1
|
Lehr S, Brückner DB, Minchington TG, Greunz-Schindler M, Merrin J, Hannezo E, Kicheva A. Self-organized pattern formation in the developing mouse neural tube by a temporal relay of BMP signaling. Dev Cell 2025; 60:567-580.e14. [PMID: 39603235 DOI: 10.1016/j.devcel.2024.10.024] [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: 11/13/2023] [Revised: 05/08/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Developing tissues interpret dynamic changes in morphogen activity to generate cell type diversity. To quantitatively study bone morphogenetic protein (BMP) signaling dynamics in the mouse neural tube, we developed an embryonic stem cell differentiation system tailored for growing tissues. Differentiating cells form striking self-organized patterns of dorsal neural tube cell types driven by sequential phases of BMP signaling that are observed both in vitro and in vivo. Data-driven biophysical modeling showed that these dynamics result from coupling fast negative feedback with slow positive regulation of signaling by the specification of an endogenous BMP source. Thus, in contrast to relays that propagate morphogen signaling in space, we identify a BMP signaling relay that operates in time. This mechanism allows for a rapid initial concentration-sensitive response that is robustly terminated, thereby regulating balanced sequential cell type generation. Our study provides an experimental and theoretical framework to understand how signaling dynamics are exploited in developing tissues.
Collapse
Affiliation(s)
- Stefanie Lehr
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria
| | - David B Brückner
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria
| | | | | | - Jack Merrin
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria.
| | - Anna Kicheva
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria.
| |
Collapse
|
2
|
Sharma V, Sachan N, Sarkar B, Mutsuddi M, Mukherjee A. E3 ubiquitin ligase Deltex facilitates the expansion of Wingless gradient and antagonizes Wingless signaling through a conserved mechanism of transcriptional effector Armadillo/β-catenin degradation. eLife 2024; 12:RP88466. [PMID: 38900140 PMCID: PMC11189633 DOI: 10.7554/elife.88466] [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: 06/21/2024] Open
Abstract
The Wnt/Wg pathway controls myriads of biological phenomena throughout the development and adult life of all organisms across the phyla. Thus, an aberrant Wnt signaling is associated with a wide range of pathologies in humans. Tight regulation of Wnt/Wg signaling is required to maintain proper cellular homeostasis. Here, we report a novel role of E3 ubiquitin ligase Deltex in Wg signaling regulation. Drosophila dx genetically interacts with wg and its pathway components. Furthermore, Dx LOF results in a reduced spreading of Wg while its over-expression expands the diffusion gradient of the morphogen. We attribute this change in Wg gradient to the endocytosis of Wg through Dx which directly affects the short- and long-range Wg targets. We also demonstrate the role of Dx in regulating Wg effector Armadillo where Dx down-regulates Arm through proteasomal degradation. We also showed the conservation of Dx function in the mammalian system where DTX1 is shown to bind with β-catenin and facilitates its proteolytic degradation, spotlighting a novel step that potentially modulates Wnt/Wg signaling cascade.
Collapse
Affiliation(s)
- Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu UniversityVaranasiIndia
- Department of Integrative Biology and Physiology, University of California Los AngelesLos AngelesUnited States
| | - Nalani Sachan
- Department of Cell Biology, NYU Langone Medical CenterNew YorkUnited States
| | - Bappi Sarkar
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu UniversityVaranasiIndia
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu UniversityVaranasiIndia
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu UniversityVaranasiIndia
| |
Collapse
|
3
|
Harish RK, Gupta M, Zöller D, Hartmann H, Gheisari A, Machate A, Hans S, Brand M. Real-time monitoring of an endogenous Fgf8a gradient attests to its role as a morphogen during zebrafish gastrulation. Development 2023; 150:dev201559. [PMID: 37665167 PMCID: PMC10565248 DOI: 10.1242/dev.201559] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Morphogen gradients impart positional information to cells in a homogenous tissue field. Fgf8a, a highly conserved growth factor, has been proposed to act as a morphogen during zebrafish gastrulation. However, technical limitations have so far prevented direct visualization of the endogenous Fgf8a gradient and confirmation of its morphogenic activity. Here, we monitor Fgf8a propagation in the developing neural plate using a CRISPR/Cas9-mediated EGFP knock-in at the endogenous fgf8a locus. By combining sensitive imaging with single-molecule fluorescence correlation spectroscopy, we demonstrate that Fgf8a, which is produced at the embryonic margin, propagates by diffusion through the extracellular space and forms a graded distribution towards the animal pole. Overlaying the Fgf8a gradient curve with expression profiles of its downstream targets determines the precise input-output relationship of Fgf8a-mediated patterning. Manipulation of the extracellular Fgf8a levels alters the signaling outcome, thus establishing Fgf8a as a bona fide morphogen during zebrafish gastrulation. Furthermore, by hindering Fgf8a diffusion, we demonstrate that extracellular diffusion of the protein from the source is crucial for it to achieve its morphogenic potential.
Collapse
Affiliation(s)
- Rohit Krishnan Harish
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Mansi Gupta
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Daniela Zöller
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Hella Hartmann
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- CMCB Technology Platform, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Ali Gheisari
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- CMCB Technology Platform, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Anja Machate
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Stefan Hans
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Michael Brand
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| |
Collapse
|
4
|
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
|
5
|
Fixsen BR, Han CZ, Zhou Y, Spann NJ, Saisan P, Shen Z, Balak C, Sakai M, Cobo I, Holtman IR, Warden AS, Ramirez G, Collier JG, Pasillas MP, Yu M, Hu R, Li B, Belhocine S, Gosselin D, Coufal NG, Ren B, Glass CK. SALL1 enforces microglia-specific DNA binding and function of SMADs to establish microglia identity. Nat Immunol 2023; 24:1188-1199. [PMID: 37322178 PMCID: PMC10307637 DOI: 10.1038/s41590-023-01528-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 05/04/2023] [Indexed: 06/17/2023]
Abstract
Spalt-like transcription factor 1 (SALL1) is a critical regulator of organogenesis and microglia identity. Here we demonstrate that disruption of a conserved microglia-specific super-enhancer interacting with the Sall1 promoter results in complete and specific loss of Sall1 expression in microglia. By determining the genomic binding sites of SALL1 and leveraging Sall1 enhancer knockout mice, we provide evidence for functional interactions between SALL1 and SMAD4 required for microglia-specific gene expression. SMAD4 binds directly to the Sall1 super-enhancer and is required for Sall1 expression, consistent with an evolutionarily conserved requirement of the TGFβ and SMAD homologs Dpp and Mad for cell-specific expression of Spalt in the Drosophila wing. Unexpectedly, SALL1 in turn promotes binding and function of SMAD4 at microglia-specific enhancers while simultaneously suppressing binding of SMAD4 to enhancers of genes that become inappropriately activated in enhancer knockout microglia, thereby enforcing microglia-specific functions of the TGFβ-SMAD signaling axis.
Collapse
Affiliation(s)
- Bethany R Fixsen
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Claudia Z Han
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Yi Zhou
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Nathanael J Spann
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Payam Saisan
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Zeyang Shen
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Christopher Balak
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Mashito Sakai
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Isidoro Cobo
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Inge R Holtman
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Anna S Warden
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | | | - Jana G Collier
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Martina P Pasillas
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Miao Yu
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Rong Hu
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Bin Li
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Sarah Belhocine
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Quebec, Quebec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Quebec, Quebec, Canada
| | - David Gosselin
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Quebec, Quebec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Quebec, Quebec, Canada
| | - Nicole G Coufal
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
- Department of Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Pierini G, Dahmann C. Hedgehog morphogen gradient is robust towards variations in tissue morphology in Drosophila. Sci Rep 2023; 13:8454. [PMID: 37231029 DOI: 10.1038/s41598-023-34632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
During tissue development, gradients of secreted signaling molecules known as morphogens provide cells with positional information. The mechanisms underlying morphogen spreading have been widely studied, however, it remains largely unexplored whether the shape of morphogen gradients is influenced by tissue morphology. Here, we developed an analysis pipeline to quantify the distribution of proteins within a curved tissue. We applied it to the Hedgehog morphogen gradient in the Drosophila wing and eye-antennal imaginal discs, which are flat and curved tissues, respectively. Despite a different expression profile, the slope of the Hedgehog gradient was comparable between the two tissues. Moreover, inducing ectopic folds in wing imaginal discs did not affect the slope of the Hedgehog gradient. Suppressing curvature in the eye-antennal imaginal disc also did not alter the Hedgehog gradient slope but led to ectopic Hedgehog expression. In conclusion, through the development of an analysis pipeline that allows quantifying protein distribution in curved tissues, we show that the Hedgehog gradient is robust towards variations in tissue morphology.
Collapse
Affiliation(s)
- Giulia Pierini
- School of Science, Technische Universität Dresden, 01062, Dresden, Germany
| | - Christian Dahmann
- School of Science, Technische Universität Dresden, 01062, Dresden, Germany.
- Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062, Dresden, Germany.
| |
Collapse
|
7
|
Bauer M, Aguilar G, Wharton KA, Matsuda S, Affolter M. Heterodimerization-dependent secretion of bone morphogenetic proteins in Drosophila. Dev Cell 2023; 58:645-659.e4. [PMID: 37054707 PMCID: PMC10303954 DOI: 10.1016/j.devcel.2023.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/13/2023] [Accepted: 03/09/2023] [Indexed: 04/15/2023]
Abstract
Combinatorial signaling is key to instruct context-dependent cell behaviors. During embryonic development, adult homeostasis, and disease, bone morphogenetic proteins (BMPs) act as dimers to instruct specific cellular responses. BMP ligands can form both homodimers or heterodimers; however, obtaining direct evidence of the endogenous localization and function of each form has proven challenging. Here, we make use of precise genome editing and direct protein manipulation via protein binders to dissect the existence and functional relevance of BMP homodimers and heterodimers in the Drosophila wing imaginal disc. This approach identified in situ the existence of Dpp (BMP2/4)/Gbb (BMP5/6/7/8) heterodimers. We found that Gbb is secreted in a Dpp-dependent manner in the wing imaginal disc. Dpp and Gbb form a gradient of heterodimers, whereas neither Dpp nor Gbb homodimers are evident under endogenous physiological conditions. We find that the formation of heterodimers is critical for obtaining optimal signaling and long-range BMP distribution.
Collapse
Affiliation(s)
- Milena Bauer
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Gustavo Aguilar
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | | | - Shinya Matsuda
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| | - Markus Affolter
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| |
Collapse
|
8
|
Friesen S, Hariharan IK. Coordinated growth of linked epithelia is mediated by the Hippo pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.26.530099. [PMID: 36993542 PMCID: PMC10054945 DOI: 10.1101/2023.02.26.530099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
An epithelium in a living organism seldom develops in isolation. Rather, most epithelia are tethered to other epithelial or non-epithelial tissues, necessitating growth coordination between layers. We investigated how two tethered epithelial layers of the Drosophila larval wing imaginal disc, the disc proper (DP) and the peripodial epithelium (PE), coordinate their growth. DP growth is driven by the morphogens Hedgehog (Hh) and Dpp, but regulation of PE growth is poorly understood. We find that the PE adapts to changes in growth rates of the DP, but not vice versa, suggesting a "leader and follower" mechanism. Moreover, PE growth can occur by cell shape changes, even when proliferation is inhibited. While Hh and Dpp pattern gene expression in both layers, growth of the DP is exquisitely sensitive to Dpp levels, while growth of the PE is not; the PE can achieve an appropriate size even when Dpp signaling is inhibited. Instead, both the growth of the PE and its accompanying cell shape changes require the activity of two components of the mechanosensitive Hippo pathway, the DNA-binding protein Scalloped (Sd) and its co-activator (Yki), which could allow the PE to sense and respond to forces generated by DP growth. Thus, an increased reliance on mechanically-dependent growth mediated by the Hippo pathway, at the expense of morphogen-dependent growth, enables the PE to evade layer-intrinsic growth control mechanisms and coordinate its growth with the DP. This provides a potential paradigm for growth coordination between different components of a developing organ.
Collapse
Affiliation(s)
- Sophia Friesen
- Department of Molecular and Cell Biology, University of California, Berkeley
| | - Iswar K. Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley
| |
Collapse
|
9
|
Diaz-Torres E, Muñoz-Nava LM, Nahmad M. Coupling cell proliferation rates to the duration of recruitment controls final size of the Drosophila wing. Proc Biol Sci 2022; 289:20221167. [PMID: 36476003 PMCID: PMC9554725 DOI: 10.1098/rspb.2022.1167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022] Open
Abstract
Organ growth driven by cell proliferation is an exponential process. As a result, even small variations in proliferation rates, when integrated over a relatively long developmental time, will lead to large differences in size. How organs robustly control their final size despite perturbations in cell proliferation rates throughout development is a long-standing question in biology. Using a mathematical model, we show that in the developing wing of the fruit fly, Drosophila melanogaster, variations in proliferation rates of wing-committed cells are inversely proportional to the duration of cell recruitment, a differentiation process in which a population of undifferentiated cells adopt the wing fate by expressing the selector gene, vestigial. A time-course experiment shows that vestigial-expressing cells increase exponentially while recruitment takes place, but slows down when recruitable cells start to vanish, suggesting that undifferentiated cells may be driving proliferation of wing-committed cells. When this observation is incorporated in our model, we show that the duration of cell recruitment robustly determines a final wing size even when cell proliferation rates of wing-committed cells are perturbed. Finally, we show that this control mechanism fails when perturbations in proliferation rates affect both wing-committed and recruitable cells, providing an experimentally testable hypothesis of our model.
Collapse
Affiliation(s)
- Elizabeth Diaz-Torres
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Luis Manuel Muñoz-Nava
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| | - Marcos Nahmad
- Department of Physiology, Biophysics, and Neurosciences, Centre for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Av. Instituto Politecnico Nacional 2508, Colonia San Pedro Zacatenco, Mexico City 07360, Mexico
| |
Collapse
|
10
|
Abstract
Metazoan embryos develop from a single cell into three-dimensional structured organisms while groups of genetically identical cells attain specialized identities. Cells of the developing embryo both create and accurately interpret morphogen gradients to determine their positions and make specific decisions in response. Here, we first cover intellectual roots of morphogen and positional information concepts. Focusing on animal embryos, we then provide a review of current understanding on how morphogen gradients are established and how their spans are controlled. Lastly, we cover how gradients evolve in time and space during development, and how they encode information to control patterning. In sum, we provide a list of patterning principles for morphogen gradients and review recent advances in quantitative methodologies elucidating information provided by morphogens.
Collapse
Affiliation(s)
- M. Fethullah Simsek
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, 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
| |
Collapse
|
11
|
Sharma V, Sarkar B, Mutsuddi M, Mukherjee A. Deltex modulates Dpp morphogen gradient formation and affects the Dpp signaling in Drosophila. J Cell Sci 2022; 135:276290. [PMID: 35950520 DOI: 10.1242/jcs.259658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/29/2022] [Indexed: 11/20/2022] Open
Abstract
Deltex (Dx) is a context-dependent regulator of Notch signaling and regulates Notch signaling in a non-canonical fashion by facilitating the endocytosis of its receptor. In an RNAi- based modifier screen of kinases and phosphatases Thickveins (Tkv), the receptor of Decapentaplegic (Dpp), was identified as one of the interactors of Dx. Dpp, a Drosophila TGF-β/Bone Morphogenetic Protein homolog acts as a morphogen to specify cell fate along the anterior-posterior axis of the wing. Tight regulation of Dpp signaling is thus indispensable for its proper functioning. Here we present Dx as a novel modulator of Dpp signaling. We show evidence for the very first time that dx genetically interacts with dpp and its pathway components. Immunocytochemical analysis shows that Dx co-localizes with Dpp and its receptor Tkv in the Drosophila third instar larval tissues. Further, Dx is also seen to modulate the expression of dpp and its target genes. Here, we attribute this modulation to the endocytosis and trafficking of Dpp through Dx. This study thus presents a whole new avenue of Dpp signaling regulation via the cytoplasmic protein Dx.
Collapse
Affiliation(s)
- Vartika Sharma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| | - Bappi Sarkar
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi-221005, Uttar Pradesh, India
| |
Collapse
|
12
|
Lee HC, Hastings C, Oliveira NMM, Pérez-Carrasco R, Page KM, Wolpert L, Stern CD. 'Neighbourhood watch' model: embryonic epiblast cells assess positional information in relation to their neighbours. Development 2022; 149:275390. [PMID: 35438131 PMCID: PMC9188750 DOI: 10.1242/dev.200295] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/11/2022] [Indexed: 12/21/2022]
Abstract
In many developing and regenerating systems, tissue pattern is established through gradients of informative morphogens, but we know little about how cells interpret these. Using experimental manipulation of early chick embryos, including misexpression of an inducer (VG1 or ACTIVIN) and an inhibitor (BMP4), we test two alternative models for their ability to explain how the site of primitive streak formation is positioned relative to the rest of the embryo. In one model, cells read morphogen concentrations cell-autonomously. In the other, cells sense changes in morphogen status relative to their neighbourhood. We find that only the latter model can account for the experimental results, including some counter-intuitive predictions. This mechanism (which we name the ‘neighbourhood watch’ model) illuminates the classic ‘French Flag Problem’ and how positional information is interpreted by a sheet of cells in a large developing system. Summary: In a large developing system, the chick embryo before gastrulation, cells may interpret gradients of positional signals relative to their neighbours to position the primitive streak, establishing bilateral symmetry.
Collapse
Affiliation(s)
- Hyung Chul Lee
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Cato Hastings
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Nidia M M Oliveira
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rubén Pérez-Carrasco
- Department of Life Sciences, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Karen M Page
- Department of Mathematics, University College London, Gower Street, London WC1E 6BT, UK
| | - Lewis Wolpert
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Claudio D Stern
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
13
|
Long GY, Yang JP, Jin DC, Yang H, Zhou C, Wang Z, Yang XB. Silencing of Decapentaplegic (Dpp) gene inhibited the wing expansion in the white-backed planthopper, Sogatella furcifera (Horváth) (Hemiptera: Delphacidae). ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 110:e21879. [PMID: 35247285 DOI: 10.1002/arch.21879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/21/2021] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
The Decapentaplegic gene controls wing patterning and spreading by regulating downstream genes in many insect species. However, the molecular characteristics, expression, and biological function of Dpp in Sogatella furcifera remain poorly understood. In this study, we cloned the Dpp gene from S. furcifera and examined its expression levels in different development stages, wing typed adults, and tissues. Then, the function of SfDpp gene was analyzed using an RNA interference (RNAi)-based approach. The results showed that the full-length complementary DNA of the SfDpp gene consists of 1034 bp and contains a 954-bp open reading frame encoding 317 amino acids. SfDpp has a transforming growth factor-β (TGF-β) propeptide superfamily domain and a TGF-β superfamily domain, typical of members of the TGF-β superfamily. Quantitative real-time polymerase chain reaction showed that the expression of SfDpp reached its highest expression level 40 min after eclosion. RNAi-based gene silencing inhibited transcript levels of the corresponding messenger RNA in S. furcifera nymphs injected with double-stranded RNA of SfDpp and resulted in death of 29.17% and 26.67% of 4th and 5th instar nymphs, respectively. The wing deformity rate of the adults was 74.12% and 3.41% after SfDpp gene silencing in 4th and 5th instar nymphs, respectively. Examining wing development-associated genes showed that two target genes of Dpp (Vestigial and Spalt) were both dramatically downregulated after SfDpp was silenced. Our results demonstrate that downregulated SfDpp in early development causes wing expansion failure in S. furcifera. Thus, Dpp may be a target gene for restricting the migration of rice-damaging planthoppers.
Collapse
Affiliation(s)
- Gui-Yun Long
- Institute of Entomology, Guizhou University; Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, and Scientific Observing and Experimental Station of Crop Pests in Guiyang, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Guiyang, China
| | - Jia-Peng Yang
- Institute of Entomology, Guizhou University; Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, and Scientific Observing and Experimental Station of Crop Pests in Guiyang, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Guiyang, China
| | - Dao-Chao Jin
- Institute of Entomology, Guizhou University; Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, and Scientific Observing and Experimental Station of Crop Pests in Guiyang, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Guiyang, China
| | - Hong Yang
- Institute of Entomology, Guizhou University; Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, and Scientific Observing and Experimental Station of Crop Pests in Guiyang, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Guiyang, China
- College of Tobacco Science of Guizhou University, Guiyang, China
| | - Cao Zhou
- Institute of Entomology, Guizhou University; Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, and Scientific Observing and Experimental Station of Crop Pests in Guiyang, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Guiyang, China
- College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Zhao Wang
- Institute of Entomology, Guizhou University; Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, and Scientific Observing and Experimental Station of Crop Pests in Guiyang, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Guiyang, China
- College of Environment and Life Sciences, Kaili University, Kaili, China
| | - Xi-Bin Yang
- Institute of Entomology, Guizhou University; Provincial Key Laboratory for Agricultural Pest Management of Mountainous Regions, and Scientific Observing and Experimental Station of Crop Pests in Guiyang, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Guiyang, China
| |
Collapse
|
14
|
Akiyama T, Seidel CW, Gibson MC. The feedback regulator nord controls Dpp/BMP signaling via extracellular interaction with dally in the Drosophila wing. Dev Biol 2022; 488:91-103. [DOI: 10.1016/j.ydbio.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/27/2022]
|
15
|
An exciting period of Drosophila developmental biology: Of imaginal discs, clones, compartments, parasegments and homeotic genes". Dev Biol 2022; 484:12-21. [PMID: 35120908 DOI: 10.1016/j.ydbio.2022.01.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/22/2022]
Abstract
In this review we recall a number of important discoveries that took place in Drosophila during seventies and eighties of the last century. The development of cell lineage methods and of powerful modifications of same, such as the Minute technique, led to the discovery of compartments and provided a clearer picture of the body organization: it came to be seen as a chain of metameric lineage units along the A/P body axis. Further, genetic screens allowed the identification of the genes involved in the establishment of the metameric scaffold - the segmentation genes- and also of Hox genes that are responsible for the specific development of individual body parts. As cloning methods became available, many of the most relevant of these developmental genes were cloned and a molecular analysis of development initiated. The discovery of the homeobox, a molecular mark of the Hox and other relevant developmental genes, allowed the finding of Hox genes in animal species, like humans, in which they could not be identified by genetic methods. Analysis of the structure and function of Hox genes provided a general image of the genetic design of the metazoan body.
Collapse
|
16
|
Yang S, Wu X, Daoutidou EI, Zhang Y, Shimell M, Chuang KH, Peterson AJ, O'Connor MB, Zheng X. The NDNF-like factor Nord is a Hedgehog-induced extracellular BMP modulator that regulates Drosophila wing patterning and growth. eLife 2022; 11:e73357. [PMID: 35037619 PMCID: PMC8856659 DOI: 10.7554/elife.73357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/15/2022] [Indexed: 11/23/2022] Open
Abstract
Hedgehog (Hh) and Bone Morphogenetic Proteins (BMPs) pattern the developing Drosophila wing by functioning as short- and long-range morphogens, respectively. Here, we show that a previously unknown Hh-dependent mechanism fine-tunes the activity of BMPs. Through genome-wide expression profiling of the Drosophila wing imaginal discs, we identify nord as a novel target gene of the Hh signaling pathway. Nord is related to the vertebrate Neuron-Derived Neurotrophic Factor (NDNF) involved in congenital hypogonadotropic hypogonadism and several types of cancer. Loss- and gain-of-function analyses implicate Nord in the regulation of wing growth and proper crossvein patterning. At the molecular level, we present biochemical evidence that Nord is a secreted BMP-binding protein and localizes to the extracellular matrix. Nord binds to Decapentaplegic (Dpp) or the heterodimer Dpp-Glass-bottom boat (Gbb) to modulate their release and activity. Furthermore, we demonstrate that Nord is a dosage-dependent BMP modulator, where low levels of Nord promote and high levels inhibit BMP signaling. Taken together, we propose that Hh-induced Nord expression fine-tunes both the range and strength of BMP signaling in the developing Drosophila wing.
Collapse
Affiliation(s)
- Shu Yang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Xuefeng Wu
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Euphrosyne I Daoutidou
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Ya Zhang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - MaryJane Shimell
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Kun-Han Chuang
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| | - Aidan J Peterson
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Michael B O'Connor
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of MinnesotaMinneapolisUnited States
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology and the GW Cancer Center, George Washington University School of Medicine and Health SciencesWashingtonUnited States
| |
Collapse
|
17
|
Azpiazu N, Morata G. Chromatin remodelling and retrotransposons activities during regeneration in Drosophila. Dev Biol 2021; 482:7-16. [PMID: 34822846 DOI: 10.1016/j.ydbio.2021.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/03/2022]
Abstract
Regeneration is a response mechanism aiming to reconstruct lost or damaged structures. To achieve this, the cells repopulating the lost tissue often have to change their original identity, a process that involves chromatin remodelling.We have analysed the issue of chromatin remodelling during regeneration in the wing disc of Drosophila . In this disc the ablation of the central region (the pouch) induces the regenerative response of the cells from the lateral region (the hinge), which reconstitute the wing pouch. We have examined euchromatin and heterochromatin histone marks during the process and find that heterochromatin marks disappear but are recovered when regeneration is complete. Euchromatin marks are not modified. We also describe the transcription of two retrotransposons, Roo and F-element in the regenerating cells. We have established a temporal correlation between the alterations of heterochromatin marks and the levels of transcription of two retrotransposons, Roo and F-element, both during embryonic development and in the regeneration process.
Collapse
Affiliation(s)
| | - Ginés Morata
- Centro de Biología Molecular CSIC-UAM, Madrid, Spain
| |
Collapse
|
18
|
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
|
19
|
Léopold P. Sizes, proportions and environment. C R Biol 2021; 344:165-175. [PMID: 34213854 DOI: 10.5802/crbiol.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 11/24/2022]
Abstract
The sizes of living organisms range over twenty orders of magnitude. Within the same species, the size of individuals also varies according to the environmental conditions to which they are subjected. From the studies conducted on organisms as diverse as the drosophila, the salamander or the mouse, laws and conserved mechanisms emerge that shed light on the fundamental aspects of growth, but also on more medical issues such as tissue regeneration, metabolic homeostasis and cancer.
Collapse
Affiliation(s)
- Pierre Léopold
- Institut Curie, PSL Research University, CNRS UMR3215, INSERM U934, UPMC Paris-Sorbonne, 26 Rue d'Ulm, 75005, Paris, France
| |
Collapse
|
20
|
Madamanchi A, Mullins MC, Umulis DM. Diversity and robustness of bone morphogenetic protein pattern formation. Development 2021; 148:dev192344. [PMID: 33795238 PMCID: PMC8034876 DOI: 10.1242/dev.192344] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pattern formation by bone morphogenetic proteins (BMPs) demonstrates remarkable plasticity and utility in several contexts, such as early embryonic development, tissue patterning and the maintenance of stem cell niches. BMPs pattern tissues over many temporal and spatial scales: BMP gradients as short as 1-2 cell diameters maintain the stem cell niche of the Drosophila germarium over a 24-h cycle, and BMP gradients of several hundred microns establish dorsal-ventral tissue specification in Drosophila, zebrafish and Xenopus embryos in timescales between 30 min and several hours. The mechanisms that shape BMP signaling gradients are also incredibly diverse. Although ligand diffusion plays a dominant role in forming the gradient, a cast of diffusible and non-diffusible regulators modulate gradient formation and confer robustness, including scale invariance and adaptability to perturbations in gene expression and growth. In this Review, we document the diverse ways that BMP gradients are formed and refined, and we identify the core principles that they share to achieve reliable performance.
Collapse
Affiliation(s)
- Aasakiran Madamanchi
- Agricultural and Biological Engineering. Purdue University, West Lafayette, IN 47907, USA
- Polytechnic Institute, Purdue University, West Lafayette, IN 47907, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David M Umulis
- Agricultural and Biological Engineering. Purdue University, West Lafayette, IN 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| |
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: 7] [Impact Index Per Article: 1.8] [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
|
Abstract
Half a century after Lewis Wolpert's seminal conceptual advance on how cellular fates distribute in space, we provide a brief historical perspective on how the concept of positional information emerged and influenced the field of developmental biology and beyond. We focus on a modern interpretation of this concept in terms of information theory, largely centered on its application to cell specification in the early Drosophila embryo. We argue that a true physical variable (position) is encoded in local concentrations of patterning molecules, that this mapping is stochastic, and that the processes by which positions and corresponding cell fates are determined based on these concentrations need to take such stochasticity into account. With this approach, we shift the focus from biological mechanisms, molecules, genes and pathways to quantitative systems-level questions: where does positional information reside, how it is transformed and accessed during development, and what fundamental limits it is subject to?
Collapse
Affiliation(s)
- Gašper Tkačik
- Institute of Science and Technology Austria, Am Campus 1, AT-3400 Klosterneuburg, Austria
| | - Thomas Gregor
- Joseph Henry Laboratories of Physics and the Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Developmental and Stem Cell Biology, UMR3738, Institut Pasteur, FR-75015 Paris, France
| |
Collapse
|
23
|
Hu Y, Moczek AP. Wing serial homologues and the diversification of insect outgrowths: insights from the pupae of scarab beetles. Proc Biol Sci 2021; 288:20202828. [PMID: 33467999 DOI: 10.1098/rspb.2020.2828] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Modification of serially homologous structures is a common avenue towards functional innovation in developmental evolution, yet ancestral affinities among serial homologues may be obscured as structure-specific modifications accumulate over time. We sought to assess the degree of homology to wings of three types of body wall projections commonly observed in scarab beetles: (i) the dorsomedial support structures found on the second and third thoracic segments of pupae, (ii) the abdominal support structures found bilaterally in most abdominal segments of pupae, and (iii) the prothoracic horns which depending on species and sex may be restricted to pupae or also found in adults. We functionally investigated 14 genes within, as well as two genes outside, the canonical wing gene regulatory network to compare and contrast their role in the formation of each of the three presumed wing serial homologues. We found 11 of 14 wing genes to be functionally required for the proper formation of lateral and dorsal support structures, respectively, and nine for the formation of prothoracic horns. At the same time, we document multiple instances of divergence in gene function across our focal structures. Collectively, our results support the hypothesis that dorsal and lateral support structures as well as prothoracic horns share a developmental origin with insect wings. Our findings suggest that the morphological and underlying gene regulatory diversification of wing serial homologues across species, life stages and segments has contributed significantly to the extraordinary diversity of arthropod appendages and outgrowths.
Collapse
Affiliation(s)
- Yonggang Hu
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Armin P Moczek
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
24
|
Alba V, Carthew JE, Carthew RW, Mani M. Global constraints within the developmental program of the Drosophila wing. eLife 2021; 10:66750. [PMID: 34180394 PMCID: PMC8257256 DOI: 10.7554/elife.66750] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/25/2021] [Indexed: 12/23/2022] Open
Abstract
Organismal development is a complex process, involving a vast number of molecular constituents interacting on multiple spatio-temporal scales in the formation of intricate body structures. Despite this complexity, development is remarkably reproducible and displays tolerance to both genetic and environmental perturbations. This robustness implies the existence of hidden simplicities in developmental programs. Here, using the Drosophila wing as a model system, we develop a new quantitative strategy that enables a robust description of biologically salient phenotypic variation. Analyzing natural phenotypic variation across a highly outbred population and variation generated by weak perturbations in genetic and environmental conditions, we observe a highly constrained set of wing phenotypes. Remarkably, the phenotypic variants can be described by a single integrated mode that corresponds to a non-intuitive combination of structural variations across the wing. This work demonstrates the presence of constraints that funnel environmental inputs and genetic variation into phenotypes stretched along a single axis in morphological space. Our results provide quantitative insights into the nature of robustness in complex forms while yet accommodating the potential for evolutionary variations. Methodologically, we introduce a general strategy for finding such invariances in other developmental contexts.
Collapse
Affiliation(s)
- Vasyl Alba
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States,NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
| | - James E Carthew
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States
| | - Richard W Carthew
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States,Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Madhav Mani
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States,NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States,Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| |
Collapse
|
25
|
Bairzin JCD, Emmons-Bell M, Hariharan IK. The Hippo pathway coactivator Yorkie can reprogram cell fates and create compartment-boundary-like interactions at clone margins. SCIENCE ADVANCES 2020; 6:6/50/eabe8159. [PMID: 33298454 PMCID: PMC7725458 DOI: 10.1126/sciadv.abe8159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/22/2020] [Indexed: 06/12/2023]
Abstract
During development, tissue-specific patterns of gene expression are established by transcription factors and then stably maintained via epigenetic mechanisms. Cancer cells often express genes that are inappropriate for that tissue or developmental stage. Here, we show that high activity levels of Yki, the Hippo pathway coactivator that causes overgrowth in Drosophila imaginal discs, can also disrupt cell fates by altering expression of selector genes like engrailed (en) and Ultrabithorax (Ubx). Posterior clones expressing activated Yki can down-regulate en and express an anterior selector gene, cubitus interruptus (ci). The microRNA bantam and the chromatin regulator Taranis both function downstream of Yki in promoting ci expression. The boundary between Yki-expressing posterior clones and surrounding wild-type cells acquires properties reminiscent of the anteroposterior compartment boundary; Hedgehog signaling pathway activation results in production of Dpp. Thus, at least in principle, heterotypic interactions between Yki-expressing cells and their neighbors could activate boundary-specific signaling mechanisms.
Collapse
Affiliation(s)
- Joanna C D Bairzin
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Maya Emmons-Bell
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, 515 Weill Hall, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
26
|
Affiliation(s)
- Naama Barkai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
27
|
Nagel AC, Maier D, Scharpf J, Ketelhut M, Preiss A. Limited Availability of General Co-Repressors Uncovered in an Overexpression Context during Wing Venation in Drosophila melanogaster. Genes (Basel) 2020; 11:genes11101141. [PMID: 32998295 PMCID: PMC7601384 DOI: 10.3390/genes11101141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022] Open
Abstract
Cell fate is determined by the coordinated activity of different pathways, including the conserved Notch pathway. Activation of Notch results in the transcription of Notch targets that are otherwise silenced by repressor complexes. In Drosophila, the repressor complex comprises the transcription factor Suppressor of Hairless (Su(H)) bound to the Notch antagonist Hairless (H) and the general co-repressors Groucho (Gro) and C-terminal binding protein (CtBP). The latter two are shared by different repressors from numerous pathways, raising the possibility that they are rate-limiting. We noted that the overexpression during wing development of H mutants HdNT and HLD compromised in Su(H)-binding induced ectopic veins. On the basis of the role of H as Notch antagonist, overexpression of Su(H)-binding defective H isoforms should be without consequence, implying different mechanisms but repression of Notch signaling activity. Perhaps excess H protein curbs general co-repressor availability. Supporting this model, nearly normal wings developed upon overexpression of H mutant isoforms that bound neither Su(H) nor co-repressor Gro and CtBP. Excessive H protein appeared to sequester general co-repressors, resulting in specific vein defects, indicating their limited availability during wing vein development. In conclusion, interpretation of overexpression phenotypes requires careful consideration of possible dominant negative effects from interception of limiting factors.
Collapse
|
28
|
Wang J, Dahmann C. Establishing compartment boundaries in Drosophila wing imaginal discs: An interplay between selector genes, signaling pathways and cell mechanics. Semin Cell Dev Biol 2020; 107:161-169. [PMID: 32732129 DOI: 10.1016/j.semcdb.2020.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 01/02/2023]
Abstract
The partitioning of cells into groups or 'compartments' separated by straight and sharp boundaries is important for tissue formation in animal development. Cells from neighboring compartments are characterized by distinct fates and functions and their continuous separation at compartment boundaries maintains proper tissue organization. Signaling across compartment boundaries can induce the local expression of morphogens that in turn direct growth and patterning of the surrounding cells. Compartment boundaries play therefore an important role in tissue development. Compartment boundaries were first identified in the early 1970s in the Drosophila wing. Here, we review the role of compartment boundaries in growth and patterning of the developing wing and then discuss the genetic and physical mechanisms underlying cell separation at compartment boundaries in this tissue.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
29
|
Upadhyay A, Peterson AJ, Kim MJ, O'Connor MB. Muscle-derived Myoglianin regulates Drosophila imaginal disc growth. eLife 2020; 9:e51710. [PMID: 32633716 PMCID: PMC7371420 DOI: 10.7554/elife.51710] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 07/04/2020] [Indexed: 01/05/2023] Open
Abstract
Organ growth and size are finely tuned by intrinsic and extrinsic signaling molecules. In Drosophila, the BMP family member Dpp is produced in a limited set of imaginal disc cells and functions as a classic morphogen to regulate pattern and growth by diffusing throughout imaginal discs. However, the role of TGFβ/Activin-like ligands in disc growth control remains ill-defined. Here, we demonstrate that Myoglianin (Myo), an Activin family member, and a close homolog of mammalian Myostatin (Mstn), is a muscle-derived extrinsic factor that uses canonical dSmad2-mediated signaling to regulate wing size. We propose that Myo is a myokine that helps mediate an allometric relationship between muscles and their associated appendages.
Collapse
Affiliation(s)
- Ambuj Upadhyay
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| | - Aidan J Peterson
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| |
Collapse
|
30
|
The Drosophila melanogaster Metabolic Response against Parasitic Nematode Infection Is Mediated by TGF-β Signaling. Microorganisms 2020; 8:microorganisms8070971. [PMID: 32610560 PMCID: PMC7409035 DOI: 10.3390/microorganisms8070971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 01/01/2023] Open
Abstract
The nematode Heterorhabditis bacteriophora, its mutualistic bacterium Photorhabdus luminescens, and the fruit fly Drosophila melanogaster establish a unique system to study the basis of infection in relation to host metabolism. Our previous results indicate that the Transforming Growth Factor β (TGF-β) signaling pathway participates in the D. melanogaster metabolic response against nematode parasitism. However, our understanding of whether the presence of Photorhabdus bacteria in Heterorhabditis nematodes affects the metabolic state of D. melanogaster during infection is limited. Here, we investigated the involvement of TGF-β signaling branches, Activin and Bone Morphogenetic Protein (BMP), in the D. melanogaster metabolic response against axenic (lacking bacteria) or symbiotic (containing bacteria) H. bacteriophora infection. We show that BMP signaling mediates lipid metabolism against axenic or symbiotic H. bacteriophora and alters the size of fat body lipid droplets against symbiotic nematode infection. Also, following symbiotic H. bacteriophora infection, Activin signaling modulates sugar metabolism. Our results indicate that Activin and BMP signaling interact with the D. melanogaster metabolic response to H. bacteriophora infection regardless of the presence or absence of Photorhabdus. These findings provide evidence for the role of TGF-β signaling in host metabolism, which could lead to the development of novel treatments for parasitic diseases.
Collapse
|
31
|
Vadde BVL, Roeder AHK. Can the French flag and reaction-diffusion models explain flower patterning? Celebrating the 50th anniversary of the French flag model. JOURNAL OF EXPERIMENTAL BOTANY 2020; 71:2886-2897. [PMID: 32016398 DOI: 10.1093/jxb/eraa065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/01/2020] [Indexed: 05/25/2023]
Abstract
It has been 50 years since Lewis Wolpert introduced the French flag model proposing the patterning of different cell types based on threshold concentrations of a morphogen diffusing in the tissue. Sixty-seven years ago, Alan Turing introduced the idea of patterns initiating de novo from a reaction-diffusion network. Together these models have been used to explain many patterning events in animal development, so here we take a look at their applicability to flower development. First, although many plant transcription factors move through plasmodesmata from cell to cell, in the flower there is little evidence that they specify fate in a concentration-dependent manner, so they cannot yet be described as morphogens. Secondly, the reaction-diffusion model appears to be a reasonably good description of the formation of spots of pigment on petals, although additional nuances are present. Thirdly, aspects of both of these combine in a new fluctuation-based patterning system creating the scattered pattern of giant cells in Arabidopsis sepals. In the future, more precise imaging and manipulations of the dynamics of patterning networks combined with mathematical modeling will allow us to better understand how the multilayered complex and beautiful patterns of flowers emerge de novo.
Collapse
Affiliation(s)
- Batthula Vijaya Lakshmi Vadde
- Weill Institute for Cell and Molecular Biology and School of Integrative Plant Science, Section of Plant Biology, Cornell University, Ithaca, NY, USA
| | - Adrienne H K Roeder
- Weill Institute for Cell and Molecular Biology and School of Integrative Plant Science, Section of Plant Biology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
32
|
Waghmare I, Wang X, Page-McCaw A. Dally-like protein sequesters multiple Wnt ligands in the Drosophila germarium. Dev Biol 2020; 464:88-102. [PMID: 32473955 DOI: 10.1016/j.ydbio.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/02/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
Cells in multicellular organisms rely on secreted ligands for development and morphogenesis. Several mechanisms modulate the availability and distribution of secreted ligands, determining their ability to signal locally and at long range from their source. One of these mechanisms is Dally-like protein (Dlp), a cell-surface glypican that exhibits biphasic functions in Drosophila wing discs, promoting Wg signaling at long-range from Wg source cells and inhibiting Wg signaling near source cells. In the germarium at the tip of the ovary, Dlp promotes long-range distribution of Wg from cap cells to follicle stem cells. However, the germarium also expresses other Wnts - Wnt2, Wnt4, and Wnt6 - that function locally in escort cells to promote oogenesis. Whether and how local functions of these Wnts are regulated remains unknown. Here we show that the dlp overexpression phenotype is multifaceted and phenocopies multiple Wnt loss-of-function phenotypes. Each aspect of dlp overexpression phenotype is suppressed by co-expression of individual Wnts, and the suppression pattern exhibited by each Wnt suggests that Wnts have functional specificity in the germarium. Further, dlp knockdown phenocopies Wnt gain-of-function phenotypes. Together these data show that Dlp inhibits the functions of each Wnt. All four Wnts co-immunoprecipitate with Dlp in S2R+ cells, suggesting that in the germarium, Dlp sequesters Wnts to inhibit local paracrine Wnt signaling. Our results indicate that Dlp modulates the availability of multiple extracellular Wnts for local paracrine Wnt signaling in the germarium.
Collapse
Affiliation(s)
- Indrayani Waghmare
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| | - Xiaoxi Wang
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| |
Collapse
|
33
|
Haupaix N, Manceau M. The embryonic origin of periodic color patterns. Dev Biol 2020; 460:70-76. [DOI: 10.1016/j.ydbio.2019.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/02/2019] [Indexed: 01/29/2023]
|
34
|
Huizar F, Soundarrajan D, Paravitorghabeh R, Zartman J. Interplay between morphogen-directed positional information systems and physiological signaling. Dev Dyn 2020; 249:328-341. [PMID: 31794137 PMCID: PMC7328709 DOI: 10.1002/dvdy.140] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 12/16/2022] Open
Abstract
The development of an organism from an undifferentiated single cell into a spatially complex structure requires spatial patterning of cell fates across tissues. Positional information, proposed by Lewis Wolpert in 1969, has led to the characterization of many components involved in regulating morphogen signaling activity. However, how morphogen gradients are established, maintained, and interpreted by cells still is not fully understood. Quantitative and systems-based approaches are increasingly needed to define general biological design rules that govern positional information systems in developing organisms. This short review highlights a selective set of studies that have investigated the roles of physiological signaling in modulating and mediating morphogen-based pattern formation. Similarities between neural transmission and morphogen-based pattern formation mechanisms suggest underlying shared principles of active cell-based communication. Within larger tissues, neural networks provide directed information, via physiological signaling, that supplements positional information through diffusion. Further, mounting evidence demonstrates that physiological signaling plays a role in ensuring robustness of morphogen-based signaling. We conclude by highlighting several outstanding questions regarding the role of physiological signaling in morphogen-based pattern formation. Elucidating how physiological signaling impacts positional information is critical for understanding the close coupling of developmental and cellular processes in the context of development, disease, and regeneration.
Collapse
Affiliation(s)
- Francisco Huizar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, South Bend, Indiana
- Bioengineering Graduate Program, University of Notre Dame, South Bend, Indiana
| | - Dharsan Soundarrajan
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, South Bend, Indiana
| | - Ramezan Paravitorghabeh
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, South Bend, Indiana
| | - Jeremiah Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, South Bend, Indiana
- Bioengineering Graduate Program, University of Notre Dame, South Bend, Indiana
| |
Collapse
|
35
|
Barrio L, Milán M. Regulation of Anisotropic Tissue Growth by Two Orthogonal Signaling Centers. Dev Cell 2020; 52:659-672.e3. [PMID: 32084357 DOI: 10.1016/j.devcel.2020.01.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 11/15/2022]
Abstract
The Drosophila wing has served as a paradigm to mechanistically characterize the role of morphogens in patterning and growth. Wingless (Wg) and Decapentaplegic (Dpp) are expressed in two orthogonal signaling centers, and their gradients organize patterning by regulating the expression of well-defined target genes. By contrast, graded activity of these morphogens is not an absolute requirement for wing growth. Despite their permissive role in regulating growth, here we show that Wg and Dpp are utilized in a non-interchangeable manner by the two existing orthogonal signaling centers to promote preferential growth along the two different axes of the developing wing. Our data indicate that these morphogens promote anisotropic growth by making use of distinct and non-interchangeable molecular mechanisms. Whereas Dpp drives growth along the anterior-posterior axis by maintaining Brinker levels below a growth-repressing threshold, Wg exerts its action along the proximal-distal axis through a double repression mechanism involving T cell factor (TCF).
Collapse
Affiliation(s)
- Lara Barrio
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Marco Milán
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats, Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
36
|
Camacho-Aguilar E, Warmflash A. Insights into mammalian morphogen dynamics from embryonic stem cell systems. Curr Top Dev Biol 2020; 137:279-305. [PMID: 32143746 PMCID: PMC7713707 DOI: 10.1016/bs.ctdb.2019.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Morphogens play an essential role in cell fate specification and patterning including in laying out the mammalian body plan during gastrulation. In vivo studies have shed light on the signaling pathways involved in this process and the phenotypes associated with their disruption, however, several important open questions remain regarding how morphogens function in space and time. Self-organized patterning systems based on embryonic stem cells have emerged as a powerful platform for beginning to address these questions that is complementary to in vivo approaches. Here we review recent progress in understanding morphogen signaling dynamics and patterning in early mammalian development by taking advantage of cutting-edge embryonic stem cell technology.
Collapse
Affiliation(s)
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, TX, United States; Department of Bioengineering, Rice University, Houston, TX, United States.
| |
Collapse
|
37
|
Rahimi N, Averbukh I, Carmon S, Schejter ED, Barkai N, Shilo BZ. Dynamics of Spaetzle morphogen shuttling in the Drosophila embryo shapes gastrulation patterning. Development 2019; 146:146/21/dev181487. [PMID: 31719046 DOI: 10.1242/dev.181487] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/07/2019] [Indexed: 12/27/2022]
Abstract
Establishment of morphogen gradients in the early Drosophila embryo is challenged by a diffusible sextracellular milieu, and by rapid nuclear divisions that occur at the same time. To understand how a sharp gradient is formed within this dynamic environment, we followed the generation of graded nuclear Dorsal protein, the hallmark of pattern formation along the dorso-ventral axis, in live embryos. The dynamics indicate that a sharp extracellular gradient is formed through diffusion-based shuttling of the Spaetzle (Spz) morphogen that progresses through several nuclear divisions. Perturbed shuttling in wntD mutant embryos results in a flat activation peak and aberrant gastrulation. Re-entry of Dorsal into the nuclei at the final division cycle plays an instructive role, as the residence time of Dorsal in each nucleus is translated to the amount of zygotic transcript that will be produced, thereby guiding graded accumulation of specific zygotic transcripts that drive patterned gastrulation. We conclude that diffusion-based ligand shuttling, coupled with dynamic readout, establishes a refined pattern within the diffusible environment of early embryos.
Collapse
Affiliation(s)
- Neta Rahimi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Inna Averbukh
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shari Carmon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Naama Barkai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ben-Zion Shilo
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
38
|
Svendsen PC, Phillips LA, Deshwar AR, Ryu JR, Najand N, Brook WJ. The selector genes midline and H15 control ventral leg pattern by both inhibiting Dpp signaling and specifying ventral fate. Dev Biol 2019; 455:19-31. [PMID: 31299230 DOI: 10.1016/j.ydbio.2019.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 04/27/2019] [Accepted: 05/28/2019] [Indexed: 01/13/2023]
Abstract
mid and H15 encode Tbx20 transcription factors that specify ventral pattern in the Drosophila leg. We find that there are at least two pathways for mid and H15 specification of ventral fate. In the first pathway, mid and H15 negatively regulate Dpp, the dorsal signal in leg development. mid and H15 block the dorsalizing effects of Dpp signaling in the ventral leg. In loss- and gain-of-function experiments in imaginal discs, we show that mid and H15 block the accumulation of phospho-Mad, the activated form of the Drosophila pSmad1/5 homolog. In a second pathway, we find mid and H15 must also directly promote ventral fate because simultaneously blocking Dpp signaling in mid H15 mutants does not rescue the ventral to dorsal transformation in most ventral leg structures. We show that mid and H15 act as transcriptional repressors in ventral leg development. The two genes repress the Dpp target gene Dad, the laterally expressed gene Upd, and the mid VLE enhancer. This repression depends on the eh1 domain, a binding site for the Groucho co-repressor, and is likely direct because Mid localizes to target gene enhancers in PCR-ChIP assays. A mid allele mutant for the repressing domain (eh1), mideh1, was found to be compromised in gain-of-function assays and in rescue of mid H15 loss-of-function. We propose that mid and H15 specify ventral fate through inhibition of Dpp signaling and through coordinating the repression of genes in the ventral leg.
Collapse
Affiliation(s)
- Pia C Svendsen
- Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada
| | - Lindsay A Phillips
- Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada
| | - Ashish R Deshwar
- Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada
| | - Jae-Ryeon Ryu
- Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada
| | - Nima Najand
- Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada
| | - William J Brook
- Alberta Children's Hospital Research Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N4N1, Canada.
| |
Collapse
|
39
|
Pickering J, Chinnaiya K, Towers M. An autoregulatory cell cycle timer integrates growth and specification in chick wing digit development. eLife 2019; 8:47625. [PMID: 31545166 PMCID: PMC6777937 DOI: 10.7554/elife.47625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/23/2019] [Indexed: 12/30/2022] Open
Abstract
A fundamental question is how proliferation and growth are timed during embryogenesis. Although it has been suggested that the cell cycle could be a timer, the underlying mechanisms remain elusive. Here we describe a cell cycle timer that operates in Sonic hedgehog (Shh)-expressing polarising region cells of the chick wing bud. Our data are consistent with Shh signalling stimulating polarising region cell proliferation via Cyclin D2, and then inhibiting proliferation via a Bmp2-p27kip1 pathway. When Shh signalling is blocked, polarising region cells over-proliferate and form an additional digit, which can be prevented by applying Bmp2 or by inhibiting D cyclin activity. In addition, Bmp2 also restores posterior digit identity in the absence of Shh signalling, thus indicating that it specifies antero-posterior (thumb to little finger) positional values. Our results reveal how an autoregulatory cell cycle timer integrates growth and specification and are widely applicable to many tissues.
Collapse
Affiliation(s)
- Joseph Pickering
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Kavitha Chinnaiya
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Matthew Towers
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
40
|
Deng M, Wang Y, Zhang L, Yang Y, Huang S, Wang J, Ge H, Ishibashi T, Yan Y. Single cell transcriptomic landscapes of pattern formation, proliferation and growth in Drosophila wing imaginal discs. Development 2019; 146:dev.179754. [PMID: 31455604 DOI: 10.1242/dev.179754] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022]
Abstract
Organ formation relies on the orchestration of pattern formation, proliferation and growth during development. How these processes are integrated at the individual cell level remains unclear. In the past decades, studies using Drosophila wing imaginal discs as a model system have provided valuable insights into pattern formation, growth control and regeneration. Here, we provide single cell transcriptomic landscapes of pattern formation, proliferation and growth of wing imaginal discs. We found that patterning information is robustly maintained in the single cell transcriptomic data and can provide reference matrices for computationally mapping single cells into discrete spatial domains. Assignment of wing disc single cells to spatial subregions facilitates examination of patterning refinement processes. We also clustered single cells into different proliferation and growth states and evaluated the correlation between cell proliferation/growth states and spatial patterning. Furthermore, single cell transcriptomic analyses allowed us to quantitatively examine disturbances of differentiation, proliferation and growth in a well-established tumor model. We provide a database to explore these datasets at http://drosophilayanlab-virtual-wingdisc.ust.hk:3838/v2/This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Mingxi Deng
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Wang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Lina Zhang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yang Yang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Shengshuo Huang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jiguang Wang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Hao Ge
- Beijing International Center for Mathematical Research and Biomedical Pioneering Innovation Center, Peking University, Peking, China 100871
| | - Toyotaka Ishibashi
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yan Yan
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China .,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
41
|
Wolf D, Smylla TK, Reichmuth J, Hoffmeister P, Kober L, Zimmermann M, Turkiewicz A, Borggrefe T, Nagel AC, Oswald F, Preiss A, Maier D. Nucleo-cytoplasmic shuttling of Drosophila Hairless/Su(H) heterodimer as a means of regulating Notch dependent transcription. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1520-1532. [PMID: 31326540 DOI: 10.1016/j.bbamcr.2019.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/02/2019] [Accepted: 07/16/2019] [Indexed: 12/21/2022]
Abstract
Activation and repression of Notch target genes is mediated by transcription factor CSL, known as Suppressor of Hairless (Su(H)) in Drosophila and CBF1 or RBPJ in human. CSL associates either with co-activator Notch or with co-repressors such as Drosophila Hairless. The nuclear translocation of transcription factor CSL relies on co-factor association, both in mammals and in Drosophila. The Drosophila CSL orthologue Su(H) requires Hairless for repressor complex formation. Based on its role in transcriptional silencing, H protein would be expected to be strictly nuclear. However, H protein is also cytosolic, which may relate to its role in the stabilization and nuclear translocation of Su(H) protein. Here, we investigate the function of the predicted nuclear localization signals (NLS 1-3) and single nuclear export signal (NES) of co-repressor Hairless using GFP-fusion proteins, reporter assays and in vivo analyses using Hairless wild type and shuttling-defective Hairless mutants. We identify NLS3 and NES to be critical for Hairless function. In fact, H⁎NLS3 mutant flies match H null mutants, whereas H⁎NLS3⁎NES double mutants display weaker phenotypes in agreement with a crucial role for NES in H export. As expected for a transcriptional repressor, Notch target genes are deregulated in H⁎NLS3 mutant cells, demonstrating nuclear requirement for its activity. Importantly, we reveal that Su(H) protein strictly follows Hairless protein localization. Together, we propose that shuttling between the nucleo-cytoplasmic compartments provides the possibility to fine tune the regulation of Notch target gene expression by balancing of Su(H) protein availability for Notch activation.
Collapse
Affiliation(s)
- Dorina Wolf
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Thomas K Smylla
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Jan Reichmuth
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Philipp Hoffmeister
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Ludmilla Kober
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Mirjam Zimmermann
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Aleksandra Turkiewicz
- Justus-Liebig University of Giessen Institute of Biochemistry, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Tilman Borggrefe
- Justus-Liebig University of Giessen Institute of Biochemistry, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Anja C Nagel
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Franz Oswald
- University Medical Center Ulm, Center for Internal Medicine, Department of Internal Medicine I, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Anette Preiss
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany
| | - Dieter Maier
- University of Hohenheim, Institute of Genetics (240a), Garbenstr. 30, 70599 Stuttgart, Germany.
| |
Collapse
|
42
|
Ozakman Y, Eleftherianos I. TGF-β Signaling Interferes With the Drosophila Innate Immune and Metabolic Response to Parasitic Nematode Infection. Front Physiol 2019; 10:716. [PMID: 31316388 PMCID: PMC6611403 DOI: 10.3389/fphys.2019.00716] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 05/23/2019] [Indexed: 01/23/2023] Open
Abstract
The common fruit fly, Drosophila melanogaster, is an outstanding model to study the molecular basis of anti-pathogen immunity. The parasitic nematode Heterorhabditis gerrardi, together with its mutualistic bacteria Photorhabdus asymbiotica, infects a wide range of insects, including D. melanogaster. Recently, we have shown that transforming growth factor-β (TGF-ß) signaling in D. melanogaster is regulated in response to parasitic nematode infection. In the current study, we investigated the contribution of two TGF-ß signaling branches, the activin and the bone morphogenetic protein (BMP), to D. melanogaster immune function against H. gerrardi. We used D. melanogaster larvae carrying mutations in the genes coding for the TGF-ß extracellular ligands daw and dpp. We have demonstrated that the number of circulating hemocytes in uninfected daw and dpp mutants decreases twofold compared to background controls, yet no significant changes in hemocyte numbers and survival of the TGF-ß mutants are observed upon nematode infection. However, we have shown that nematode-infected daw mutants express Dual oxidase at higher levels and phenoloxidase activity at lower levels compared to their background controls. To elucidate the contribution of TGF-ß signaling in the metabolic response of D. melanogaster to parasitic nematodes, we estimated lipid and carbohydrate levels in daw and dpp mutant larvae infected with H. gerrardi. We have found that both nematode-infected mutants contain lipid droplets of larger size, with daw mutant larvae also containing elevated glycogen levels. Overall, our findings indicate that the regulation of activin and BMP branches of TGF-ß signaling can alter the immune and metabolic processes in D. melanogaster during response to parasitic nematode infection. Results from this study shed light on the molecular signaling pathways insects activate to regulate mechanisms for fighting potent nematode parasites, which could lead to the identification of novel management strategies for the control of damaging pests.
Collapse
Affiliation(s)
- Yaprak Ozakman
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, The George Washington University, Washington, DC, United States
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Laboratory, Department of Biological Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
43
|
Tian A, Duwadi D, Benchabane H, Ahmed Y. Essential long-range action of Wingless/Wnt in adult intestinal compartmentalization. PLoS Genet 2019; 15:e1008111. [PMID: 31194729 PMCID: PMC6563961 DOI: 10.1371/journal.pgen.1008111] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Signal transduction activated by Wingless/Wnt ligands directs cell proliferation and fate specification in metazoans, and its overactivation underlies the development of the vast majority of colorectal cancers. In the conventional model, the secretion and movement of Wingless to cells distant from its source of synthesis are essential for long-range signaling in tissue patterning. However, this model was upended recently by an unanticipated finding: replacement of wild-type Drosophila Wingless with a membrane-tethered form produced viable adults with largely normal external morphology, which suggested that Wingless secretion and movement are dispensable for tissue patterning. Herein, we tested this foundational principle in the adult intestine, where Wingless signaling gradients coincide with all major boundaries between compartments. We find that the critical roles of Wingless during adult intestinal development, which include regulation of target gene activation, boundary formation, stem cell proliferation, epithelial cell fate specification, muscle differentiation, gut folding, and signaling crosstalk with the Decapentaplegic pathway, are all disrupted by Wingless tethering. These findings provide new evidence that supports the requirement for the direct, long-range action of Wingless in tissue patterning, with relevance for animal development, tissue homeostasis and Wnt-driven disease.
Collapse
Affiliation(s)
- Ai Tian
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Deepesh Duwadi
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Hassina Benchabane
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| | - Yashi Ahmed
- Department of Molecular and Systems Biology and the Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States of America
| |
Collapse
|
44
|
Connahs H, Tlili S, van Creij J, Loo TYJ, Banerjee TD, Saunders TE, Monteiro A. Activation of butterfly eyespots by Distal-less is consistent with a reaction-diffusion process. Development 2019; 146:dev169367. [PMID: 30992277 PMCID: PMC6526720 DOI: 10.1242/dev.169367] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 04/05/2019] [Indexed: 01/08/2023]
Abstract
Eyespots on the wings of nymphalid butterflies represent colorful examples of pattern formation, yet the developmental origins and mechanisms underlying eyespot center differentiation are still poorly understood. Using CRISPR-Cas9 we re-examine the function of Distal-less (Dll) as an activator or repressor of eyespots, a topic that remains controversial. We show that the phenotypic outcome of CRISPR mutations depends upon which specific exon is targeted. In Bicyclus anynana, exon 2 mutations are associated with both missing and ectopic eyespots, and also exon skipping. Exon 3 mutations, which do not lead to exon skipping, produce only null phenotypes, including missing eyespots, lighter wing coloration and loss of scales. Reaction-diffusion modeling of Dll function, using Wnt and Dpp as candidate morphogens, accurately replicates these complex crispant phenotypes. These results provide new insight into the function of Dll as a potential activator of eyespot development, scale growth and melanization, and suggest that the tuning of Dll expression levels can generate a diversity of eyespot phenotypes, including their appearance on the wing.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Heidi Connahs
- Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Sham Tlili
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Jelle van Creij
- Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Tricia Y J Loo
- Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Tirtha Das Banerjee
- Department of Biological Sciences, National University of Singapore, Singapore 117558
| | - Timothy E Saunders
- Department of Biological Sciences, National University of Singapore, Singapore 117558
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Institute of Molecular and Cell Biology, A*Star, Proteos, Singapore 138673
| | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, Singapore 117558
- Yale-NUS College, Singapore 138527
| |
Collapse
|
45
|
Lu Y, Yao Y, Li Z. Ectopic Dpp signaling promotes stem cell competition through EGFR signaling in the Drosophila testis. Sci Rep 2019; 9:6118. [PMID: 30992503 PMCID: PMC6467874 DOI: 10.1038/s41598-019-42630-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 04/02/2019] [Indexed: 12/28/2022] Open
Abstract
Stem cell competition could select the fittest stem cells and potentially control tumorigenesis. However, little is known about the underlying molecular mechanisms. Here, we find that ectopic Decapentaplegic (Dpp) signal activation by expressing a constitutively active form of Thickveins (TkvCA) in cyst stem cells (CySCs) leads to competition between CySCs and germline stem cells (GSCs) for niche occupancy and GSC loss. GSCs are displaced from the niche and undergo differentiation. Interestingly, we find that induction of TkvCA results in elevated expression of vein, which further activates Epidermal Growth Factor Receptor (EGFR) signaling in CySCs to promote their proliferation and compete GSCs out of the niche. Our findings elucidate the important role of Dpp signaling in regulating stem cell competition and tumorigenesis, which could be shed light on tumorigenesis and cancer treatment in mammals.
Collapse
Affiliation(s)
- Yanfen Lu
- College of Plant Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Changping District, Beijing, 102206, China
| | - Yuncong Yao
- College of Plant Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Changping District, Beijing, 102206, China.
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing, 100048, China.
| |
Collapse
|
46
|
The genes expression difference between winged and wingless bird cherry-oat aphid Rhopalosiphum padi based on transcriptomic data. Sci Rep 2019; 9:4754. [PMID: 30894649 PMCID: PMC6426873 DOI: 10.1038/s41598-019-41348-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/23/2019] [Indexed: 01/05/2023] Open
Abstract
Aphids produce wing and wingless morphs, depending on the environmental conditions during their complex life cycles. Wing and wingless variations play an important role in migration and host alternation, affecting the migration and host alternation processes. Several transcriptional studies have concentrated on aphids and sought to determine how an organism perceives environmental cues and responds in a plastic manner, but the underlying mechanisms have remained unclear. Therefore, to better understand the molecular mechanisms underlying the wing polyphenism of this fascinating phenomenon, we provide the first report concerning the wing development of aphids in bird cherry-oat aphid Rhopalosiphum padi with comparative transcriptional analysis of all the developmental stages by RNA-Seq. We identified several candidate genes related to biogenic amines and hormones that may be specifically involved in wing development. Moreover, we found that the third instar stage might be a critical stage for visibility of alternative morphs as well as changes in the expression of thirty-three genes associated with wing development. Several genes, i.e., Wnt2, Fng, Uba1, Hh, Foxo, Dpp, Brk, Ap, Dll, Hth, Tsh, Nub, Scr, Antp, Ubx, Asc, Srf and Fl, had different expression levels in different developmental stages and may play important roles in regulating wing polyphenism.
Collapse
|
47
|
Brodskiy PA, Wu Q, Soundarrajan DK, Huizar FJ, Chen J, Liang P, Narciso C, Levis MK, Arredondo-Walsh N, Chen DZ, Zartman JJ. Decoding Calcium Signaling Dynamics during Drosophila Wing Disc Development. Biophys J 2019; 116:725-740. [PMID: 30704858 PMCID: PMC6382932 DOI: 10.1016/j.bpj.2019.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/04/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023] Open
Abstract
The robust specification of organ development depends on coordinated cell-cell communication. This process requires signal integration among multiple pathways, relying on second messengers such as calcium ions. Calcium signaling encodes a significant portion of the cellular state by regulating transcription factors, enzymes, and cytoskeletal proteins. However, the relationships between the inputs specifying cell and organ development, calcium signaling dynamics, and final organ morphology are poorly understood. Here, we have designed a quantitative image-analysis pipeline for decoding organ-level calcium signaling. With this pipeline, we extracted spatiotemporal features of calcium signaling dynamics during the development of the Drosophila larval wing disc, a genetic model for organogenesis. We identified specific classes of wing phenotypes that resulted from calcium signaling pathway perturbations, including defects in gross morphology, vein differentiation, and overall size. We found four qualitative classes of calcium signaling activity. These classes can be ordered based on agonist stimulation strength Gαq-mediated signaling. In vivo calcium signaling dynamics depend on both receptor tyrosine kinase/phospholipase C γ and G protein-coupled receptor/phospholipase C β activities. We found that spatially patterned calcium dynamics correlate with known differential growth rates between anterior and posterior compartments. Integrated calcium signaling activity decreases with increasing tissue size, and it responds to morphogenetic perturbations that impact organ growth. Together, these findings define how calcium signaling dynamics integrate upstream inputs to mediate multiple response outputs in developing epithelial organs.
Collapse
Affiliation(s)
- Pavel A Brodskiy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Qinfeng Wu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Dharsan K Soundarrajan
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Francisco J Huizar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Jianxu Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Peixian Liang
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Cody Narciso
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Megan K Levis
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | | | - Danny Z Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Jeremiah J Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana.
| |
Collapse
|
48
|
Smylla TK, Meier M, Preiss A, Maier D. The Notch repressor complex in Drosophila: in vivo analysis of Hairless mutants using overexpression experiments. Dev Genes Evol 2019; 229:13-24. [PMID: 30612166 DOI: 10.1007/s00427-018-00624-2] [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: 09/14/2018] [Accepted: 12/17/2018] [Indexed: 10/27/2022]
Abstract
During development of higher animals, the Notch signalling pathway governs cell type specification by mediating appropriate gene expression responses. In the absence of signalling, Notch target genes are silenced by repressor complexes. In the model organism Drosophila melanogaster, the repressor complex includes the transcription factor Suppressor of Hairless [Su(H)] and Hairless (H) plus general co-repressors. Recent crystal structure analysis of the Drosophila Notch repressor revealed details of the Su(H)-H complex. They were confirmed by mutational analyses of either protein; however, only Su(H) mutants have been further studied in vivo. Here, we analyse three H variants predicted to affect Su(H) binding. To this end, amino acid replacements Phenylalanine 237, Leucines 245 and 247, as well as Tryptophan 258 to Alanine were introduced into the H protein. A cell-based reporter assay indicates substantial loss of Su(H) binding to the respective mutant proteins HFA, HLLAA and HWA. For in vivo analysis, UAS-lines HFA, HLLAA and HWA were generated to allow spatially restricted overexpression. In these assays, all three mutants resembled the HLD control, shown before to lack Su(H) binding, indicating a strong reduction of H activity. For example, the H variants were impaired in wing margin formation, but unexpectedly induced ectopic wing venation. Concurrent overexpression with Su(H), however, suggests that all mutant H protein isoforms are still able to bind Su(H) in vivo. We conclude that a weakening of the cohesion in the H-Su(H) repressor complex is sufficient for disrupting its in vivo functionality.
Collapse
Affiliation(s)
- Thomas K Smylla
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| | - Markus Meier
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| | - Anette Preiss
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany
| | - Dieter Maier
- Institut für Genetik, Universität Hohenheim, Garbenstr. 30, 70599, Stuttgart, Germany.
| |
Collapse
|
49
|
Vollmer J, Casares F, Iber D. Growth and size control during development. Open Biol 2018; 7:rsob.170190. [PMID: 29142108 PMCID: PMC5717347 DOI: 10.1098/rsob.170190] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/17/2017] [Indexed: 11/30/2022] Open
Abstract
The size and shape of organs are characteristic for each species. Even when organisms develop to different sizes due to varying environmental conditions, such as nutrition, organ size follows species-specific rules of proportionality to the rest of the body, a phenomenon referred to as allometry. Therefore, for a given environment, organs stop growth at a predictable size set by the species's genotype. How do organs stop growth? How can related species give rise to organs of strikingly different size? No definitive answer has been given to date. One of the major models for the studies of growth termination is the vinegar fly Drosophila melanogaster. Therefore, this review will focus mostly on work carried out in Drosophila to try to tease apart potential mechanisms and identify routes for further investigation. One general rule, found across the animal kingdom, is that the rate of growth declines with developmental time. Therefore, answers to the problem of growth termination should explain this seemingly universal fact. In addition, growth termination is intimately related to the problems of robustness (i.e. precision) and plasticity in organ size, symmetric and asymmetric organ development, and of how the ‘target’ size depends on extrinsic, environmental factors.
Collapse
Affiliation(s)
- Jannik Vollmer
- D-BSSE, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland.,Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| | - Fernando Casares
- CABD, CSIC-Universidad Pablo de Olavide-JA, 41013 Seville, Spain
| | - Dagmar Iber
- D-BSSE, ETH Zürich, Mattenstrasse 26, 4058 Basel, Switzerland .,Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| |
Collapse
|
50
|
Akiyama T, User SD, Gibson MC. Somatic clones heterozygous for recessive disease alleles of BMPR1A exhibit unexpected phenotypes in Drosophila. eLife 2018; 7:35258. [PMID: 29745898 PMCID: PMC5963922 DOI: 10.7554/elife.35258] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/02/2018] [Indexed: 01/02/2023] Open
Abstract
The majority of mutations studied in animal models are designated as recessive based on the absence of visible phenotypes in germline heterozygotes. Accordingly, genetic studies primarily rely on homozygous loss-of-function to determine gene requirements, and a conceptually-related ‘two-hit model’ remains the central paradigm in cancer genetics. Here we investigate pathogenesis due to somatic mutation in epithelial tissues, a process that predominantly generates heterozygous cell clones. To study somatic mutation in Drosophila, we generated inducible alleles that mimic human Juvenile polyposis-associated BMPR1A mutations. Unexpectedly, four of these mutations had no phenotype in heterozygous carriers but exhibited clear tissue-level effects when present in somatic clones of heterozygous cells. We conclude that these alleles are indeed recessive when present in the germline, but nevertheless deleterious when present in heterozygous clones. This unforeseen effect, deleterious heteromosaicism, suggests a ‘one-hit’ mechanism for disease initiation that may explain some instances of pathogenesis associated with spontaneous mutation.
Collapse
Affiliation(s)
- Takuya Akiyama
- Stowers Institute for Medical Research, Kansas City, United States
| | - Sırma D User
- Stowers Institute for Medical Research, Kansas City, United States
| | - Matthew C Gibson
- Stowers Institute for Medical Research, Kansas City, United States.,Department of Anatomy and Cell Biology, The University of Kansas School of Medicine, Kansas City, United States
| |
Collapse
|