1
|
Matakatsu H, Fehon RG. Dachsous and Fat coordinately repress the Dachs-Dlish-Approximated complex to control growth. J Cell Biol 2024; 223:e202406119. [PMID: 39373700 PMCID: PMC11461286 DOI: 10.1083/jcb.202406119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/11/2024] [Indexed: 10/08/2024] Open
Abstract
Two protocadherins, Dachsous and Fat, regulate organ growth in Drosophila via the Hippo pathway. Dachsous and Fat bind heterotypically to regulate the abundance and subcellular localization of a "core complex" consisting of Dachs, Dlish, and Approximated. This complex localizes to the junctional cortex where it represses Warts. Dachsous is believed to promote growth by recruiting and stabilizing this complex, while Fat represses growth by promoting its degradation. Here, we examine the functional relationships between the intracellular domains of Dachsous and Fat and the core complex. While Dachsous promotes the accumulation of core complex proteins in puncta, it is not required for their assembly. Indeed, the core complex accumulates maximally in the absence of both Dachsous and Fat. Furthermore, Dachsous represses growth in the absence of Fat by removing the core complex from the junctional cortex. Fat similarly recruits core complex components but promotes their degradation. Our findings reveal that Dachsous and Fat coordinately constrain tissue growth by repressing the core complex.
Collapse
Affiliation(s)
- Hitoshi Matakatsu
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Richard G. Fehon
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Tripathi BK, Irvine KD. Contributions of the Dachsous intracellular domain to Dachsous-Fat signaling. Development 2024; 151:dev202919. [PMID: 39503213 PMCID: PMC11634027 DOI: 10.1242/dev.202919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
The protocadherins Fat and Dachsous regulate organ growth, shape, patterning, and planar cell polarity. Although Dachsous and Fat have been described as ligand and receptor, respectively, in a signal transduction pathway, there is also evidence for bidirectional signaling. Here, we assess signaling downstream of Dachsous through analysis of its intracellular domain. Genomic deletions of conserved sequences within dachsous identified regions of the intracellular domain that contribute to Dachsous activity. Deletion of the A motif increased Dachsous protein levels and decreased wing size. Deletion of the D motif decreased Dachsous levels at cell membranes, increased wing size, and disrupted wing, leg and hindgut patterning and planar cell polarity. Co-immunoprecipitation experiments established that the D motif is necessary and sufficient for association of Dachsous with key partners, including Lowfat, Dachs, Spiny-legs, Fat and MyoID. Subdivision of the D motif identified distinct regions that preferentially contribute to different Dachsous activities. Our results identify motifs that are essential for Dachsous function and are consistent with the hypothesis that the key function of Dachsous is regulation of Fat.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D. Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
3
|
Matakatsu H, Fehon RG. Dachsous and Fat coordinately repress the Dachs-Dlish-Approximated complex to control growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599638. [PMID: 38948705 PMCID: PMC11212998 DOI: 10.1101/2024.06.18.599638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Two protocadherins, Dachsous (Ds) and Fat (Ft), regulate organ growth in Drosophila via the Hippo pathway. Ds and Ft bind heterotypically to regulate the abundance and subcellular localization of a 'core complex' consisting of Dachs, Dlish and Approximated. This complex localizes to the junctional cortex where it promotes growth by repressing the pathway kinase Warts. Ds is believed to promote growth by recruiting and stabilizing the core complex at the junctional cortex, while Ft represses growth by promoting degradation of core complex components. Here, we examine the functions of intracellular domains of Ds and Ft and their relationship to the core complex. While Ds promotes accumulation of the core complex proteins in cortical puncta, it is not required for core complex assembly. Indeed, the core complex assembles maximally in the absence of both Ds and Ft. Furthermore, while Ds promotes growth in the presence of Ft, it represses growth in the absence of Ft by removing the core complex from the junctional cortex. Ft similarly recruits core complex components, however it normally promotes their degradation. Our findings reveal that Ds and Ft constrain tissue growth by repressing the default 'on' state of the core complex.
Collapse
|
4
|
Liu A, O’Connell J, Wall F, Carthew RW. Scaling between cell cycle duration and wing growth is regulated by Fat-Dachsous signaling in Drosophila. eLife 2024; 12:RP91572. [PMID: 38842917 PMCID: PMC11156469 DOI: 10.7554/elife.91572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024] Open
Abstract
The atypical cadherins Fat and Dachsous (Ds) signal through the Hippo pathway to regulate growth of numerous organs, including the Drosophila wing. Here, we find that Ds-Fat signaling tunes a unique feature of cell proliferation found to control the rate of wing growth during the third instar larval phase. The duration of the cell cycle increases in direct proportion to the size of the wing, leading to linear-like growth during the third instar. Ds-Fat signaling enhances the rate at which the cell cycle lengthens with wing size, thus diminishing the rate of wing growth. We show that this results in a complex but stereotyped relative scaling of wing growth with body growth in Drosophila. Finally, we examine the dynamics of Fat and Ds protein distribution in the wing, observing graded distributions that change during growth. However, the significance of these dynamics is unclear since perturbations in expression have negligible impact on wing growth.
Collapse
Affiliation(s)
- Andrew Liu
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
- NSF-Simons National Institute for Theory and Mathematics in BiologyChicagoUnited States
| | - Jessica O’Connell
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Farley Wall
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Richard W Carthew
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
- NSF-Simons National Institute for Theory and Mathematics in BiologyChicagoUnited States
| |
Collapse
|
5
|
Liu A, O’Connell J, Wall F, Carthew RW. Scaling between cell cycle duration and wing growth is regulated by Fat-Dachsous signaling in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.01.551465. [PMID: 38645118 PMCID: PMC11030236 DOI: 10.1101/2023.08.01.551465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The atypical cadherins Fat and Dachsous (Ds) signal through the Hippo pathway to regulate growth of numerous organs, including the Drosophila wing. Here, we find that Ds-Fat signaling tunes a unique feature of cell proliferation found to control the rate of wing growth during the third instar larval phase. The duration of the cell cycle increases in direct proportion to the size of the wing, leading to linear-like growth during the third instar. Ds-Fat signaling enhances the rate at which the cell cycle lengthens with wing size, thus diminishing the rate of wing growth. We show that this results in a complex but stereotyped relative scaling of wing growth with body growth in Drosophila. Finally, we examine the dynamics of Fat and Ds protein distribution in the wing, observing graded distributions that change during growth. However, the significance of these dynamics is unclear since perturbations in expression have negligible impact on wing growth.
Collapse
Affiliation(s)
- Andrew Liu
- Department of Molecular Biosciences, Northwestern University, Evanston IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston IL
| | - Jessica O’Connell
- Department of Molecular Biosciences, Northwestern University, Evanston IL
| | - Farley Wall
- Department of Molecular Biosciences, Northwestern University, Evanston IL
| | - Richard W. Carthew
- Department of Molecular Biosciences, Northwestern University, Evanston IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston IL
| |
Collapse
|
6
|
Tripathi BK, Irvine KD. Contributions of the Dachsous intracellular domain to Dachsous-Fat signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587940. [PMID: 38617303 PMCID: PMC11014530 DOI: 10.1101/2024.04.03.587940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The protocadherins Fat and Dachsous regulate organ growth, shape, patterning, and planar cell polarity. Although Dachsous and Fat have been described as ligand and receptor, respectively, in a signal transduction pathway, there is also evidence for bidirectional signaling. Here we assess signaling downstream of Dachsous through analysis of its intracellular domain. Genomic deletions of conserved sequences within dachsous identified regions of the intracellular domain required for normal development. Deletion of the A motif increased Dachsous protein levels and decreased wing size. Deletion of the D motif decreased Dachsous levels at cell membranes, increased wing size, and disrupted wing, leg and hindgut patterning and planar cell polarity. Co-immunoprecipitation experiments established that the D motif is necessary and sufficient for association of Dachsous with four key partners: Lowfat, Dachs, Spiny-legs, and MyoID. Subdivision of the D motif identified distinct regions that are preferentially responsible for association with Lft versus Dachs. Our results identify motifs that are essential for Dachsous function and are consistent with the hypothesis that the key function of Dachsous is regulation of Fat.
Collapse
|
7
|
Nita A, Moroishi T. Hippo pathway in cell-cell communication: emerging roles in development and regeneration. Inflamm Regen 2024; 44:18. [PMID: 38566194 PMCID: PMC10986044 DOI: 10.1186/s41232-024-00331-8] [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: 01/25/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
The Hippo pathway is a central regulator of tissue growth that has been widely studied in mammalian organ development, regeneration, and cancer biology. Although previous studies have convincingly revealed its cell-autonomous functions in controlling cell fate, such as cell proliferation, survival, and differentiation, accumulating evidence in recent years has revealed its non-cell-autonomous functions. This pathway regulates cell-cell communication through direct interactions, soluble factors, extracellular vesicles, and the extracellular matrix, providing a range of options for controlling diverse biological processes. Consequently, the Hippo pathway not only dictates the fate of individual cells but also triggers multicellular responses involving both tissue-resident cells and infiltrating immune cells. Here, we have highlighted the recent understanding of the molecular mechanisms by which the Hippo pathway controls cell-cell communication and discuss its importance in tissue homeostasis, especially in development and regeneration.
Collapse
Affiliation(s)
- Akihiro Nita
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
8
|
Gou J, Zhang T, Othmer HG. The Interaction of Mechanics and the Hippo Pathway in Drosophila melanogaster. Cancers (Basel) 2023; 15:4840. [PMID: 37835534 PMCID: PMC10571775 DOI: 10.3390/cancers15194840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/10/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
Drosophila melanogaster has emerged as an ideal system for studying the networks that control tissue development and homeostasis and, given the similarity of the pathways involved, controlled and uncontrolled growth in mammalian systems. The signaling pathways used in patterning the Drosophila wing disc are well known and result in the emergence of interaction of these pathways with the Hippo signaling pathway, which plays a central role in controlling cell proliferation and apoptosis. Mechanical effects are another major factor in the control of growth, but far less is known about how they exert their control. Herein, we develop a mathematical model that integrates the mechanical interactions between cells, which occur via adherens and tight junctions, with the intracellular actin network and the Hippo pathway so as to better understand cell-autonomous and non-autonomous control of growth in response to mechanical forces.
Collapse
Affiliation(s)
- Jia Gou
- Department of Mathematics, University of California, Riverside, CA 92507, USA;
| | - Tianhao Zhang
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Hans G. Othmer
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
9
|
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
|
10
|
Kowalczyk W, Romanelli L, Atkins M, Hillen H, Bravo González-Blas C, Jacobs J, Xie J, Soheily S, Verboven E, Moya IM, Verhulst S, de Waegeneer M, Sansores-Garcia L, van Huffel L, Johnson RL, van Grunsven LA, Aerts S, Halder G. Hippo signaling instructs ectopic but not normal organ growth. Science 2022; 378:eabg3679. [DOI: 10.1126/science.abg3679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The Hippo signaling pathway is widely considered a master regulator of organ growth because of the prominent overgrowth phenotypes caused by experimental manipulation of its activity. Contrary to this model, we show here that removing Hippo transcriptional output did not impair the ability of the mouse liver and
Drosophila
eyes to grow to their normal size. Moreover, the transcriptional activity of the Hippo pathway effectors Yap/Taz/Yki did not correlate with cell proliferation, and hyperactivation of these effectors induced gene expression programs that did not recapitulate normal development. Concordantly, a functional screen in
Drosophila
identified several Hippo pathway target genes that were required for ectopic overgrowth but not normal growth. Thus, Hippo signaling does not instruct normal growth, and the Hippo-induced overgrowth phenotypes are caused by the activation of abnormal genetic programs.
Collapse
Affiliation(s)
- W. Kowalczyk
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - L. Romanelli
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - M. Atkins
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Biological Sciences, Sam Houston State University, Huntsville, TX, USA
| | - H. Hillen
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - C. Bravo González-Blas
- VIB Center for Brain and Disease Research and KU Leuven Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - J. Jacobs
- VIB Center for Brain and Disease Research and KU Leuven Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - J. Xie
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - S. Soheily
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - E. Verboven
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - I. M. Moya
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
- Facultad de Ingeniería y Ciencias Aplicadas, Universidad de Las Américas, Quito, Ecuador
| | - S. Verhulst
- Department for Cell Biology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussel-Jette, Belgium
| | - M. de Waegeneer
- VIB Center for Brain and Disease Research and KU Leuven Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - L. Sansores-Garcia
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - L. van Huffel
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - R. L. Johnson
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L. A. van Grunsven
- Department for Cell Biology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussel-Jette, Belgium
| | - S. Aerts
- VIB Center for Brain and Disease Research and KU Leuven Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - G. Halder
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Chorro A, Verma B, Homfeldt M, Ibáñez B, Lawrence PA, Casal J. Planar cell polarity: intracellular asymmetry and supracellular gradients of Dachsous. Open Biol 2022; 12:220195. [PMID: 36476047 PMCID: PMC9554717 DOI: 10.1098/rsob.220195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The slope of a supracellular molecular gradient has long been thought to orient and coordinate planar cell polarity (PCP). Here we demonstrate and measure that gradient. Dachsous (Ds) is a conserved and elemental molecule of PCP; Ds forms intercellular bridges with another cadherin molecule, Fat (Ft), an interaction modulated by the Golgi protein Four-jointed (Fj). Using genetic mosaics and tagged Ds, we measure Ds in vivo in membranes of individual cells over a whole metamere of the Drosophila abdomen. We find as follows. (i) A supracellular gradient rises from head to tail in the anterior compartment (A) and then falls in the posterior compartment (P). (ii) There is more Ds in the front than the rear membranes of all cells in the A compartment, except that compartment's most anterior and most posterior cells. There is more Ds in the rear than in the front membranes of all cells of the P compartment. (iii) The loss of Fj removes intracellular asymmetry anteriorly in the segment and reduces it elsewhere. Additional experiments show that Fj makes PCP more robust. Using Dachs (D) as a molecular indicator of polarity, we confirm that opposing gradients of PCP meet slightly out of register with compartment boundaries.
Collapse
Affiliation(s)
- Adrià Chorro
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Bhavna Verma
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Maylin Homfeldt
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Beatríz Ibáñez
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Peter A. Lawrence
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - José Casal
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| |
Collapse
|
12
|
Brittle A, Warrington SJ, Strutt H, Manning E, Tan SE, Strutt D. Distinct mechanisms of planar polarization by the core and Fat-Dachsous planar polarity pathways in the Drosophila wing. Cell Rep 2022; 40:111419. [PMID: 36170824 PMCID: PMC9631118 DOI: 10.1016/j.celrep.2022.111419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Planar polarity describes the coordinated polarization of cells within a tissue plane, and in animals can be determined by the “core” or Fat-Dachsous pathways. Current models for planar polarity establishment involve two components: tissue-level “global” cues that determine the overall axis of polarity and cell-level feedback-mediated cellular polarity amplification. Here, we investigate the contributions of global cues versus cellular feedback amplification in the core and Fat-Dachsous pathways during Drosophila pupal wing development. We present evidence that these pathways generate planar polarity via distinct mechanisms. Core pathway function is consistent with strong feedback capable of self-organizing cell polarity, which can then be aligned with the tissue axis via weak or transient global cues. Conversely, generation of cell polarity by the Ft-Ds pathway depends on strong global cues in the form of graded patterns of gene expression, which can then be amplified by weak feedback mechanisms. The core and Fat-Dachsous planar polarity pathways function via distinct mechanisms The core can self-organize planar polarity and be oriented by weak upstream cues Fat-Dachsous are oriented by strong gradient cues but show poor self-organization
Collapse
Affiliation(s)
- Amy Brittle
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | | | - Helen Strutt
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Elizabeth Manning
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Su Ee Tan
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - David Strutt
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| |
Collapse
|
13
|
McKenna KZ, Nijhout HF. The development of shape. Modular control of growth in the lepidopteran forewing. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2022; 338:170-180. [PMID: 34710273 DOI: 10.1002/jez.b.23101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/08/2021] [Accepted: 10/11/2021] [Indexed: 12/28/2022]
Abstract
The mechanisms by which tissues and organs achieve their final size and shape during development are largely unknown. Although we have learned much about the mechanisms that control growth, little is known about how those play out to achieve a structure's specific final size and shape. The wings of insects are attractive systems for the study of the control of morphogenesis, because they are perfectly flat and two-dimensional, composed of two closely appressed cellular monolayers in which morphogenetic processes can be easily visualized. The wings of Lepidoptera arise from imaginal disks whose structure is always perfectly congruent with that of the adult wing, so that it is possible to fate-map corresponding positions on the larval disk to those of the adult wing. Here we show that the forewing imaginal disks of Junonia coenia are subdivided into four domains, with characteristic patterns of expression of known patterning genes Spalt (Sal), Engrailed (En), and Cubitus interruptus (Ci). We show that DNA and protein synthesis, as well as mitoses, are spatially patterned in a domain-specific way. Knockdown of Sal and En using produced domain-specific reductions in the shape of the forewing. Knockdown of signaling pathways involved in the regulation of growth likewise altered the shape of the forewing in a domain-specific way. Our results reveal a multi-level regulation of forewing shape involving hormones and growth-regulating genes.
Collapse
|
14
|
Gridnev A, Misra JR. Emerging Mechanisms of Growth and Patterning Regulation by Dachsous and Fat Protocadherins. Front Cell Dev Biol 2022; 10:842593. [PMID: 35372364 PMCID: PMC8967653 DOI: 10.3389/fcell.2022.842593] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/08/2022] [Indexed: 01/14/2023] Open
Abstract
Dachsous (Ds) and Fat are evolutionarily conserved cell adhesion molecules that play a critical role in development of multiple organ systems, where they coordinate tissue growth and morphogenesis. Much of our understanding of Ds-Fat signaling pathway comes from studies in Drosophila, where they initiate a signaling pathway that regulate growth by influencing Hippo signaling and morphogenesis by regulating Planar Cell Polarity (PCP). In this review, we discuss recent advances in our understanding of the mechanisms by which Ds-Fat signaling pathway regulates these critical developmental processes. Further, we discuss the progress in our understanding about how they function in mammals.
Collapse
|
15
|
Abstract
The Drosophila wing imaginal disc is a tissue of undifferentiated cells that are precursors of the wing and most of the notum of the adult fly. The wing disc first forms during embryogenesis from a cluster of ∼30 cells located in the second thoracic segment, which invaginate to form a sac-like structure. They undergo extensive proliferation during larval stages to form a mature larval wing disc of ∼35,000 cells. During this time, distinct cell fates are assigned to different regions, and the wing disc develops a complex morphology. Finally, during pupal stages the wing disc undergoes morphogenetic processes and then differentiates to form the adult wing and notum. While the bulk of the wing disc comprises epithelial cells, it also includes neurons and glia, and is associated with tracheal cells and muscle precursor cells. The relative simplicity and accessibility of the wing disc, combined with the wealth of genetic tools available in Drosophila, have combined to make it a premier system for identifying genes and deciphering systems that play crucial roles in animal development. Studies in wing imaginal discs have made key contributions to many areas of biology, including tissue patterning, signal transduction, growth control, regeneration, planar cell polarity, morphogenesis, and tissue mechanics.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
16
|
Moore KS, Moore R, Fulmer DB, Guo L, Gensemer C, Stairley R, Glover J, Beck TC, Morningstar JE, Biggs R, Muhkerjee R, Awgulewitsch A, Norris RA. DCHS1, Lix1L, and the Septin Cytoskeleton: Molecular and Developmental Etiology of Mitral Valve Prolapse. J Cardiovasc Dev Dis 2022; 9:62. [PMID: 35200715 PMCID: PMC8874669 DOI: 10.3390/jcdd9020062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Mitral valve prolapse (MVP) is a common cardiac valve disease that often progresses to serious secondary complications requiring surgery. MVP manifests as extracellular matrix disorganization and biomechanically incompetent tissues in the adult setting. However, MVP has recently been shown to have a developmental basis, as multiple causal genes expressed during embryonic development have been identified. Disease phenotypes have been observed in mouse models with human MVP mutations as early as birth. This study focuses on the developmental function of DCHS1, one of the first genes to be shown as causal in multiple families with non-syndromic MVP. By using various biochemical techniques as well as mouse and cell culture models, we demonstrate a unique link between DCHS1-based cell adhesions and the septin-actin cytoskeleton through interactions with cytoplasmic protein Lix1-Like (LIX1L). This DCHS1-LIX1L-SEPT9 axis interacts with and promotes filamentous actin organization to direct cell-ECM alignment and valve tissue shape.
Collapse
Affiliation(s)
- Kelsey S. Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Reece Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Diana B. Fulmer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Lilong Guo
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Cortney Gensemer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Rebecca Stairley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Janiece Glover
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Tyler C. Beck
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Jordan E. Morningstar
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Rachel Biggs
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Rupak Muhkerjee
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Alexander Awgulewitsch
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.S.M.); (R.M.); (L.G.); (C.G.); (R.S.); (J.G.); (T.C.B.); (J.E.M.); (R.B.); (A.A.)
| |
Collapse
|
17
|
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: 2.8] [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
|
18
|
Zhu H. Elucidate growth control mechanisms using reconstructed spatiotemporal distributions of signaling events. Comput Struct Biotechnol J 2021; 19:3618-3627. [PMID: 34257840 PMCID: PMC8249872 DOI: 10.1016/j.csbj.2021.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/19/2021] [Accepted: 06/12/2021] [Indexed: 11/30/2022] Open
Abstract
A fundamental biological question is how diverse and complex signaling and patterning is controlled correctly to generate distinct tissues, organs, and body plans, but incorrectly in diseased cells and tissues. Signaling pathways important for growth control have been identified, but to identify the mechanisms their transient and context-dependent interactions encode is more difficult. Currently computational systems biology aims to infer the control mechanisms by investigating quantitative changes of gene expression and protein concentrations, but this inference is difficult in nature. We propose it is desirable to explicitly simulate events and orders of gene regulation and protein interactions, which better elucidate control mechanisms, and report a method and tool with three examples. The Drosophila wing model includes Wnt, PCP, and Hippo pathways and mechanical force, incorporates well-confirmed experimental findings, and generates novel results. The other two examples illustrate the building of three-dimensional and large-scale models. These examples support that reconstructed spatiotemporal distributions of key signaling events help elucidate growth control mechanisms. As biologists pay increasing attention to disordered signaling in diseased cells, to develop new modeling methods and tools for conducting new computational studies is important.
Collapse
Affiliation(s)
- Hao Zhu
- Bioinformatics Section, School of Basic Medical Sciences, Southern Medical University, Shatai Road, Guangzhou 510515, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
19
|
The novel FAT4 activator jujuboside A suppresses NSCLC tumorigenesis by activating HIPPO signaling and inhibiting YAP nuclear translocation. Pharmacol Res 2021; 170:105723. [PMID: 34116210 DOI: 10.1016/j.phrs.2021.105723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/29/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022]
Abstract
FAT atypical cadherin 4 (FAT4) has been identified as a tumor suppressor in lung cancers. However, no agent for lung cancer treatment targeting FAT4 has been used in the clinic. Jujuboside A (JUA) is a major active compound in Semen Ziziphi Spinosae. Semen Ziziphi Spinosae is a traditional Chinese herbal medicine used clinically for tumor treatment to improve patients' quality of life. However, the anti-lung cancer activity and the underlying mechanisms of JUA are not yet fully understood. Here, we demonstrated the anti-lung cancer activity of JUA in two lung cancer mice models and three non-small cell lung cancer (NSCLC) cell lines, and further illustrated its underlying mechanisms. JUA suppressed the occurrence and development of lung cancer and extended mice survival in vivo, and suppressed NSCLC cell activities through cell cycle arrest, proliferation suppression, stemness inhibition and senescence promotion. Moreover, JUA directly bound with and activated FAT4, subsequently activating FAT4-HIPPO signaling and inhibiting YAP nuclear translocation. Knockdown of FAT4 diminished JUA's effects on HIPPO signaling, YAP nuclear translocation, cell proliferation and cellular senescence. In conclusion, JUA significantly suppressed NSCLC tumorigenesis by regulating FAT4-HIPPO-YAP signaling. Our findings suggest that JUA is a novel FAT4 activator that can be developed as a promising NSCLC therapeutic agent targeting the FAT4-HIPPO-YAP pathway.
Collapse
|
20
|
Yi C, Spitters TWGM, Al-Far EADA, Wang S, Xiong T, Cai S, Yan X, Guan K, Wagner M, El-Armouche A, Antos CL. A calcineurin-mediated scaling mechanism that controls a K +-leak channel to regulate morphogen and growth factor transcription. eLife 2021; 10:e60691. [PMID: 33830014 PMCID: PMC8110307 DOI: 10.7554/elife.60691] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 04/07/2021] [Indexed: 01/10/2023] Open
Abstract
The increase in activity of the two-pore potassium-leak channel Kcnk5b maintains allometric juvenile growth of adult zebrafish appendages. However, it remains unknown how this channel maintains allometric growth and how its bioelectric activity is regulated to scale these anatomical structures. We show the activation of Kcnk5b is sufficient to activate several genes that are part of important development programs. We provide in vivo transplantation evidence that the activation of gene transcription is cell autonomous. We also show that Kcnk5b will induce the expression of different subsets of the tested developmental genes in different cultured mammalian cell lines, which may explain how one electrophysiological stimulus can coordinately regulate the allometric growth of diverse populations of cells in the fin that use different developmental signals. We also provide evidence that the post-translational modification of serine 345 in Kcnk5b by calcineurin regulates channel activity to scale the fin. Thus, we show how an endogenous bioelectric mechanism can be regulated to promote coordinated developmental signaling to generate and scale a vertebrate appendage.
Collapse
Affiliation(s)
- Chao Yi
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of SciencesShanghaiChina
| | - Tim WGM Spitters
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
| | | | - Sen Wang
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of SciencesShanghaiChina
| | - TianLong Xiong
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of SciencesShanghaiChina
| | - Simian Cai
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
| | - Xin Yan
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
| | - Kaomei Guan
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
| | - Michael Wagner
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
- Klinik für Innere Medizin und Kardiologie, Herzzentrum Dresden, Technische Universität DresdenDresdenGermany
| | - Ali El-Armouche
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
| | - Christopher L Antos
- School of Life Sciences and Technology, ShanghaiTech UniversityShanghaiChina
- Institut für Pharmakologie und Toxikologie, Technische Universität DresdenDresdenGermany
| |
Collapse
|
21
|
Zecca M, Struhl G. A unified mechanism for the control of Drosophila wing growth by the morphogens Decapentaplegic and Wingless. PLoS Biol 2021; 19:e3001111. [PMID: 33657096 PMCID: PMC8148325 DOI: 10.1371/journal.pbio.3001111] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 05/25/2021] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
Development of the Drosophila wing-a paradigm of organ development-is governed by 2 morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Both proteins are produced by defined subpopulations of cells and spread outwards, forming gradients that control gene expression and cell pattern as a function of concentration. They also control growth, but how is unknown. Most studies have focused on Dpp and yielded disparate models in which cells throughout the wing grow at similar rates in response to the grade or temporal change in Dpp concentration or to the different amounts of Dpp "equalized" by molecular or mechanical feedbacks. In contrast, a model for Wg posits that growth is governed by a progressive expansion in morphogen range, via a mechanism in which a minimum threshold of Wg sustains the growth of cells within the wing and recruits surrounding "pre-wing" cells to grow and enter the wing. This mechanism depends on the capacity of Wg to fuel the autoregulation of vestigial (vg)-the selector gene that specifies the wing state-both to sustain vg expression in wing cells and by a feed-forward (FF) circuit of Fat (Ft)/Dachsous (Ds) protocadherin signaling to induce vg expression in neighboring pre-wing cells. Here, we have subjected Dpp to the same experimental tests used to elucidate the Wg model and find that it behaves indistinguishably. Hence, we posit that both morphogens act together, via a common mechanism, to control wing growth as a function of morphogen range.
Collapse
Affiliation(s)
- Myriam Zecca
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Gary Struhl
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| |
Collapse
|
22
|
Greenfeld H, Lin J, Mullins MC. The BMP signaling gradient is interpreted through concentration thresholds in dorsal-ventral axial patterning. PLoS Biol 2021; 19:e3001059. [PMID: 33481775 PMCID: PMC7857602 DOI: 10.1371/journal.pbio.3001059] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/03/2021] [Accepted: 01/07/2021] [Indexed: 12/24/2022] Open
Abstract
Bone Morphogenetic Protein (BMP) patterns the dorsal–ventral (DV) embryonic axis in all vertebrates, but it is unknown how cells along the DV axis interpret and translate the gradient of BMP signaling into differential gene activation that will give rise to distinct cell fates. To determine the mechanism of BMP morphogen interpretation in the zebrafish gastrula, we identified 57 genes that are directly activated by BMP signaling. By using Seurat analysis of single-cell RNA sequencing (scRNA-seq) data, we found that these genes are expressed in at least 3 distinct DV domains of the embryo. We distinguished between 3 models of BMP signal interpretation in which cells activate distinct gene expression through interpretation of thresholds of (1) the BMP signaling gradient slope; (2) the BMP signal duration; or (3) the level of BMP signal activation. We tested these 3 models using quantitative measurements of phosphorylated Smad5 (pSmad5) and by examining the spatial relationship between BMP signaling and activation of different target genes at single-cell resolution across the embryo. We found that BMP signaling gradient slope or BMP exposure duration did not account for the differential target gene expression domains. Instead, we show that cells respond to 3 distinct levels of BMP signaling activity to activate and position target gene expression. Together, we demonstrate that distinct pSmad5 threshold levels activate spatially distinct target genes to pattern the DV axis. This study tested three models of how a BMP morphogen gradient is translated into differential gene activation that specifies distinct cell fates, finding that BMP signal concentration thresholds, not gradient shape or signal duration, position three distinct gene activation domains.
Collapse
Affiliation(s)
- Hannah Greenfeld
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Jerome Lin
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Mary C. Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
23
|
Xu L, Zhang J, Zhan A, Wang Y, Ma X, Jie W, Cao Z, Omar MAA, He K, Li F. Identification and Analysis of MicroRNAs Associated with Wing Polyphenism in the Brown Planthopper, Nilaparvata lugens. Int J Mol Sci 2020; 21:E9754. [PMID: 33371331 PMCID: PMC7767257 DOI: 10.3390/ijms21249754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 12/27/2022] Open
Abstract
Many insects are capable of developing two types of wings (i.e., wing polyphenism) to adapt to various environments. Though the roles of microRNAs (miRNAs) in regulating animal growth and development have been well studied, their potential roles in modulating wing polyphenism remain largely elusive. To identify wing polyphenism-related miRNAs, we isolated small RNAs from 1st to 5th instar nymphs of long-wing (LW) and short-wing (SW) strains of the brown planthopper (BPH), Nilaparvata lugens. Small RNA libraries were then constructed and sequenced, yielding 158 conserved and 96 novel miRNAs. Among these, 122 miRNAs were differentially expressed between the two BPH strains. Specifically, 47, 2, 27 and 41 miRNAs were more highly expressed in the 1st, 3rd, 4th and 5th instars, respectively, of the LW strain compared with the SW strain. In contrast, 47, 3, 29 and 25 miRNAs were more highly expressed in the 1st, 3rd, 4th and 5th instars, respectively, of the SW strain compared with the LW strain. Next, we predicted the targets of these miRNAs and carried out Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis. We found that a number of pathways might be involved in wing form determination, such as the insulin, MAPK, mTOR, FoxO and thyroid hormone signaling pathways and the thyroid hormone synthesis pathway. Thirty and 45 differentially expressed miRNAs targeted genes in the insulin signaling and insect hormone biosynthesis pathways, respectively, which are related to wing dimorphism. Among these miRNAs, Nlu-miR-14-3p, Nlu-miR-9a-5p and Nlu-miR-315-5p, were confirmed to interact with insulin receptors (NlInRs) in dual luciferase reporter assays. These discoveries are helpful for understanding the miRNA-mediated regulatory mechanism of wing polyphenism in BPHs and shed new light on how insects respond to environmental cues through developmental plasticity.
Collapse
Affiliation(s)
- Le Xu
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (L.X.); (A.Z.); (X.M.); (Z.C.); (M.A.A.O.); (F.L.)
| | - Jiao Zhang
- College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China; (J.Z.); (W.J.)
| | - Anran Zhan
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (L.X.); (A.Z.); (X.M.); (Z.C.); (M.A.A.O.); (F.L.)
| | - Yaqin Wang
- State Key Laboratory of Rice Biology, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China;
| | - Xingzhou Ma
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (L.X.); (A.Z.); (X.M.); (Z.C.); (M.A.A.O.); (F.L.)
- College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China; (J.Z.); (W.J.)
| | - Wencai Jie
- College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China; (J.Z.); (W.J.)
| | - Zhenghong Cao
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (L.X.); (A.Z.); (X.M.); (Z.C.); (M.A.A.O.); (F.L.)
| | - Mohamed A. A. Omar
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (L.X.); (A.Z.); (X.M.); (Z.C.); (M.A.A.O.); (F.L.)
- Department of Plant Protection, Faculty of Agriculture (Saba Basha), Alexandria University, Alexandria 21531, Egypt
| | - Kang He
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (L.X.); (A.Z.); (X.M.); (Z.C.); (M.A.A.O.); (F.L.)
| | - Fei Li
- State Key Laboratory of Rice Biology & Ministry of Agricultural and Rural Affairs Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (L.X.); (A.Z.); (X.M.); (Z.C.); (M.A.A.O.); (F.L.)
| |
Collapse
|
24
|
Pietra S, Ng K, Lawrence PA, Casal J. Planar cell polarity in the larval epidermis of Drosophila and the role of microtubules. Open Biol 2020; 10:200290. [PMID: 33295841 PMCID: PMC7776564 DOI: 10.1098/rsob.200290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We investigate planar cell polarity (PCP) in the Drosophila larval epidermis. The intricate pattern of denticles depends on only one system of PCP, the Dachsous/Fat system. Dachsous molecules in one cell bind to Fat molecules in a neighbour cell to make intercellular bridges. The disposition and orientation of these Dachsous–Fat bridges allows each cell to compare two neighbours and point its denticles towards the neighbour with the most Dachsous. Measurements of the amount of Dachsous reveal a peak at the back of the anterior compartment of each segment. Localization of Dachs and orientation of ectopic denticles help reveal the polarity of every cell. We discuss whether these findings support our gradient model of Dachsous activity. Several groups have proposed that Dachsous and Fat fix the direction of PCP via oriented microtubules that transport PCP proteins to one side of the cell. We test this proposition in the larval cells and find that most microtubules grow perpendicularly to the axis of PCP. We find no meaningful bias in the polarity of microtubules aligned close to that axis. We also reexamine published data from the pupal abdomen and find no evidence supporting the hypothesis that microtubular orientation draws the arrow of PCP.
Collapse
Affiliation(s)
- Stefano Pietra
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - KangBo Ng
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Peter A Lawrence
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - José Casal
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| |
Collapse
|
25
|
Control of Drosophila wing size by morphogen range and hormonal gating. Proc Natl Acad Sci U S A 2020; 117:31935-31944. [PMID: 33257577 DOI: 10.1073/pnas.2018196117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The stereotyped dimensions of animal bodies and their component parts result from tight constraints on growth. Yet, the mechanisms that stop growth when organs reach the right size are unknown. Growth of the Drosophila wing-a classic paradigm-is governed by two morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Wing growth during larval life ceases when the primordium attains full size, concomitant with the larval-to-pupal molt orchestrated by the steroid hormone ecdysone. Here, we block the molt by genetically dampening ecdysone production, creating an experimental paradigm in which the wing stops growing at the correct size while the larva continues to feed and gain body mass. Under these conditions, we show that wing growth is limited by the ranges of Dpp and Wg, and by ecdysone, which regulates the cellular response to their signaling activities. Further, we present evidence that growth terminates because of the loss of two distinct modes of morphogen action: 1) maintenance of growth within the wing proper and 2) induced growth of surrounding "pre-wing" cells and their recruitment into the wing. Our results provide a precedent for the control of organ size by morphogen range and the hormonal gating of morphogen action.
Collapse
|
26
|
Vea IM, Shingleton AW. Network-regulated organ allometry: The developmental regulation of morphological scaling. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e391. [PMID: 32567243 DOI: 10.1002/wdev.391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/30/2020] [Accepted: 05/23/2020] [Indexed: 12/11/2022]
Abstract
Morphological scaling relationships, or allometries, describe how traits grow coordinately and covary among individuals in a population. The developmental regulation of scaling is essential to generate correctly proportioned adults across a range of body sizes, while the mis-regulation of scaling may result in congenital birth defects. Research over several decades has identified the developmental mechanisms that regulate the size of individual traits. Nevertheless, we still have poor understanding of how these mechanisms work together to generate correlated size variation among traits in response to environmental and genetic variation. Conceptually, morphological scaling can be generated by size-regulatory factors that act directly on multiple growing traits (trait-autonomous scaling), or indirectly via hormones produced by central endocrine organs (systemically regulated scaling), and there are a number of well-established examples of such mechanisms. There is much less evidence, however, that genetic and environmental variation actually acts on these mechanisms to generate morphological scaling in natural populations. More recent studies indicate that growing organs can themselves regulate the growth of other organs in the body. This suggests that covariation in trait size can be generated by network-regulated scaling mechanisms that respond to changes in the growth of individual traits. Testing this hypothesis, and one of the main challenges of understanding morphological scaling, requires connecting mechanisms elucidated in the laboratory with patterns of scaling observed in the natural world. This article is categorized under: Establishment of Spatial and Temporal Patterns > Regulation of Size, Proportion, and Timing Comparative Development and Evolution > Organ System Comparisons Between Species.
Collapse
Affiliation(s)
- Isabelle M Vea
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Alexander W Shingleton
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
27
|
Gou J, Stotsky JA, Othmer HG. Growth control in the Drosophila wing disk. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1478. [PMID: 31917525 DOI: 10.1002/wsbm.1478] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
The regulation of size and shape is a fundamental requirement of biological development and has been a subject of scientific study for centuries, but we still lack an understanding of how organisms know when to stop growing. Imaginal wing disks of the fruit fly Drosophila melanogaster, which are precursors of the adult wings, are an archetypal tissue for studying growth control. The growth of the disks is dependent on many inter- and intra-organ factors such as morphogens, mechanical forces, nutrient levels, and hormones that influence gene expression and cell growth. Extracellular signals are transduced into gene-control signals via complex signal transduction networks, and since cells typically receive many different signals, a mechanism for integrating the signals is needed. Our understanding of the effect of morphogens on tissue-level growth regulation via individual pathways has increased significantly in the last half century, but our understanding of how multiple biochemical and mechanical signals are integrated to determine whether or not a cell decides to divide is still rudimentary. Numerous fundamental questions are involved in understanding the decision-making process, and here we review the major biochemical and mechanical pathways involved in disk development with a view toward providing a basis for beginning to understand how multiple signals can be integrated at the cell level, and how this translates into growth control at the level of the imaginal disk. This article is categorized under: Analytical and Computational Methods > Computational Methods Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Jia Gou
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Jay A Stotsky
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
28
|
Simsek MF, Özbudak EM. Spatial Fold Change of FGF Signaling Encodes Positional Information for Segmental Determination in Zebrafish. Cell Rep 2019; 24:66-78.e8. [PMID: 29972792 PMCID: PMC6063364 DOI: 10.1016/j.celrep.2018.06.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/08/2018] [Accepted: 06/05/2018] [Indexed: 01/15/2023] Open
Abstract
Signal gradients encode instructive information for numerous decision-making processes during embryonic development. A striking example of precise, scalable tissue-level patterning is the segmentation of somites—the precursors of the vertebral column—during which the fibroblast growth factor (FGF), Wnt, and retinoic acid (RA) pathways establish spatial gradients. Despite decades of studies proposing roles for all three pathways, the dynamic feature of these gradients that encodes instructive information determining segment sizes remained elusive. We developed a non-elongating tail explant system, integrated quantitative measurements with computational modeling, and tested alternative models to show that positional information is encoded solely by spatial fold change (SFC) in FGF signal output. Neighboring cells measure SFC to accurately position the determination front and thus determine segment size. The SFC model successfully recapitulates results of spatiotemporal perturbation experiments on both explants and intact embryos, and it shows that Wnt signaling acts permissively upstream of FGF signaling and that RA gradient is dispensable. Simsek et al. use an elongation-arrested 3D explant system, integrated with quantitative measurements and computational modeling, to show that positional information for segmentation is encoded solely by spatial fold change (SFC) in FGF signal output. Neighboring cells measure SFC to accurately determine somite segment sizes. Wnt signaling acts permissively upstream of FGF signaling.
Collapse
Affiliation(s)
- M Fethullah Simsek
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ertuğrul M Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
29
|
Mangione F, Martín-Blanco E. The Dachsous/Fat/Four-Jointed Pathway Directs the Uniform Axial Orientation of Epithelial Cells in the Drosophila Abdomen. Cell Rep 2019; 25:2836-2850.e4. [PMID: 30517870 DOI: 10.1016/j.celrep.2018.11.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/05/2018] [Accepted: 11/06/2018] [Indexed: 12/19/2022] Open
Abstract
The achievement of the final form of an individual requires not only the control of cell size and differentiation but also integrative directional cues to instruct cell movements, positions, and orientations. In Drosophila, the adult epidermis of the abdomen is created de novo by histoblasts. As these expand and fuse, they uniformly orient along the anteroposterior axis. We found that the Dachsous/Fat/Four-jointed (Ds/Ft/Fj) pathway is key for their alignment. The refinement of the tissue-wide expression of the atypical cadherins Ds and Ft result in their polarization and directional adhesiveness. Mechanistically, the axially oriented changes in histoblasts respond to the redesign of the epithelial field. We suggest that the role of Ds/Ft/Fj in long-range oriented cell alignment is a general function and that the regulation of the expression of its components will be crucial in other morphogenetic models or during tissue repair.
Collapse
Affiliation(s)
- Federica Mangione
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Científic de Barcelona, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Enrique Martín-Blanco
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Científic de Barcelona, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| |
Collapse
|
30
|
McKenna KZ, Tao D, Nijhout HF. Exploring the Role of Insulin Signaling in Relative Growth: A Case Study on Wing-Body Scaling in Lepidoptera. Integr Comp Biol 2019; 59:1324-1337. [DOI: 10.1093/icb/icz080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abstract
Adult forms emerge from the relative growth of the body and its parts. Each appendage and organ has a unique pattern of growth that influences the size and shape it attains. This produces adult size relationships referred to as static allometries, which have received a great amount of attention in evolutionary and developmental biology. However, many questions remain unanswered, for example: What sorts of developmental processes coordinate growth? And how do these processes change given variation in body size? It has become increasingly clear that nutrition is one of the strongest influences on size relationships. In insects, nutrition acts via insulin/TOR signaling to facilitate inter- and intra-specific variation in body size and appendage size. Yet, the mechanism by which insulin signaling influences the scaling of growth remains unclear. Here we will discuss the potential roles of insulin signaling in wing-body scaling in Lepidoptera. We analyzed the growth of wings in animals reared on different diet qualities that induce a range of body sizes not normally present in our laboratory populations. By growing wings in tissue culture, we survey how perturbation and stimulation of insulin/TOR signaling influences wing growth. To conclude, we will discuss the implications of our findings for the development and evolution of organismal form.
Collapse
Affiliation(s)
| | - Della Tao
- Department of Biology, Duke University, Durham, NC 27708, USA
| | | |
Collapse
|
31
|
Mirth CK, Shingleton AW. Coordinating Development: How Do Animals Integrate Plastic and Robust Developmental Processes? Front Cell Dev Biol 2019; 7:8. [PMID: 30788342 PMCID: PMC6372504 DOI: 10.3389/fcell.2019.00008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/16/2019] [Indexed: 02/02/2023] Open
Abstract
Our developmental environment significantly affects myriad aspects of our biology, including key life history traits, morphology, physiology, and our susceptibility to disease. This environmentally-induced variation in phenotype is known as plasticity. In many cases, plasticity results from alterations in the rate of synthesis of important developmental hormones. However, while developmental processes like organ growth are sensitive to environmental conditions, others like patterning - the process that generates distinct cell identities - remain robust to perturbation. This is particularly surprising given that the same hormones that regulate organ growth also regulate organ patterning. In this review, we revisit the current approaches that address how organs coordinate their growth and pattern, and outline our hypotheses for understanding how organs achieve correct pattern across a range of sizes.
Collapse
Affiliation(s)
- Christen K Mirth
- School of Biological Sciences, Monash University, Clayton, VIC, Australia
| | - Alexander W Shingleton
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
32
|
Early girl is a novel component of the Fat signaling pathway. PLoS Genet 2019; 15:e1007955. [PMID: 30699121 PMCID: PMC6370246 DOI: 10.1371/journal.pgen.1007955] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2019] [Accepted: 01/11/2019] [Indexed: 01/05/2023] Open
Abstract
The Drosophila protocadherins Dachsous and Fat regulate growth and tissue polarity by modulating the levels, membrane localization and polarity of the atypical myosin Dachs. Localization to the apical junctional membrane is critical for Dachs function, and the adapter protein Vamana/Dlish and palmitoyl transferase Approximated are required for Dachs membrane localization. However, how Dachs levels are regulated is poorly understood. Here we identify the early girl gene as playing an essential role in Fat signaling by limiting the levels of Dachs protein. early girl mutants display overgrowth of the wings and reduced cross vein spacing, hallmark features of mutations affecting Fat signaling. Genetic experiments reveal that it functions in parallel with Fat to regulate Dachs. early girl encodes an E3 ubiquitin ligase, physically interacts with Dachs, and regulates its protein stability. Concomitant loss of early girl and approximated results in accumulation of Dachs and Vamana in cytoplasmic punctae, suggesting that it also regulates their trafficking to the apical membrane. Our findings establish a crucial role for early girl in Fat signaling, involving regulation of Dachs and Vamana, two key downstream effectors of this pathway. During development, organs grow to achieve a consistent final size. The evolutionarily conserved Hippo signaling network plays a central role in organ size control, and when dysregulated can be associated with cancer and other diseases. Fat signaling is one of several upstream pathways that impinge on Hippo signaling to regulate organ growth. We describe here identification of the Drosophila early girl gene as a new component of the Fat signaling pathway. We show that Early girl controls Fat signaling by regulating the levels of the Dachs protein. However Early girl differs from other Fat signaling regulators in that it doesn’t influence planar cell polarity or control the polarity of Dachs localization. early girl encodes a conserved protein that is predicted to influence protein stability, and it can physically associate with Dachs. We also discovered that Early girl acts together with another protein, called Approximated, to regulate the sub-cellular localization of Dachs and a Dachs-interacting protein called Vamana. Altogether, our observations establish Early girl as an essential component of Fat signaling that acts to regulate the levels and localization of Dachs and Vamana.
Collapse
|
33
|
Fat-regulated adaptor protein Dlish binds the growth suppressor Expanded and controls its stability and ubiquitination. Proc Natl Acad Sci U S A 2019; 116:1319-1324. [PMID: 30606799 PMCID: PMC6347691 DOI: 10.1073/pnas.1811891116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To regulate the growth and size of organs, cells can use information from their neighbors to modify intracellular mediators of cell proliferation. The intracellular Hippo pathway is a widely utilized nexus for growth control in animals, but its regulation by extracellular signals is not fully understood. We here identify a pathway that regulates organ size in Drosophila, triggered by the transmembrane receptor, the giant protocadherin Fat. We show that the Fat-regulated SH3 domain adaptor protein Dlish binds to and reduces the stability of the growth suppressor Expanded, a known regulator of the Hippo pathway. The destabilization of Expanded by Dlish works in parallel to a previously established pathway in which Dlish increases levels of the growth-stimulating protein Dachs. The Drosophila protocadherin Fat controls organ size through the Hippo pathway, but the biochemical links to the Hippo pathway components are still poorly defined. We previously identified Dlish, an SH3 domain protein that physically interacts with Fat and the type XX myosin Dachs, and showed that Fat’s regulation of Dlish levels and activity helps limit Dachs-mediated inhibition of Hippo pathway activity. We here characterize a parallel growth control pathway downstream of Fat and Dlish. Using immunoprecipitation and mass spectrometry to search for Dlish partners, we find that Dlish binds the FERM domain growth repressor Expanded (Ex); Dlish SH3 domains directly bind sites in the Ex C terminus. We further show that, in vivo, Dlish reduces the subapical accumulation of Ex, and that loss of Dlish blocks the destabilization of Ex caused by loss of Fat. Moreover, Dlish can bind the F-box E3 ubiquitin ligase Slimb and promote Slimb-mediated ubiquitination of Expanded in vitro. Both the in vitro and in vivo effects of Dlish on Ex require Slimb, strongly suggesting that Dlish destabilizes Ex by helping recruit Slimb-containing E3 ubiquitin ligase complexes to Ex.
Collapse
|
34
|
Gou J, Lin L, Othmer HG. A Model for the Hippo Pathway in the Drosophila Wing Disc. Biophys J 2018; 115:737-747. [PMID: 30041810 PMCID: PMC6103738 DOI: 10.1016/j.bpj.2018.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/21/2018] [Accepted: 07/02/2018] [Indexed: 01/18/2023] Open
Abstract
Although significant progress has been made toward understanding morphogen-mediated patterning in development, control of the size and shape of tissues via local and global signaling is poorly understood. In particular, little is known about how cell-cell interactions are involved in the control of tissue size. The Hippo pathway in the Drosophila wing disc involves cell-cell interactions via cadherins, which lead to modulation of Yorkie, a cotranscriptional factor that affects control of the cell cycle and growth, and studies involving over- and underexpression of components of this pathway reveal conditions that lead to tissue over- or undergrowth. Here, we develop a mathematical model of the Hippo pathway that can qualitatively explain these observations, made in both whole-disc mutants and mutant-clone experiments. We find that a number of nonintuitive experimental results can be explained by subtle changes in the balances between inputs to the Hippo pathway and suggest some predictions that can be tested experimentally. We also show that certain components of the pathway are polarized at the single-cell level, which replicates observations of planar cell polarity. Because the signal transduction and growth control pathways are highly conserved between Drosophila and mammalian systems, the model we formulate can be used as a framework to guide future experimental work on the Hippo pathway in both Drosophila and mammalian systems.
Collapse
Affiliation(s)
- Jia Gou
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Lin Lin
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
35
|
Abstract
The Hippo signal transduction pathway is an important regulator of organ growth and cell differentiation, and its deregulation contributes to the development of cancer. The activity of the Hippo pathway is strongly dependent on cell junctions, cellular architecture, and the mechanical properties of the microenvironment. In this review, we discuss recent advances in our understanding of how cell junctions transduce signals from the microenvironment and control the activity of the Hippo pathway. We also discuss how these mechanisms may control organ growth during development and regeneration, and how defects in them deregulate Hippo signaling in cancer cells.
Collapse
Affiliation(s)
- Ruchan Karaman
- VIB Center for Cancer Biology, University of Leuven, 3000 Leuven, Belgium.,Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| | - Georg Halder
- VIB Center for Cancer Biology, University of Leuven, 3000 Leuven, Belgium.,Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| |
Collapse
|
36
|
Hust J, Lavine MD, Worthington AM, Zinna R, Gotoh H, Niimi T, Lavine L. The Fat-Dachsous signaling pathway regulates growth of horns in Trypoxylus dichotomus, but does not affect horn allometry. JOURNAL OF INSECT PHYSIOLOGY 2018; 105:85-94. [PMID: 29366850 DOI: 10.1016/j.jinsphys.2018.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 06/07/2023]
Abstract
Males of the Asian rhinoceros beetle, Trypoxylus dichotomus, possess exaggerated head and thoracic horns that scale dramatically out of proportion to body size. While studies of insulin signaling suggest that this pathway regulates nutrition-dependent growth including exaggerated horns, what regulates disproportionate growth has yet to be identified. The Fat signaling pathway is a potential candidate for regulating disproportionate growth of sexually-selected traits, a hypothesis we advanced in a previous paper (Gotoh et al., 2015). To investigate the role of Fat signaling in the growth and scaling of the sexually dimorphic, condition-dependent traits of the in the Asian rhinoceros beetle T. dichotomus, we used RNA interference to knock down expression of fat and its co-receptor dachsous. Knockdown of fat, and to a lesser degree dachsous, caused shortening and widening of appendages, including the head and thoracic horns. However, scaling of horns to body size was not affected. Our results show that Fat signaling regulates horn growth in T. dichotomus as it does in appendage growth in other insects. However, we provide evidence that Fat signaling does not mediate the disproportionate, positive allometric growth of horns in T. dichotomus.
Collapse
Affiliation(s)
- James Hust
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Mark D Lavine
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Amy M Worthington
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Robert Zinna
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Hiroki Gotoh
- Department of Entomology, Washington State University, Pullman, WA 99164, United States; Lab of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - T Niimi
- Lab of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya 464-8601, Japan; Division of Evolutionary Developmental Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
| | - Laura Lavine
- Department of Entomology, Washington State University, Pullman, WA 99164, United States.
| |
Collapse
|
37
|
Abstract
In his classic book On Growth and Form, D'Arcy Thompson discussed the necessity of a physical and mathematical approach to understanding the relationship between growth and form. The past century has seen extraordinary advances in our understanding of biological components and processes contributing to organismal morphogenesis, but the mathematical and physical principles involved have not received comparable attention. The most obvious entry of physics into morphogenesis is via tissue mechanics. In this Review, we discuss the fundamental role of mechanical interactions between cells induced by growth in shaping a tissue. Non-uniform growth can lead to accumulation of mechanical stress, which in the context of two-dimensional sheets of tissue can specify the shape it assumes in three dimensions. A special class of growth patterns - conformal growth - does not lead to the accumulation of stress and can generate a rich variety of planar tissue shapes. Conversely, mechanical stress can provide a regulatory feedback signal into the growth control circuit. Both theory and experiment support a key role for mechanical interactions in shaping tissues and, via mechanical feedback, controlling epithelial growth.
Collapse
Affiliation(s)
- Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway NJ 08854, USA
| | - Boris I Shraiman
- Department of Physics, Kavli Institute for Theoretical Physics, University of California, Santa Barbara, CA 93101, USA
| |
Collapse
|
38
|
Dye NA, Popović M, Spannl S, Etournay R, Kainmüller D, Ghosh S, Myers EW, Jülicher F, Eaton S. Cell dynamics underlying oriented growth of the Drosophila wing imaginal disc. Development 2017; 144:4406-4421. [PMID: 29038308 DOI: 10.1242/dev.155069] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022]
Abstract
Quantitative analysis of the dynamic cellular mechanisms shaping the Drosophila wing during its larval growth phase has been limited, impeding our ability to understand how morphogen patterns regulate tissue shape. Such analysis requires explants to be imaged under conditions that maintain both growth and patterning, as well as methods to quantify how much cellular behaviors change tissue shape. Here, we demonstrate a key requirement for the steroid hormone 20-hydroxyecdysone (20E) in the maintenance of numerous patterning systems in vivo and in explant culture. We find that low concentrations of 20E support prolonged proliferation in explanted wing discs in the absence of insulin, incidentally providing novel insight into the hormonal regulation of imaginal growth. We use 20E-containing media to observe growth directly and to apply recently developed methods for quantitatively decomposing tissue shape changes into cellular contributions. We discover that whereas cell divisions drive tissue expansion along one axis, their contribution to expansion along the orthogonal axis is cancelled by cell rearrangements and cell shape changes. This finding raises the possibility that anisotropic mechanical constraints contribute to growth orientation in the wing disc.
Collapse
Affiliation(s)
- Natalie A Dye
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Marko Popović
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187 Dresden, Germany
| | - Stephanie Spannl
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Raphaël Etournay
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany.,Unité de Génétique et Physiologie de l'Audition UMRS 1120, Département de Neurosciences, Institut Pasteur, 75015 Paris, France
| | - Dagmar Kainmüller
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany.,Janelia Farm Research Campus, 19700 Helix Dr, Ashburn, VA 20147, USA
| | - Suhrid Ghosh
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Eugene W Myers
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany.,Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187 Dresden, Germany .,Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Suzanne Eaton
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany .,Biotechnologisches Zentrum, Technische Universität Dresden, Tatzberg 47/49, 01309 Dresden, Germany
| |
Collapse
|
39
|
Difference in Dachsous Levels between Migrating Cells Coordinates the Direction of Collective Cell Migration. Dev Cell 2017; 42:479-497.e10. [DOI: 10.1016/j.devcel.2017.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/19/2017] [Accepted: 07/31/2017] [Indexed: 12/21/2022]
|
40
|
Loza O, Heemskerk I, Gordon-Bar N, Amir-Zilberstein L, Jung Y, Sprinzak D. A synthetic planar cell polarity system reveals localized feedback on Fat4-Ds1 complexes. eLife 2017; 6:e24820. [PMID: 28826487 PMCID: PMC5576920 DOI: 10.7554/elife.24820] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 08/09/2017] [Indexed: 12/21/2022] Open
Abstract
The atypical cadherins Fat and Dachsous (Ds) have been found to underlie planar cell polarity (PCP) in many tissues. Theoretical models suggest that polarity can arise from localized feedbacks on Fat-Ds complexes at the cell boundary. However, there is currently no direct evidence for the existence or mechanism of such feedbacks. To directly test the localized feedback model, we developed a synthetic biology platform based on mammalian cells expressing the human Fat4 and Ds1. We show that Fat4-Ds1 complexes accumulate on cell boundaries in a threshold-like manner and exhibit dramatically slower dynamics than unbound Fat4 and Ds1. This suggests a localized feedback mechanism based on enhanced stability of Fat4-Ds1 complexes. We also show that co-expression of Fat4 and Ds1 in the same cells is sufficient to induce polarization of Fat4-Ds1 complexes. Together, these results provide direct evidence that localized feedbacks on Fat4-Ds1 complexes can give rise to PCP.
Collapse
Affiliation(s)
- Olga Loza
- Department of Biochemistry and Molecular Biology, Wise Faculty of Life ScienceTel Aviv UniversityTel AvivIsrael
| | - Idse Heemskerk
- Department of BiosciencesRice UniversityHoustonUnited States
| | - Nadav Gordon-Bar
- Department of Biochemistry and Molecular Biology, Wise Faculty of Life ScienceTel Aviv UniversityTel AvivIsrael
| | - Liat Amir-Zilberstein
- Department of Biochemistry and Molecular Biology, Wise Faculty of Life ScienceTel Aviv UniversityTel AvivIsrael
| | - Yunmin Jung
- Department of Biochemistry and Molecular Biology, Wise Faculty of Life ScienceTel Aviv UniversityTel AvivIsrael
| | - David Sprinzak
- Department of Biochemistry and Molecular Biology, Wise Faculty of Life ScienceTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
41
|
A Kinome RNAi Screen in Drosophila Identifies Novel Genes Interacting with Lgl, aPKC, and Crb Cell Polarity Genes in Epithelial Tissues. G3-GENES GENOMES GENETICS 2017; 7:2497-2509. [PMID: 28611255 PMCID: PMC5555457 DOI: 10.1534/g3.117.043513] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In both Drosophila melanogaster and mammalian systems, epithelial structure and underlying cell polarity are essential for proper tissue morphogenesis and organ growth. Cell polarity interfaces with multiple cellular processes that are regulated by the phosphorylation status of large protein networks. To gain insight into the molecular mechanisms that coordinate cell polarity with tissue growth, we screened a boutique collection of RNAi stocks targeting the kinome for their capacity to modify Drosophila “cell polarity” eye and wing phenotypes. Initially, we identified kinase or phosphatase genes whose depletion modified adult eye phenotypes associated with the manipulation of cell polarity complexes (via overexpression of Crb or aPKC). We next conducted a secondary screen to test whether these cell polarity modifiers altered tissue overgrowth associated with depletion of Lgl in the wing. These screens identified Hippo, Jun kinase (JNK), and Notch signaling pathways, previously linked to cell polarity regulation of tissue growth. Furthermore, novel pathways not previously connected to cell polarity regulation of tissue growth were identified, including Wingless (Wg/Wnt), Ras, and lipid/Phospho-inositol-3-kinase (PI3K) signaling pathways. Additionally, we demonstrated that the “nutrient sensing” kinases Salt Inducible Kinase 2 and 3 (SIK2 and 3) are potent modifiers of cell polarity phenotypes and regulators of tissue growth. Overall, our screen has revealed novel cell polarity-interacting kinases and phosphatases that affect tissue growth, providing a platform for investigating molecular mechanisms coordinating cell polarity and tissue growth during development.
Collapse
|
42
|
Wortman JC, Nahmad M, Zhang PC, Lander AD, Yu CC. Expanding signaling-molecule wavefront model of cell polarization in the Drosophila wing primordium. PLoS Comput Biol 2017; 13:e1005610. [PMID: 28671940 PMCID: PMC5515495 DOI: 10.1371/journal.pcbi.1005610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 07/18/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
In developing tissues, cell polarization and proliferation are regulated by morphogens and signaling pathways. Cells throughout the Drosophila wing primordium typically show subcellular localization of the unconventional myosin Dachs on the distal side of cells (nearest the center of the disc). Dachs localization depends on the spatial distribution of bonds between the protocadherins Fat (Ft) and Dachsous (Ds), which form heterodimers between adjacent cells; and the Golgi kinase Four-jointed (Fj), which affects the binding affinities of Ft and Ds. The Fj concentration forms a linear gradient while the Ds concentration is roughly uniform throughout most of the wing pouch with a steep transition region that propagates from the center to the edge of the pouch during the third larval instar. Although the Fj gradient is an important cue for polarization, it is unclear how the polarization is affected by cell division and the expanding Ds transition region, both of which can alter the distribution of Ft-Ds heterodimers around the cell periphery. We have developed a computational model to address these questions. In our model, the binding affinity of Ft and Ds depends on phosphorylation by Fj. We assume that the asymmetry of the Ft-Ds bond distribution around the cell periphery defines the polarization, with greater asymmetry promoting cell proliferation. Our model predicts that this asymmetry is greatest in the radially-expanding transition region that leaves polarized cells in its wake. These cells naturally retain their bond distribution asymmetry after division by rapidly replenishing Ft-Ds bonds at new cell-cell interfaces. Thus we predict that the distal localization of Dachs in cells throughout the pouch requires the movement of the Ds transition region and the simple presence, rather than any specific spatial pattern, of Fj. In the tissues of a developing organism, specialized proteins can control cell growth and give cells a sense of direction, e.g., which way is the head or the tail, by having their concentration vary throughout the tissue. In cells of the developing fruit fly wing, a protein called Dachs localizes on the side of the cell closest to the center of the tissue, indicating a directionality. The localization of Dachs is determined by the spatial distribution, around the periphery of a cell, of intercellular bonds of the proteins Fat and Dachsous between adjacent cells. Here we asked how this cell directionality is affected when cells divide and when the concentration of Dachsous changes over time. We use a computational model to show that as the circular step-up region of the Dachsous concentration profile sweeps radially outward, like rings radiating outward from where a pebble was dropped in a pond, it leaves polarized cells in its wake. Our model also shows how cells can naturally recover their directionality after cell division.
Collapse
Affiliation(s)
- Juliana C. Wortman
- Department of Physics and Astronomy, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
| | - Marcos Nahmad
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
| | - Peng Cheng Zhang
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
| | - Arthur D. Lander
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
| | - Clare C. Yu
- Department of Physics and Astronomy, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
43
|
Configuring a robust nervous system with Fat cadherins. Semin Cell Dev Biol 2017; 69:91-101. [PMID: 28603077 DOI: 10.1016/j.semcdb.2017.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/26/2017] [Accepted: 06/07/2017] [Indexed: 01/14/2023]
Abstract
Atypical Fat cadherins represent a small but versatile group of signaling molecules that influence proliferation and tissue polarity. With huge extracellular domains and intracellular domains harboring many independent protein interaction sites, Fat cadherins are poised to translate local cell adhesion events into a variety of cell behaviors. The need for such global coordination is particularly prominent in the nervous system, where millions of morphologically diverse neurons are organized into functional networks. As we learn more about their biological functions and molecular properties, increasing evidence suggests that Fat cadherins mediate contact-induced changes that ultimately impose a structure to developing neuronal circuits.
Collapse
|
44
|
|
45
|
Montes AJ, Morata G. Homeostatic response to blocking cell division in Drosophila imaginal discs: Role of the Fat/Dachsous (Ft/Ds) pathway. Dev Biol 2017; 424:113-123. [PMID: 28300568 DOI: 10.1016/j.ydbio.2017.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/08/2017] [Accepted: 03/10/2017] [Indexed: 12/28/2022]
Abstract
One major problem in developmental biology is the identification of the mechanisms that control the final size of tissues and organs. We are addressing this issue in the imaginal discs of Drosophila by analysing the response to blocking cell division in large domains in the wing and leg discs. The affected domains may be zones of restricted lineage like compartments, or zones of open lineage that may integrate cells from the surrounding territory. Our results reveal the existence of a powerful homeostatic mechanism that can compensate for gross differences in growth rates and builds structures of normal size. This mechanism functions at the level of whole discs, inducing additional cell proliferation to generate the cells that populate the cell division-arrested territory and generating an active recruitment process to integrate those cells. The activation of this response mechanism is mediated by alterations in the normal activity of PCP genes of the Fat/Ds system: in discs mutant for dachs, ds or four jointed the response mechanism is not activated.
Collapse
Affiliation(s)
| | - Ginés Morata
- Centro de Biología Molecular CSIC-UAM, Madrid, Spain.
| |
Collapse
|
46
|
Keira Y, Wada M, Ishikawa HO. Regulation of Drosophila Development by the Golgi Kinase Four-Jointed. Curr Top Dev Biol 2017; 123:143-179. [DOI: 10.1016/bs.ctdb.2016.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
47
|
Kale A, Rimesso G, Baker NE. Local Cell Death Changes the Orientation of Cell Division in the Developing Drosophila Wing Imaginal Disc Without Using Fat or Dachsous as Orienting Signals. PLoS One 2016; 11:e0167637. [PMID: 28030539 PMCID: PMC5193341 DOI: 10.1371/journal.pone.0167637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/17/2016] [Indexed: 12/21/2022] Open
Abstract
Drosophila imaginal disc cells exhibit preferred cell division orientations according to location within the disc. These orientations are altered if cell death occurs within the epithelium, such as is caused by cell competition or by genotypes affecting cell survival. Both normal cell division orientations, and their orientations after cell death, depend on the Fat-Dachsous pathway of planar cell polarity (PCP). The hypothesis that cell death initiates a planar polarity signal was investigated. When clones homozygous for the pineapple eye (pie) mutation were made to initiate cell death, neither Dachsous nor Fat was required in pie cells for the re-orientation of nearby cells, indicating a distinct signal for this PCP pathway. Dpp and Wg were also not needed for pie clones to re-orient cell division. Cell shapes were evaluated in wild type and mosaic wing discs to assess mechanical consequences of cell loss. Although proximal wing disc cells and cells close to the dorso-ventral boundary were elongated in their preferred cell division axes in wild type discs, cell shapes in much of the wing pouch were symmetrical on average and did not predict their preferred division axis. Cells in pie mutant clones were slightly larger than their normal counterparts, consistent with mechanical stretching following cell loss, but no bias in cell shape was detected in the surrounding cells. These findings indicate that an unidentified signal influences PCP-dependent cell division orientation in imaginal discs.
Collapse
Affiliation(s)
- Abhijit Kale
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NYC, NY, United States of America
| | - Gerard Rimesso
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NYC, NY, United States of America
| | - Nicholas E. Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NYC, NY, United States of America
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NYC, NY, United States of America
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NYC, NY, United States of America
- * E-mail:
| |
Collapse
|
48
|
Matakatsu H, Blair SS, Fehon RG. The palmitoyltransferase Approximated promotes growth via the Hippo pathway by palmitoylation of Fat. J Cell Biol 2016; 216:265-277. [PMID: 28031421 PMCID: PMC5223609 DOI: 10.1083/jcb.201609094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/19/2016] [Accepted: 12/09/2016] [Indexed: 01/07/2023] Open
Abstract
The palmitoyl transferase Approximated regulates signaling by the protocadherin Fat to control tissue growth upstream of the Hippo pathway in Drosophila. Matakatsu et al. show that palmitoylation of the intracellular domain of Fat by Approximated negatively regulates Fat and its ability to restrict growth. The large protocadherin Fat functions to promote Hippo pathway activity in restricting tissue growth. Loss of Fat leads to accumulation of the atypical myosin Dachs at the apical junctional region, which in turn promotes growth by inhibiting Warts. We previously identified Approximated (App), a DHHC domain palmitoyltransferase, as a negative regulator of Fat signaling in growth control. We show here that App promotes growth by palmitoylating the intracellular domain of Fat, and that palmitoylation negatively regulates Fat function. Independently, App also recruits Dachs to the apical junctional region through protein–protein association, thereby stimulating Dachs’s activity in promoting growth. Further, we show that palmitoylation by App functions antagonistically to phosphorylation by Discs-overgrown, which activates Fat. Together, these findings suggest a model in which App promotes Dachs activity by simultaneously repressing Fat via posttranslational modification and recruiting Dachs to the apical junctional region, thereby promoting tissue growth.
Collapse
Affiliation(s)
- Hitoshi Matakatsu
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637.,Department of Zoology, University of Wisconsin, Madison, WI 53706
| | - Seth S Blair
- Department of Zoology, University of Wisconsin, Madison, WI 53706
| | - Richard G Fehon
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
49
|
Vollmer J, Iber D. An Unbiased Analysis of Candidate Mechanisms for the Regulation of Drosophila Wing Disc Growth. Sci Rep 2016; 6:39228. [PMID: 27995964 PMCID: PMC5172366 DOI: 10.1038/srep39228] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/16/2016] [Indexed: 11/25/2022] Open
Abstract
The control of organ size presents a fundamental open problem in biology. A declining growth rate is observed in all studied higher animals, and the growth limiting mechanism may therefore be evolutionary conserved. Most studies of organ growth control have been carried out in Drosophila imaginal discs. We have previously shown that the area growth rate in the Drosophila eye primordium declines inversely proportional to the increase in its area, which is consistent with a dilution mechanism for growth control. Here, we show that a dilution mechanism cannot explain growth control in the Drosophila wing disc. We computationally evaluate a range of alternative candidate mechanisms and show that the experimental data can be best explained by a biphasic growth law. However, also logistic growth and an exponentially declining growth rate fit the data very well. The three growth laws correspond to fundamentally different growth mechanisms that we discuss. Since, as we show, a fit to the available experimental growth kinetics is insufficient to define the underlying mechanism of growth control, future experimental studies must focus on the molecular mechanisms to define the mechanism of growth control.
Collapse
Affiliation(s)
- Jannik Vollmer
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058, Basel, Switzerland
| | - Dagmar Iber
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058, Basel, Switzerland
| |
Collapse
|
50
|
Brun-Usan M, Marín-Riera M, Grande C, Truchado-Garcia M, Salazar-Ciudad I. A set of simple cell processes is sufficient to model spiral cleavage. Development 2016; 144:54-62. [PMID: 27888194 DOI: 10.1242/dev.140285] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/14/2016] [Indexed: 11/20/2022]
Abstract
During cleavage, different cellular processes cause the zygote to become partitioned into a set of cells with a specific spatial arrangement. These processes include the orientation of cell division according to: an animal-vegetal gradient; the main axis (Hertwig's rule) of the cell; and the contact areas between cells or the perpendicularity between consecutive cell divisions (Sachs' rule). Cell adhesion and cortical rotation have also been proposed to be involved in spiral cleavage. We use a computational model of cell and tissue biomechanics to account for the different existing hypotheses about how the specific spatial arrangement of cells in spiral cleavage arises during development. Cell polarization by an animal-vegetal gradient, a bias to perpendicularity between consecutive cell divisions (Sachs' rule), cortical rotation and cell adhesion, when combined, reproduce the spiral cleavage, whereas other combinations of processes cannot. Specifically, cortical rotation is necessary at the 8-cell stage to direct all micromeres in the same direction. By varying the relative strength of these processes, we reproduce the spatial arrangement of cells in the blastulae of seven different invertebrate species.
Collapse
Affiliation(s)
- Miguel Brun-Usan
- Genomics, Bioinformatics and Evolution, Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain.,Evo-devo Helsinki community, Centre of Excellence in Computational and Experimental Developmental Biology, Institute of Biotechnology, University of Helsinki, PO Box 56, Helsinki FIN-00014, Finland
| | - Miquel Marín-Riera
- Genomics, Bioinformatics and Evolution, Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain.,Evo-devo Helsinki community, Centre of Excellence in Computational and Experimental Developmental Biology, Institute of Biotechnology, University of Helsinki, PO Box 56, Helsinki FIN-00014, Finland
| | - Cristina Grande
- Departamento de Biología Molecular and Centro de Biología Molecular, 'Severo Ochoa' (CSIC, Universidad Autónoma de Madrid), Madrid, Spain.,Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Cantoblanco, Madrid 28049, Spain
| | - Marta Truchado-Garcia
- Departamento de Biología Molecular and Centro de Biología Molecular, 'Severo Ochoa' (CSIC, Universidad Autónoma de Madrid), Madrid, Spain.,Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Cantoblanco, Madrid 28049, Spain
| | - Isaac Salazar-Ciudad
- Genomics, Bioinformatics and Evolution, Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Barcelona, Spain .,Evo-devo Helsinki community, Centre of Excellence in Computational and Experimental Developmental Biology, Institute of Biotechnology, University of Helsinki, PO Box 56, Helsinki FIN-00014, Finland
| |
Collapse
|