stresses can also lead to cell death and may be associated with pathologies has been much less explored so far. Recently, the development of new perturbative approaches allowing modulation of pressure and deformation of tissues has demonstrated that compaction (the reduction of tissue size or volume) can lead to cell elimination. Here, we discuss the relevant type of stress and the parameters that could be causal to cell death from single cell to multicellular systems. We then compare the pathways and mechanisms that have been proposed to influence cell survival upon compaction. We eventually describe the relevance of compaction-induced death in vivo, and its functions in morphogenesis, tissue size regulation, tissue homoeostasis and cancer progression.
Collapse
Affiliation(s)
- Léo Valon
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, 75015, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, 75015, France
| |
Collapse
|
31
|
Escot S, Willnow D, Naumann H, Di Francescantonio S, Spagnoli FM. Robo signalling controls pancreatic progenitor identity by regulating Tead transcription factors. Nat Commun 2018; 9:5082. [PMID: 30504829 PMCID: PMC6269453 DOI: 10.1038/s41467-018-07474-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
A complex interplay of intrinsic factors and extrinsic signalling pathways controls both cell lineage commitment and maintenance of cell identity. Loss of defined cellular states is the cause of many different cancers, including pancreatic cancer. Recent findings suggest a clinical role for the conserved SLIT/ROBO signalling pathway in pancreatic cancer. However, whilst this pathway has been extensively studied in many processes, a role for Slit and Robo genes in pancreas cell identity and plasticity has not been established yet. Here, we identify Slit/Robo signalling as a key regulator of pancreatic progenitor identity. We find that Robo1 and Robo2 are required for preserving pancreatic cell identity shortly after fate induction and, subsequently, for expansion of the pancreatic progenitor pool in the mouse. Furthermore, we show that Robo receptors control the expression of Tead transcription factors as well as its downstream transcriptional activity. Our work identifies an interplay between Slit/Robo pathway and Tead intrinsic regulators, functioning as gatekeeper of pancreatic cell identity.
Collapse
Affiliation(s)
- Sophie Escot
- Lab. of Molecular and Cellular Basis of Embryonic Development, Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin, 13125, Germany
| | - David Willnow
- Lab. of Molecular and Cellular Basis of Embryonic Development, Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin, 13125, Germany.,Berlin Institute of Health (BIH), Berlin, 10178, Germany
| | - Heike Naumann
- Lab. of Molecular and Cellular Basis of Embryonic Development, Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin, 13125, Germany
| | - Silvia Di Francescantonio
- Lab. of Molecular and Cellular Basis of Embryonic Development, Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin, 13125, Germany
| | - Francesca M Spagnoli
- Lab. of Molecular and Cellular Basis of Embryonic Development, Max-Delbrueck Center for Molecular Medicine, Robert-Roessle Strasse 10, Berlin, 13125, Germany. .,Berlin Institute of Health (BIH), Berlin, 10178, Germany. .,Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
32
|
Tan S, Huan P, Liu B. An investigation of oyster TGF-β receptor genes and their potential roles in early molluscan development. Gene 2018; 663:65-71. [DOI: 10.1016/j.gene.2018.04.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/19/2018] [Accepted: 04/12/2018] [Indexed: 10/17/2022]
|
33
|
Girnius N, Davis RJ. JNK Promotes Epithelial Cell Anoikis by Transcriptional and Post-translational Regulation of BH3-Only Proteins. Cell Rep 2018; 21:1910-1921. [PMID: 29141222 DOI: 10.1016/j.celrep.2017.10.067] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 09/27/2017] [Accepted: 10/18/2017] [Indexed: 11/18/2022] Open
Abstract
Developmental morphogenesis, tissue injury, and oncogenic transformation can cause the detachment of epithelial cells. These cells are eliminated by a specialized form of apoptosis (anoikis). While the processes that contribute to this form of cell death have been studied, the underlying mechanisms remain unclear. Here, we tested the role of the cJUN NH2-terminal kinase (JNK) signaling pathway using murine models with compound JNK deficiency in mammary and kidney epithelial cells. These studies demonstrated that JNK is required for efficient anoikis in vitro and in vivo. Moreover, JNK-promoted anoikis required pro-apoptotic members of the BCL2 family of proteins. We show that JNK acts through a BAK/BAX-dependent apoptotic pathway by increasing BIM expression and phosphorylating BMF, leading to death of detached epithelial cells.
Collapse
Affiliation(s)
- Nomeda Girnius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
34
|
Akiyama T, User SD, Gibson MC. Somatic clones heterozygous for recessive disease alleles of BMPR1A exhibit unexpected phenotypes in Drosophila. eLife 2018; 7:35258. [PMID: 29745898 PMCID: PMC5963922 DOI: 10.7554/elife.35258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/02/2018] [Indexed: 01/02/2023] Open
Abstract
The majority of mutations studied in animal models are designated as recessive based on the absence of visible phenotypes in germline heterozygotes. Accordingly, genetic studies primarily rely on homozygous loss-of-function to determine gene requirements, and a conceptually-related ‘two-hit model’ remains the central paradigm in cancer genetics. Here we investigate pathogenesis due to somatic mutation in epithelial tissues, a process that predominantly generates heterozygous cell clones. To study somatic mutation in Drosophila, we generated inducible alleles that mimic human Juvenile polyposis-associated BMPR1A mutations. Unexpectedly, four of these mutations had no phenotype in heterozygous carriers but exhibited clear tissue-level effects when present in somatic clones of heterozygous cells. We conclude that these alleles are indeed recessive when present in the germline, but nevertheless deleterious when present in heterozygous clones. This unforeseen effect, deleterious heteromosaicism, suggests a ‘one-hit’ mechanism for disease initiation that may explain some instances of pathogenesis associated with spontaneous mutation.
Collapse
Affiliation(s)
- Takuya Akiyama
- Stowers Institute for Medical Research, Kansas City, United States
| | - Sırma D User
- Stowers Institute for Medical Research, Kansas City, United States
| | - Matthew C Gibson
- Stowers Institute for Medical Research, Kansas City, United States.,Department of Anatomy and Cell Biology, The University of Kansas School of Medicine, Kansas City, United States
| |
Collapse
|
35
|
Nigg EA, Schnerch D, Ganier O. Impact of Centrosome Aberrations on Chromosome Segregation and Tissue Architecture in Cancer. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2018; 82:137-144. [PMID: 29610243 DOI: 10.1101/sqb.2017.82.034421] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Centrosomes determine the disposition of microtubule networks and thereby contribute to regulate cell shape, polarity, and motility, as well as chromosome segregation during cell division. Additionally, centrioles, the core components of centrosomes, are required for the formation of cilia and flagella. Mutations in genes coding for centrosomal and centriolar proteins are responsible for several human diseases, foremost ciliopathies and developmental disorders resulting in small brains (primary microcephaly) or small body size (dwarfism). Moreover, a long-standing postulate implicates numerical and/or structural centrosome aberrations in the etiology of cancer. In this review, we will discuss recent work on the role of centrosome aberrations in the promotion of genome instability and the disruption of tissue architecture, two hallmarks of human cancers. We will emphasize recent studies on the impact of centrosome aberrations on the polarity of epithelial cells cultured in three-dimensional spheroid models. Collectively, the results from these in vitro systems suggest that different types of centrosome aberrations can promote invasive behavior through different pathways. Particularly exciting is recent evidence indicating that centrosome aberrations may trigger the dissemination of potentially metastatic cells through a non-cell-autonomous mechanism.
Collapse
Affiliation(s)
- Erich A Nigg
- Biozentrum, University of Basel, Basel CH-4056, Switzerland
| | | | - Olivier Ganier
- Biozentrum, University of Basel, Basel CH-4056, Switzerland
| |
Collapse
|
36
|
Ohsawa S, Vaughen J, Igaki T. Cell Extrusion: A Stress-Responsive Force for Good or Evil in Epithelial Homeostasis. Dev Cell 2018; 44:284-296. [PMID: 29408235 DOI: 10.1016/j.devcel.2018.01.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Epithelial tissues robustly respond to internal and external stressors via dynamic cellular rearrangements. Cell extrusion acts as a key regulator of epithelial homeostasis by removing apoptotic cells, orchestrating morphogenesis, and mediating competitive cellular battles during tumorigenesis. Here, we delineate the diverse functions of cell extrusion during development and disease. We emphasize the expanding role for apoptotic cell extrusion in exerting morphogenetic forces, as well as the strong intersection of cell extrusion with cell competition, a homeostatic mechanism that eliminates aberrant or unfit cells. While cell competition and extrusion can exert potent, tumor-suppressive effects, dysregulation of either critical homeostatic program can fuel cancer progression.
Collapse
Affiliation(s)
- Shizue Ohsawa
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - John Vaughen
- Department of Developmental Biology, Stanford School of Medicine, Beckman Center, 279 Campus Drive B300, Stanford, CA 94305, USA
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
37
|
Approximate Bayesian computation reveals the importance of repeated measurements for parameterising cell-based models of growing tissues. J Theor Biol 2018; 443:66-81. [PMID: 29391171 DOI: 10.1016/j.jtbi.2018.01.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 11/22/2022]
Abstract
The growth and dynamics of epithelial tissues govern many morphogenetic processes in embryonic development. A recent quantitative transition in data acquisition, facilitated by advances in genetic and live-imaging techniques, is paving the way for new insights to these processes. Computational models can help us understand and interpret observations, and then make predictions for future experiments that can distinguish between hypothesised mechanisms. Increasingly, cell-based modelling approaches such as vertex models are being used to help understand the mechanics underlying epithelial morphogenesis. These models typically seek to reproduce qualitative phenomena, such as cell sorting or tissue buckling. However, it remains unclear to what extent quantitative data can be used to constrain these models so that they can then be used to make quantitative, experimentally testable predictions. To address this issue, we perform an in silico study to investigate whether vertex model parameters can be inferred from imaging data, and explore methods to quantify the uncertainty of such estimates. Our approach requires the use of summary statistics to estimate parameters. Here, we focus on summary statistics of cellular packing and of laser ablation experiments, as are commonly reported from imaging studies. We find that including data from repeated experiments is necessary to generate reliable parameter estimates that can facilitate quantitative model predictions.
Collapse
|
38
|
Ma M, Cao X, Dai J, Pastor-Pareja JC. Basement Membrane Manipulation in Drosophila Wing Discs Affects Dpp Retention but Not Growth Mechanoregulation. Dev Cell 2017; 42:97-106.e4. [PMID: 28697337 DOI: 10.1016/j.devcel.2017.06.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/04/2017] [Accepted: 06/02/2017] [Indexed: 01/19/2023]
Abstract
Basement membranes (BMs) are extracellular matrix polymers basally underlying epithelia, where they regulate cell signaling and tissue mechanics. Constriction by the BM shapes Drosophila wing discs, a well-characterized model of tissue growth. Recently, the hypothesis that mechanical factors govern wing growth has received much attention, but it has not been definitively tested. In this study, we manipulated BM composition to cause dramatic changes in tissue tension. We found that increased tissue compression when perlecan was knocked down did not affect adult wing size. BM elimination, decreasing compression, reduced wing size but did not visibly affect Hippo signaling, widely postulated to mediate growth mechanoregulation. BM elimination, in contrast, attenuated signaling by bone morphogenetic protein/transforming growth factor β ligand Dpp, which was not efficiently retained within the tissue and escaped to the body cavity. Our results challenge mechanoregulation of wing growth, while uncovering a function of BMs in preserving a growth-promoting tissue environment.
Collapse
Affiliation(s)
- Mengqi Ma
- School of Life Sciences, Tsinghua University, Medical Science Building, D224, Beijing 100084, China
| | - Xueya Cao
- School of Life Sciences, Tsinghua University, Medical Science Building, D224, Beijing 100084, China
| | - Jianli Dai
- School of Life Sciences, Tsinghua University, Medical Science Building, D224, Beijing 100084, China
| | - José C Pastor-Pareja
- School of Life Sciences, Tsinghua University, Medical Science Building, D224, Beijing 100084, China.
| |
Collapse
|
39
|
Lu J, Wang D, Shen J. Hedgehog signalling is required for cell survival in Drosophila wing pouch cells. Sci Rep 2017; 7:11317. [PMID: 28900135 PMCID: PMC5595820 DOI: 10.1038/s41598-017-10550-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/10/2017] [Indexed: 11/09/2022] Open
Abstract
An appropriate balance between cell survival and cell death is essential for correct pattern formation in the animal tissues and organs. Previous studies have shown that the short-range signalling molecule Hedgehog (Hh) is required for cell proliferation and pattern formation in the Drosophila central wing discs. Signal transduction by one of the Hh targets, the morphogen Decapentaplegic (Dpp), is required for not only cell proliferation, but also cell survival in the pouch cells. However, Hh function in cell survival and cell death has not been revealed. Here, we found that loss of Hh signal activity induces considerable Caspase-dependent cell death in the wing pouch cells, and this process was independent of both Dpp signalling and Jun-N-terminal kinase (JNK) signalling. Loss of Hh induced activation of the pro-apoptotic gene hid and inhibition of diap1. Therefore, we identified an important role of Hh signalling in cell survival during Drosophila wing development.
Collapse
Affiliation(s)
- Juan Lu
- Department of Entomology, MOA Key Laboratory for monitoring and green management of crop pests, China Agricultural University, 100193, Beijing, China
| | - Dan Wang
- Department of Entomology, MOA Key Laboratory for monitoring and green management of crop pests, China Agricultural University, 100193, Beijing, China
| | - Jie Shen
- Department of Entomology, MOA Key Laboratory for monitoring and green management of crop pests, China Agricultural University, 100193, Beijing, China.
| |
Collapse
|
40
|
spalt is functionally conserved in Locusta and Drosophila to promote wing growth. Sci Rep 2017; 7:44393. [PMID: 28300136 PMCID: PMC5353606 DOI: 10.1038/srep44393] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/07/2017] [Indexed: 12/19/2022] Open
Abstract
Locusta has strong fly wings to ensure its long distance migration, but the molecular mechanism that regulates the Locusta wing development is poorly understood. To address the developmental mechanism of the Locusta flying wing, we cloned the Dpp target gene spalt (sal) and analyzed its function in wing growth in the Locusta. The Locusta wing size is apparently reduced with vein defects when sal is interfered by injection of dsRNA, indicating that sal is required for locust wing growth and vein formation. This function is conserved during the Drosophila wing development. To better understand sal’s function in wing growth, we then used Drosophila wing disc as a model for further study. We found that sal promotes cell proliferation in the whole wing disc via positive regulation of a microRNA bantam. Our results firstly unravel sal’s function in the Locusta wing growth and confirm a highly conserved function of sal in Locusta and Drosophila.
Collapse
|
41
|
Pflugfelder G, Eichinger F, Shen J. T-Box Genes in Drosophila Limb Development. Curr Top Dev Biol 2017; 122:313-354. [DOI: 10.1016/bs.ctdb.2016.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Liu S, Sun J, Wang D, Pflugfelder GO, Shen J. Fold formation at the compartment boundary of Drosophila wing requires Yki signaling to suppress JNK dependent apoptosis. Sci Rep 2016; 6:38003. [PMID: 27897227 PMCID: PMC5126554 DOI: 10.1038/srep38003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/02/2016] [Indexed: 12/18/2022] Open
Abstract
Compartment boundaries prevent cell populations of different lineage from intermingling. In many cases, compartment boundaries are associated with morphological folds. However, in the Drosophila wing imaginal disc, fold formation at the anterior/posterior (A/P) compartment boundary is suppressed, probably as a prerequisite for the formation of a flat wing surface. Fold suppression depends on optomotor-blind (omb). Omb mutant animals develop a deep apical fold at the A/P boundary of the larval wing disc and an A/P cleft in the adult wing. A/P fold formation is controlled by different signaling pathways. Jun N-terminal kinase (JNK) and Yorkie (Yki) signaling are activated in cells along the fold and are necessary for the A/P fold to develop. While JNK promotes cell shape changes and cell death, Yki target genes are required to antagonize apoptosis, explaining why both pathways need to be active for the formation of a stable fold.
Collapse
Affiliation(s)
- Suning Liu
- Department of Entomology, China Agricultural University, 100193 Beijing, China
| | - Jie Sun
- Department of Entomology, China Agricultural University, 100193 Beijing, China
| | - Dan Wang
- Department of Entomology, China Agricultural University, 100193 Beijing, China
| | - Gert O Pflugfelder
- Institute of Genetics, Johannes Gutenberg-University, 55128 Mainz, Germany
| | - Jie Shen
- Department of Entomology, China Agricultural University, 100193 Beijing, China
| |
Collapse
|
43
|
Merino MM, Levayer R, Moreno E. Survival of the Fittest: Essential Roles of Cell Competition in Development, Aging, and Cancer. Trends Cell Biol 2016; 26:776-788. [DOI: 10.1016/j.tcb.2016.05.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/20/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022]
|
44
|
Tang W, Wang D, Shen J. Asymmetric distribution of Spalt in Drosophila wing squamous and columnar epithelia ensures correct cell morphogenesis. Sci Rep 2016; 6:30236. [PMID: 27452716 PMCID: PMC4958983 DOI: 10.1038/srep30236] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 07/01/2016] [Indexed: 01/01/2023] Open
Abstract
The Drosophila wing imaginal disc is a sac-like structure that is composed of two opposing cell layers: peripodial epithelium (PE, also known as squamous epithelia) and disc proper (DP, also known as pseudostratified columnar epithelia). The molecular mechanism of cell morphogenesis has been well studied in the DP but not in the PE. Although proper Dpp signalling activity is required for proper PE formation, the detailed regulation mechanism is poorly understood. Here, we found that the Dpp target gene sal is only expressed in DP cells, not in PE cells, although pMad is present in the PE. Increasing Dpp signalling activity cannot activate Sal in PE cells. The absence of Sal in the PE is essential for PE formation. The ectopic expression of sal in PE cells is sufficient to increase the PE cell height. Down-regulation of sal in the DP reduced DP cell height. We further demonstrated that the known PE cell height regulator Lines, which can convert PE into a DP cell fate, is mediated by sal mis-activation in PE because sal-RNAi and lines co-expression largely restores PE cell morphology. By revealing the microtubule distribution, we demonstrated that Lines- and Sal-heightened PE cells are morphologically similar to the intermediate cell with cuboidal morphology.
Collapse
Affiliation(s)
- Wenqian Tang
- Department of Entomology, China Agricultural University, 100193 Beijing, China
| | - Dan Wang
- Department of Entomology, China Agricultural University, 100193 Beijing, China
| | - Jie Shen
- Department of Entomology, China Agricultural University, 100193 Beijing, China
| |
Collapse
|
45
|
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
|
46
|
Blander JM. Death in the intestinal epithelium-basic biology and implications for inflammatory bowel disease. FEBS J 2016; 283:2720-30. [PMID: 27250564 PMCID: PMC4956528 DOI: 10.1111/febs.13771] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/06/2016] [Accepted: 06/01/2016] [Indexed: 12/11/2022]
Abstract
Every 4-5 days, intestinal epithelial cells (IEC) are terminated as they reach the end of their life. This process ensures that the epithelium is comprised of the fittest cells that maintain an impermeable barrier to luminal contents and the gut microbiota, as well as the most metabolically able cells that conduct functions in nutrient absorption, digestion, and secretion of antimicrobial peptides. IEC are terminated by apical extrusion-or shedding-from the intestinal epithelial monolayer into the gut lumen. Whether death by apoptosis signals extrusion or death follows expulsion by younger IEC has been a matter of debate. Seemingly a minor detail, IEC death before or after apical extrusion bears weight on the potential contribution of apoptotic IEC to intestinal homeostasis as a consequence of their recognition by intestinal lamina propria phagocytes. In inflammatory bowel disease (IBD), excessive death is observed in the ileal and colonic epithelium. The precise mode of IEC death in IBD is not defined. A highly inflammatory milieu within the intestinal lamina propria, rich in the proinflammatory cytokine, TNF-α, increases IEC shedding and compromises barrier integrity fueling more inflammation. A milestone in the treatment of IBD, anti-TNF-α therapy, may promote mucosal healing by reversing increased and inflammation-associated IEC death. Understanding the biology and consequences of cell death in the intestinal epithelium is critical to the design of new avenues for IBD therapy.
Collapse
Affiliation(s)
- J. Magarian Blander
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
47
|
Sedzinski J, Hannezo E, Tu F, Biro M, Wallingford JB. Emergence of an Apical Epithelial Cell Surface In Vivo. Dev Cell 2016; 36:24-35. [PMID: 26766441 DOI: 10.1016/j.devcel.2015.12.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 12/22/2022]
Abstract
Epithelial sheets are crucial components of all metazoan animals, enclosing organs and protecting the animal from its environment. Epithelial homeostasis poses unique challenges, as addition of new cells and loss of old cells must be achieved without disrupting the fluid-tight barrier and apicobasal polarity of the epithelium. Several studies have identified cell biological mechanisms underlying extrusion of cells from epithelia, but far less is known of the converse mechanism by which new cells are added. Here, we combine molecular, pharmacological, and laser-dissection experiments with theoretical modeling to characterize forces driving emergence of an apical surface as single nascent cells are added to a vertebrate epithelium in vivo. We find that this process involves the interplay between cell-autonomous actin-generated pushing forces in the emerging cell and mechanical properties of neighboring cells. Our findings define the forces driving this cell behavior, contributing to a more comprehensive understanding of epithelial homeostasis.
Collapse
Affiliation(s)
- Jakub Sedzinski
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Edouard Hannezo
- Cavendish Laboratory, Department of Physics, University of Cambridge, J.J. Thomson Avenue, Cambridge CB3 0HE, UK; Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Fan Tu
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Maté Biro
- Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag 6, Newtown, NSW 2042, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - John B Wallingford
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA; Patterson Labs, University of Texas, 2401 Speedway, Austin, TX 78712, USA.
| |
Collapse
|
48
|
Gudipaty SA, Rosenblatt J. Epithelial cell extrusion: Pathways and pathologies. Semin Cell Dev Biol 2016; 67:132-140. [PMID: 27212253 DOI: 10.1016/j.semcdb.2016.05.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/13/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023]
Abstract
To remove dying or unwanted cells from an epithelium while preserving the barrier function of the layer, epithelia use a unique process called cell extrusion. To extrude, the cell fated to die emits the lipid Sphingosine 1 Phosphate (S1P), which binds the G-protein-coupled receptor Sphingosine 1 Phosphate receptor 2 (S1P2) in the neighboring cells that activates Rho-mediated contraction of an actomyosin ring circumferentially and basally. This contraction acts to squeeze the cell out apically while drawing together neighboring cells and preventing any gaps to the epithelial barrier. Epithelia can extrude out cells targeted to die by apoptotic stimuli to repair the barrier in the face of death or extrude live cells to promote cell death when epithelial cells become too crowded. Indeed, because epithelial cells naturally turn over by cell death and division at some of the highest rates in the body, epithelia depend on crowding-induced live cell extrusion to preserve constant cell numbers. If extrusion is defective, epithelial cells rapidly lose contact inhibition and form masses. Additionally, because epithelia act as the first line of defense in innate immunity, preservation of this barrier is critical for preventing pathogens from invading the body. Given its role in controlling constant cell numbers and maintaining barrier function, a number of different pathologies can result when extrusion is disrupted. Here, we review mechanisms and signaling pathways that control epithelial extrusion and discuss how defects in these mechanisms can lead to multiple diseases. We also discuss tactics pathogens have devised to hijack the extrusion process to infect and colonize epithelia.
Collapse
Affiliation(s)
- Swapna Aravind Gudipaty
- Department of Oncological Sciences, Huntsman Cancer Institute, University Of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA
| | - Jody Rosenblatt
- Department of Oncological Sciences, Huntsman Cancer Institute, University Of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA.
| |
Collapse
|
49
|
Amarnath S, Agarwala S. Cell-cycle-dependent TGFβ-BMP antagonism regulates neural tube closure by modulating tight junctions. J Cell Sci 2016; 130:119-131. [PMID: 27034139 DOI: 10.1242/jcs.179192] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 03/18/2016] [Indexed: 12/15/2022] Open
Abstract
Many organs form by invaginating and rolling flat epithelial cell sheets into tubes. Invagination of the ventral midline of the neural plate forms the median hinge point (MHP), an event that elevates the neural folds and is essential for neural tube closure (NTC). MHP formation involves dynamic spatiotemporal modulations of cell shape, but how these are achieved is not understood. Here, we show that cell-cycle-dependent BMP and TGFβ antagonism elicits MHP formation by dynamically regulating interactions between apical (PAR complex) and basolateral (LGL) polarity proteins. TGFβ and BMP-activated receptor (r)-SMADs [phosphorylated SMAD2 or SMAD3 (pSMAD2,3), or phosphorylated SMAD1, SMAD5 or SMAD8 (pSMAD1,5,8)] undergo cell-cycle-dependent modulations and nucleo-cytosolic shuttling along the apicobasal axis of the neural plate. Non-canonical TGFβ and BMP activity in the cytosol determines whether pSMAD2,3 or pSMAD1,5,8 associates with the tight junction (PAR complex) or with LGL, and whether cell shape changes can occur at the MHP. Thus, the interactions of BMP and TGFβ with polarity proteins dynamically modulate MHP formation by regulating r-SMAD competition for tight junctions and r-SMAD sequestration by LGL.
Collapse
Affiliation(s)
- Smita Amarnath
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Seema Agarwala
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA .,Institute for Cell and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA.,Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
50
|
Abstract
Widely implicated in human disease, abnormal cellular cysts reflect dramatic defects in the maintenance of epithelial integrity. A new study reports that epithelial cysts may arise as a surprisingly general consequence of clonal defects in the specification of cell identity.
Collapse
Affiliation(s)
- Yu-Ichiro Nakajima
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Matthew C Gibson
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA.
| |
Collapse
|