1
|
Julick CR, Thanintorn N, Zhang H, Tsatskis Y, Glaeser M, Qu Y, Rusch J, McNeill H. Regulation of Hippo signaling and planar cell polarity via distinct regions of the Fat intracellular domain. Development 2025; 152:dev204694. [PMID: 40377178 DOI: 10.1242/dev.204694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 05/08/2025] [Indexed: 05/18/2025]
Abstract
The large Drosophila protocadherin Fat (Ft) is a receptor for signal transduction pathways that control growth (Hippo signaling), planar cell polarity (PCP), metabolism and the proximodistal patterning of appendages. The intracellular domain (ICD) of Ft is crucial in implementing its biological functions. Six regions of high conservation (named A-F) within the ICD have been identified, as well as distinct regions mediating Hippo pathway activity that have been functionally characterized via transgenic expression rescue assays. Here, we make targeted deletions of these highly conserved residues and the putative Hippo- and PCP-regulating domains of endogenous Ft using CRISPR/Cas9. Through transcriptomic, developmental and phenotypic analyses, we show that different regions of Ft contribute uniquely to chromatin dynamics, tissue morphogenesis, PCP and metabolic regulation. We also demonstrate that different regions of Ft regulate growth in opposite directions, with regions B and F promoting growth and region D inhibiting growth. Strikingly, conserved regions D and F are key regulators of the function of Ft in Hippo activity - exhibiting opposing effects on Hippo pathway modulation - and of the conserved regions, and D is the main regulator of PCP.
Collapse
Affiliation(s)
- Cole R Julick
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Nattapon Thanintorn
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Hongtao Zhang
- School of Life Sciences, Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, Shandong 266237, China
| | - Yonit Tsatskis
- Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Megan Glaeser
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Yi Qu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health Systems, Toronto, ON M5G 1X5, Canada
| | - Jannette Rusch
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
2
|
Ding Y, Chen Q. Wnt/β-catenin signaling pathway: an attractive potential therapeutic target in osteosarcoma. Front Oncol 2025; 14:1456959. [PMID: 40028002 PMCID: PMC11867957 DOI: 10.3389/fonc.2024.1456959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/24/2024] [Indexed: 03/05/2025] Open
Abstract
Osteosarcoma (OS) is the most common bone malignancy in children and adolescents, and although current neoadjuvant chemotherapy has shown efficacy against OS, the long-term survival rate for patients with OS remains low, highlighting the need to find more effective treatments. In cancer cells, abnormal activation of signaling pathways can widely affect cell activity from growth and proliferation to apoptosis, invasion and metastasis. Wnt/β-catenin is a complex and unique signaling pathway that is considered to be one of the most important carcinogenic pathways in human cancer. Research have confirmed that the Wnt/β-catenin signaling pathway is an important driving factor for the occurrence and development of osteosarcoma, and abnormal activation of this pathway can promote the pathological processes of cell proliferation, invasion, migration, tumor angiogenesis and chemical resistance of osteosarcoma. However, inhibition of Wnt/β-catenin signaling pathway can effectively inhibit or reverse the above pathological processes. Therefore, manipulating the expression or function of the Wnt/β-catenin pathway may be a potential targeted pathway for the treatment of OS. In this review, we describe the characteristics of the Wnt/β-catenin signaling pathway and summarize the role and mechanism of this pathway in OS. This paper discusses the therapeutic significance of inhibiting or targeting Wnt/β-catenin pathway in OS and the shortcomings of current studies on this pathway in OS and the problems to be solved. This review helps us to understand the role of Wnt/β-catenin on OS, and provides a theoretical basis and new ideas for targeting Wnt/β-catenin pathway as a therapeutic target for OS.
Collapse
Affiliation(s)
- Yi Ding
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, China
- Department of Spine Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Qin Chen
- Department of Spine Surgery, Ganzhou People's Hospital, Ganzhou, China
- Department of Spine Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| |
Collapse
|
3
|
García-Castro P, Giambó-Falian I, Carvacho I, Fuentes R. Phenogenetics of cortical granule dynamics during zebrafish oocyte-to-embryo transition. Front Cell Dev Biol 2025; 13:1514461. [PMID: 39949602 PMCID: PMC11821946 DOI: 10.3389/fcell.2025.1514461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Fertilization is a critical process in sexual reproduction that involves the fusion of a capacitated sperm with a mature oocyte to form a zygote. Polyspermy, the fertilization of an oocyte by multiple sperm, leads to polyploidy and embryo lethality. Mammalian and non-mammalian oocytes have evolved mechanisms to prevent polyspermy, including fast and slow blocks. The fast block comprises membrane depolarization post-sperm fusion, temporarily preventing additional sperm fusion. The slow block, triggered by cortical granule (CG) exocytosis, involves the release of proteins that modify the zona pellucida to form a permanent barrier, avoiding the fertilization by additional sperm. The evidence shows that immature oocytes often fail to prevent polyspermy due to ineffective CG exocytosis, attributed to impaired intracellular calcium increases, lower content of this ion, and incomplete CG migration. The study of how genetic variations lead to observable phenotypes (phenogenetics) during the oocyte-to-embryo transition, have identified several maternal-effect genes in zebrafish involved in CG behavior. These genes regulate various stages of CG biology, including biosynthesis, maturation, and exocytosis. Mutations in these genes disrupt these processes, highlighting the maternal genetic control over CG properties. Zebrafish has emerged as a pivotal model for understanding the evolving genetic regulation and molecular mechanisms underlying CG biology, providing valuable insights into fertility and early embryonic development.
Collapse
Affiliation(s)
- Priscila García-Castro
- Laboratorio de Fenómica y Embriogénesis Temprana (LAFET), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Isabella Giambó-Falian
- Laboratorio de Fenómica y Embriogénesis Temprana (LAFET), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ingrid Carvacho
- Laboratorio de Canales Iónicos y Reproducción (CIR), Departamento de Medicina Translacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Ricardo Fuentes
- Laboratorio de Fenómica y Embriogénesis Temprana (LAFET), Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
4
|
Trinidad L, Fletcher AG, Strutt D. The Fat-Dachsous planar polarity pathway competes with hinge contraction to orient polarized cell behaviors during Drosophila wing morphogenesis. Curr Biol 2025; 35:422-430.e3. [PMID: 39708794 PMCID: PMC7617321 DOI: 10.1016/j.cub.2024.11.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/02/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
During tissue morphogenesis, an interplay of biochemical pathways and mechanical cues regulates polarized cell behaviors, the balance of which leads to tissues reaching their correct shape and size.1,2,3,4 A well-studied example of a biochemical regulator is the highly conserved Fat-Dachsous (Ft-Ds) pathway that coordinates planar polarized cell behaviors and growth in epithelial tissues.5,6 For instance, in the Drosophila larval wing disc, the Ft-Ds pathway acts via the atypical myosin Dachs to control tissue shape by promoting the orientation of cell divisions primarily in a proximodistal (PD) direction.7,8 Here, we investigate interactions between Ft-Ds planar polarity and mechanical forces in the developing Drosophila pupal wing. We show that in the early stages of pupal wing development (16-18 h after puparium formation), anteroposterior (AP)-oriented cell divisions and T1 transitions are controlled by the Ft-Ds pathway acting via Dachs. Shortly thereafter, PD-oriented tissue tension is induced across the wing blade by the process of hinge contraction. This opposes the control of Dachs over polarized cell behaviors in a tug-of-war fashion, resulting in more PD-oriented cell divisions and T1s. Furthermore, increased PD tissue tension stabilizes Ft along PD-oriented junctions, suggesting that biomechanical feedback on the Ft-Ds pathway resists the effects of hinge contraction on cell shape. We also show that loss of Dachs results in increased myosin-II stability at cell junctions, revealing compensatory interactions between these two myosins. Overall, we propose that Ft-Ds pathway function constitutes a mechanism whereby tissues are buffered against mechanical perturbations.
Collapse
Affiliation(s)
- Larra Trinidad
- School of Biosciences, University of Sheffield, Firth Court, Sheffield, S10 2TN, UK
| | - Alexander G Fletcher
- School of School of Mathematical and Physical Sciences, University of Sheffield, Hicks Building, Sheffield S3 7RH, UK
| | - David Strutt
- School of Biosciences, University of Sheffield, Firth Court, Sheffield, S10 2TN, UK.
| |
Collapse
|
5
|
Matakatsu H, Fehon RG. Dachsous and Fat coordinately repress the Dachs-Dlish-Approximated complex to control growth. J Cell Biol 2024; 223:e202406119. [PMID: 39373700 PMCID: PMC11461286 DOI: 10.1083/jcb.202406119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/28/2024] [Accepted: 09/11/2024] [Indexed: 10/08/2024] Open
Abstract
Two protocadherins, Dachsous and Fat, regulate organ growth in Drosophila via the Hippo pathway. Dachsous and Fat bind heterotypically to regulate the abundance and subcellular localization of a "core complex" consisting of Dachs, Dlish, and Approximated. This complex localizes to the junctional cortex where it represses Warts. Dachsous is believed to promote growth by recruiting and stabilizing this complex, while Fat represses growth by promoting its degradation. Here, we examine the functional relationships between the intracellular domains of Dachsous and Fat and the core complex. While Dachsous promotes the accumulation of core complex proteins in puncta, it is not required for their assembly. Indeed, the core complex accumulates maximally in the absence of both Dachsous and Fat. Furthermore, Dachsous represses growth in the absence of Fat by removing the core complex from the junctional cortex. Fat similarly recruits core complex components but promotes their degradation. Our findings reveal that Dachsous and Fat coordinately constrain tissue growth by repressing the core complex.
Collapse
Affiliation(s)
- Hitoshi Matakatsu
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Richard G. Fehon
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
6
|
Tripathi BK, Irvine KD. Contributions of the Dachsous intracellular domain to Dachsous-Fat signaling. Development 2024; 151:dev202919. [PMID: 39503213 PMCID: PMC11634027 DOI: 10.1242/dev.202919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
The protocadherins Fat and Dachsous regulate organ growth, shape, patterning, and planar cell polarity. Although Dachsous and Fat have been described as ligand and receptor, respectively, in a signal transduction pathway, there is also evidence for bidirectional signaling. Here, we assess signaling downstream of Dachsous through analysis of its intracellular domain. Genomic deletions of conserved sequences within dachsous identified regions of the intracellular domain that contribute to Dachsous activity. Deletion of the A motif increased Dachsous protein levels and decreased wing size. Deletion of the D motif decreased Dachsous levels at cell membranes, increased wing size, and disrupted wing, leg and hindgut patterning and planar cell polarity. Co-immunoprecipitation experiments established that the D motif is necessary and sufficient for association of Dachsous with key partners, including Lowfat, Dachs, Spiny-legs, Fat and MyoID. Subdivision of the D motif identified distinct regions that preferentially contribute to different Dachsous activities. Our results identify motifs that are essential for Dachsous function and are consistent with the hypothesis that the key function of Dachsous is regulation of Fat.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D. Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
7
|
Strutt H, Meshram D, Manning E, Madathil ACK, Strutt D. Fat-Dachsous planar polarity function requires two distinct heterophilic cadherin-cadherin binding interactions. Cell Rep 2024; 43:114722. [PMID: 39302834 PMCID: PMC11497213 DOI: 10.1016/j.celrep.2024.114722] [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: 04/17/2024] [Revised: 07/24/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Fat and Dachsous are evolutionarily conserved atypical cadherins that regulate polarized cell behaviors. In the Drosophila wing, they interact heterophilically between neighboring cells, localize asymmetrically to opposite cell ends, and control wing shape by regulating oriented cell rearrangements and divisions. Fat and Dachsous have 34 and 27 cadherin repeats, respectively, and previous work has identified trans interactions between their first four cadherin repeats. Here, we identify a second heterophilic binding site in their C-terminal cadherin repeats and show the conservation of this binding site in human Fat4 and Dachsous1. We provide evidence that both N- and C-terminal binding sites regulate the stability of Fat-Dachsous binding interactions and show that the N-terminal binding sites are partly dispensable for Fat-Dachsous function in vivo. Finally, we provide in vivo confirmation that the N-terminal repeats interact in an anti-parallel manner. We propose that multiple binding sites promote the clustering of Fat and Dachsous into a lattice-like array.
Collapse
Affiliation(s)
- Helen Strutt
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK.
| | - Dipak Meshram
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Elizabeth Manning
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | | | - David Strutt
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
8
|
Matakatsu H, Fehon RG. Dachsous and Fat coordinately repress the Dachs-Dlish-Approximated complex to control growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599638. [PMID: 38948705 PMCID: PMC11212998 DOI: 10.1101/2024.06.18.599638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Two protocadherins, Dachsous (Ds) and Fat (Ft), regulate organ growth in Drosophila via the Hippo pathway. Ds and Ft bind heterotypically to regulate the abundance and subcellular localization of a 'core complex' consisting of Dachs, Dlish and Approximated. This complex localizes to the junctional cortex where it promotes growth by repressing the pathway kinase Warts. Ds is believed to promote growth by recruiting and stabilizing the core complex at the junctional cortex, while Ft represses growth by promoting degradation of core complex components. Here, we examine the functions of intracellular domains of Ds and Ft and their relationship to the core complex. While Ds promotes accumulation of the core complex proteins in cortical puncta, it is not required for core complex assembly. Indeed, the core complex assembles maximally in the absence of both Ds and Ft. Furthermore, while Ds promotes growth in the presence of Ft, it represses growth in the absence of Ft by removing the core complex from the junctional cortex. Ft similarly recruits core complex components, however it normally promotes their degradation. Our findings reveal that Ds and Ft constrain tissue growth by repressing the default 'on' state of the core complex.
Collapse
|
9
|
Liu A, O’Connell J, Wall F, Carthew RW. Scaling between cell cycle duration and wing growth is regulated by Fat-Dachsous signaling in Drosophila. eLife 2024; 12:RP91572. [PMID: 38842917 PMCID: PMC11156469 DOI: 10.7554/elife.91572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024] Open
Abstract
The atypical cadherins Fat and Dachsous (Ds) signal through the Hippo pathway to regulate growth of numerous organs, including the Drosophila wing. Here, we find that Ds-Fat signaling tunes a unique feature of cell proliferation found to control the rate of wing growth during the third instar larval phase. The duration of the cell cycle increases in direct proportion to the size of the wing, leading to linear-like growth during the third instar. Ds-Fat signaling enhances the rate at which the cell cycle lengthens with wing size, thus diminishing the rate of wing growth. We show that this results in a complex but stereotyped relative scaling of wing growth with body growth in Drosophila. Finally, we examine the dynamics of Fat and Ds protein distribution in the wing, observing graded distributions that change during growth. However, the significance of these dynamics is unclear since perturbations in expression have negligible impact on wing growth.
Collapse
Affiliation(s)
- Andrew Liu
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
- NSF-Simons National Institute for Theory and Mathematics in BiologyChicagoUnited States
| | - Jessica O’Connell
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Farley Wall
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
| | - Richard W Carthew
- Department of Molecular Biosciences, Northwestern UniversityEvanstonUnited States
- NSF-Simons Center for Quantitative Biology, Northwestern UniversityEvanstonUnited States
- NSF-Simons National Institute for Theory and Mathematics in BiologyChicagoUnited States
| |
Collapse
|
10
|
Bu T, Wang L, Wu X, Gao S, Li X, Yun D, Yang X, Li L, Cheng CY, Sun F. The Planar Cell Polarity Protein Fat1 in Sertoli Cell Function. Endocrinology 2024; 165:bqae041. [PMID: 38553880 DOI: 10.1210/endocr/bqae041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Indexed: 04/30/2024]
Abstract
Fat (FAT atypical cadherin) and Dchs (Dachsous cadherin-related protein) in adjacent Sertoli:Sertoli, Sertoli:spermatid, and spermatid:spermatid interfaces create an important intercellular bridge whose adhesive function is in turn supported by Fjx1, a nonreceptor Ser/Thr protein kinase. This concept is derived from earlier studies of Drosophila, which has been confirmed in this and earlier reports as well. Herein, we use the approach of knockdown of Fat1 by RNAi using primary cultures of Sertoli cells that mimicked the blood-testis barrier (BTB) in vivo, and a series of coherent experiments including functional assays to monitor the Sertoli cell tight junction (TJ) permeability barrier and a functional in vitro TJ integrity assay to assess the role of Fat1 in the testis. It was shown that planar cell polarity (PCP) protein Fat1 affected Sertoli cell function through its modulation of actin and microtubule cytoskeletal function, altering their polymerization activity through the Fat1/Fjx1 complex. Furthermore, Fat1 is intimately associated with β-catenin and α-N-catenin, as well as with Prickle 1 of the Vangl1/Prickle 1 complex, another PCP core protein to support intercellular interactions to confer PCP. In summary, these findings support the notion that the Fat:Dchs and the Vangl2:Fzd PCP intercellular bridges are tightly associated with basal ES/TJ structural proteins to stabilize PCP function at the Sertoli:Sertoli, Sertoli:spermatid, and spermatid:spermatid interface to sustain spermatogenesis.
Collapse
Affiliation(s)
- Tiao Bu
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Lingling Wang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China
| | - Sheng Gao
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xinyao Li
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Damin Yun
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiwen Yang
- School of Basic Medical Science, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan 750004, China
| | - Linxi Li
- Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Chuen Yan Cheng
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Fei Sun
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
11
|
Liu A, O’Connell J, Wall F, Carthew RW. Scaling between cell cycle duration and wing growth is regulated by Fat-Dachsous signaling in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.01.551465. [PMID: 38645118 PMCID: PMC11030236 DOI: 10.1101/2023.08.01.551465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The atypical cadherins Fat and Dachsous (Ds) signal through the Hippo pathway to regulate growth of numerous organs, including the Drosophila wing. Here, we find that Ds-Fat signaling tunes a unique feature of cell proliferation found to control the rate of wing growth during the third instar larval phase. The duration of the cell cycle increases in direct proportion to the size of the wing, leading to linear-like growth during the third instar. Ds-Fat signaling enhances the rate at which the cell cycle lengthens with wing size, thus diminishing the rate of wing growth. We show that this results in a complex but stereotyped relative scaling of wing growth with body growth in Drosophila. Finally, we examine the dynamics of Fat and Ds protein distribution in the wing, observing graded distributions that change during growth. However, the significance of these dynamics is unclear since perturbations in expression have negligible impact on wing growth.
Collapse
Affiliation(s)
- Andrew Liu
- Department of Molecular Biosciences, Northwestern University, Evanston IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston IL
| | - Jessica O’Connell
- Department of Molecular Biosciences, Northwestern University, Evanston IL
| | - Farley Wall
- Department of Molecular Biosciences, Northwestern University, Evanston IL
| | - Richard W. Carthew
- Department of Molecular Biosciences, Northwestern University, Evanston IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston IL
| |
Collapse
|
12
|
Tripathi BK, Irvine KD. Contributions of the Dachsous intracellular domain to Dachsous-Fat signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587940. [PMID: 38617303 PMCID: PMC11014530 DOI: 10.1101/2024.04.03.587940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The protocadherins Fat and Dachsous regulate organ growth, shape, patterning, and planar cell polarity. Although Dachsous and Fat have been described as ligand and receptor, respectively, in a signal transduction pathway, there is also evidence for bidirectional signaling. Here we assess signaling downstream of Dachsous through analysis of its intracellular domain. Genomic deletions of conserved sequences within dachsous identified regions of the intracellular domain required for normal development. Deletion of the A motif increased Dachsous protein levels and decreased wing size. Deletion of the D motif decreased Dachsous levels at cell membranes, increased wing size, and disrupted wing, leg and hindgut patterning and planar cell polarity. Co-immunoprecipitation experiments established that the D motif is necessary and sufficient for association of Dachsous with four key partners: Lowfat, Dachs, Spiny-legs, and MyoID. Subdivision of the D motif identified distinct regions that are preferentially responsible for association with Lft versus Dachs. Our results identify motifs that are essential for Dachsous function and are consistent with the hypothesis that the key function of Dachsous is regulation of Fat.
Collapse
|
13
|
Shinwari K, Rehman HM, Xiao N, Guojun L, Khan MA, Bolkov MA, Tuzankina IA, Chereshnev VA. Novel high-risk missense mutations identification in FAT4 gene causing Hennekam syndrome and Van Maldergem syndrome 2 through molecular dynamics simulation. INFORMATICS IN MEDICINE UNLOCKED 2023. [DOI: 10.1016/j.imu.2023.101160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
14
|
Kasiah J, McNeill H. Fat and Dachsous cadherins in mammalian development. Curr Top Dev Biol 2023; 154:223-244. [PMID: 37100519 DOI: 10.1016/bs.ctdb.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Cell growth and patterning are critical for tissue development. Here we discuss the evolutionarily conserved cadherins, Fat and Dachsous, and the roles they play during mammalian tissue development and disease. In Drosophila, Fat and Dachsous regulate tissue growth via the Hippo pathway and planar cell polarity (PCP). The Drosophila wing has been an ideal tissue to observe how mutations in these cadherins affect tissue development. In mammals, there are multiple Fat and Dachsous cadherins, which are expressed in many tissues, but mutations in these cadherins that affect growth and tissue organization are context dependent. Here we examine how mutations in the Fat and Dachsous mammalian genes affect development in mammals and contribute to human disease.
Collapse
Affiliation(s)
- Jennysue Kasiah
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States.
| |
Collapse
|
15
|
Brittle A, Warrington SJ, Strutt H, Manning E, Tan SE, Strutt D. Distinct mechanisms of planar polarization by the core and Fat-Dachsous planar polarity pathways in the Drosophila wing. Cell Rep 2022; 40:111419. [PMID: 36170824 PMCID: PMC9631118 DOI: 10.1016/j.celrep.2022.111419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Planar polarity describes the coordinated polarization of cells within a tissue plane, and in animals can be determined by the “core” or Fat-Dachsous pathways. Current models for planar polarity establishment involve two components: tissue-level “global” cues that determine the overall axis of polarity and cell-level feedback-mediated cellular polarity amplification. Here, we investigate the contributions of global cues versus cellular feedback amplification in the core and Fat-Dachsous pathways during Drosophila pupal wing development. We present evidence that these pathways generate planar polarity via distinct mechanisms. Core pathway function is consistent with strong feedback capable of self-organizing cell polarity, which can then be aligned with the tissue axis via weak or transient global cues. Conversely, generation of cell polarity by the Ft-Ds pathway depends on strong global cues in the form of graded patterns of gene expression, which can then be amplified by weak feedback mechanisms.
The core and Fat-Dachsous planar polarity pathways function via distinct mechanisms The core can self-organize planar polarity and be oriented by weak upstream cues Fat-Dachsous are oriented by strong gradient cues but show poor self-organization
Collapse
Affiliation(s)
- Amy Brittle
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | | | - Helen Strutt
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Elizabeth Manning
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Su Ee Tan
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - David Strutt
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| |
Collapse
|
16
|
Ayukawa T, Akiyama M, Hozumi Y, Ishimoto K, Sasaki J, Senoo H, Sasaki T, Yamazaki M. Tissue flow regulates planar cell polarity independently of the Frizzled core pathway. Cell Rep 2022; 40:111388. [PMID: 36130497 DOI: 10.1016/j.celrep.2022.111388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 07/16/2022] [Accepted: 08/29/2022] [Indexed: 11/27/2022] Open
Abstract
Planar cell polarity (PCP) regulates the orientation of external structures. A core group of proteins that includes Frizzled forms the heart of the PCP regulatory system. Other PCP mechanisms that are independent of the core group likely exist, but their underlying mechanisms are elusive. Here, we show that tissue flow is a mechanism governing core group-independent PCP on the Drosophila notum. Loss of core group function only slightly affects bristle orientation in the adult central notum. This near-normal PCP results from tissue flow-mediated rescue of random bristle orientation during the pupal stage. Manipulation studies suggest that tissue flow can orient bristles in the opposite direction to the flow. This process is independent of the core group and implies that the apical extracellular matrix functions like a "comb" to align bristles. Our results reveal the significance of cooperation between tissue dynamics and extracellular substances in PCP establishment.
Collapse
Affiliation(s)
- Tomonori Ayukawa
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Masakazu Akiyama
- Meiji Institute for Advanced Study of Mathematical Sciences, Meiji University, Tokyo 164-8525, Japan; Faculty of Science, Academic Assembly, University of Toyama, Toyama 930-8555, Japan
| | - Yasukazu Hozumi
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Kenta Ishimoto
- Research Institute for Mathematical Sciences, Kyoto University, Kyoto 606-8502, Japan
| | - Junko Sasaki
- Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Haruki Senoo
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Takehiko Sasaki
- Department of Biochemical Pathophysiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Masakazu Yamazaki
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Akita 010-8543, Japan; Japan Science and Technology Agency, PRESTO, Saitama 332-0012, Japan.
| |
Collapse
|
17
|
Camuglia J, Chanet S, Martin AC. Morphogenetic forces planar polarize LGN/Pins in the embryonic head during Drosophila gastrulation. eLife 2022; 11:e78779. [PMID: 35796436 PMCID: PMC9262390 DOI: 10.7554/elife.78779] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/05/2022] [Indexed: 01/03/2023] Open
Abstract
Spindle orientation is often achieved by a complex of Partner of Inscuteable (Pins)/LGN, Mushroom Body Defect (Mud)/Nuclear Mitotic Apparatus (NuMa), Gαi, and Dynein, which interacts with astral microtubules to rotate the spindle. Cortical Pins/LGN recruitment serves as a critical step in this process. Here, we identify Pins-mediated planar cell polarized divisions in several of the mitotic domains of the early Drosophila embryo. We found that neither planar cell polarity pathways nor planar polarized myosin localization determined division orientation; instead, our findings strongly suggest that Pins planar polarity and force generated from mesoderm invagination are important. Disrupting Pins polarity via overexpression of a myristoylated version of Pins caused randomized division angles. We found that disrupting forces through chemical inhibitors, depletion of an adherens junction protein, or blocking mesoderm invagination disrupted Pins planar polarity and spindle orientation. Furthermore, directional ablations that separated mesoderm from mitotic domains disrupted spindle orientation, suggesting that forces transmitted from mesoderm to mitotic domains can polarize Pins and orient division during gastrulation. To our knowledge, this is the first in vivo example where mechanical force has been shown to polarize Pins to mediate division orientation.
Collapse
Affiliation(s)
- Jaclyn Camuglia
- Biology Department, Massachusetts Institute of TechnologyCambridge, MAUnited States
| | - Soline Chanet
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSLParisFrance
| | - Adam C Martin
- Biology Department, Massachusetts Institute of TechnologyCambridge, MAUnited States
| |
Collapse
|
18
|
Gridnev A, Misra JR. Emerging Mechanisms of Growth and Patterning Regulation by Dachsous and Fat Protocadherins. Front Cell Dev Biol 2022; 10:842593. [PMID: 35372364 PMCID: PMC8967653 DOI: 10.3389/fcell.2022.842593] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/08/2022] [Indexed: 01/14/2023] Open
Abstract
Dachsous (Ds) and Fat are evolutionarily conserved cell adhesion molecules that play a critical role in development of multiple organ systems, where they coordinate tissue growth and morphogenesis. Much of our understanding of Ds-Fat signaling pathway comes from studies in Drosophila, where they initiate a signaling pathway that regulate growth by influencing Hippo signaling and morphogenesis by regulating Planar Cell Polarity (PCP). In this review, we discuss recent advances in our understanding of the mechanisms by which Ds-Fat signaling pathway regulates these critical developmental processes. Further, we discuss the progress in our understanding about how they function in mammals.
Collapse
|
19
|
Abstract
The Drosophila wing imaginal disc is a tissue of undifferentiated cells that are precursors of the wing and most of the notum of the adult fly. The wing disc first forms during embryogenesis from a cluster of ∼30 cells located in the second thoracic segment, which invaginate to form a sac-like structure. They undergo extensive proliferation during larval stages to form a mature larval wing disc of ∼35,000 cells. During this time, distinct cell fates are assigned to different regions, and the wing disc develops a complex morphology. Finally, during pupal stages the wing disc undergoes morphogenetic processes and then differentiates to form the adult wing and notum. While the bulk of the wing disc comprises epithelial cells, it also includes neurons and glia, and is associated with tracheal cells and muscle precursor cells. The relative simplicity and accessibility of the wing disc, combined with the wealth of genetic tools available in Drosophila, have combined to make it a premier system for identifying genes and deciphering systems that play crucial roles in animal development. Studies in wing imaginal discs have made key contributions to many areas of biology, including tissue patterning, signal transduction, growth control, regeneration, planar cell polarity, morphogenesis, and tissue mechanics.
Collapse
Affiliation(s)
- Bipin Kumar Tripathi
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Waksman Institute, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
20
|
Dong M, Chen D, Che L, Gu N, Yin M, Du X, Shen J, Yan S. Biotoxicity Evaluation of a Cationic Star Polymer on a Predatory Ladybird and Cooperative Pest Control by Polymer-Delivered Pesticides and Ladybird. ACS APPLIED MATERIALS & INTERFACES 2022; 14:6083-6092. [PMID: 35072467 DOI: 10.1021/acsami.1c24077] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although employing nanocarriers for gene/drug delivery shows great potential in agricultural fields, the biotoxicity of nanocarriers is a major concern for large-scale applications. Herein, we synthesized a cationic star polymer (SPc) as a pesticide nanocarrier/adjuvant to evaluate its safety against a widely used predatory ladybird (Harmonia axyridis). The application of SPc at extremely high concentrations nearly did not influence the hatching of ladybird eggs but it led to the death of ladybird larvae at lethal concentration 50 (LC50) values of 43.96 and 19.85 mg/mL through the soaking and feeding methods, respectively. The oral feeding of SPc downregulated many membrane protein genes and lysosome genes significantly, and the cell membrane and nucleus in gut tissues were remarkably damaged by SPc application, revealing that the lethal mechanism might be SPc-mediated membrane damage. Furthermore, the oral feeding of SPc increased the relative abundance of Serratia bacteria in ladybird guts to result in bacterial infection. Coapplication of ladybird and SPc-loaded thiamethoxam/matrine achieved desired control efficacies of more than 80% against green peach aphids, revealing that the coapplication could overcome the slow-acting property of ladybirds. To our knowledge, this is the first attempt to investigate the polymer-mediated lethal mechanism toward natural enemies and explore the possibility of coapplying SPc-loaded pesticides and natural enemies for pest management.
Collapse
Affiliation(s)
- Min Dong
- Department of Plant Biosecurity and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, P. R. China
| | - Dingming Chen
- Department of Plant Biosecurity and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, P. R. China
| | - Lin Che
- Department of Plant Biosecurity and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, P. R. China
| | - Na Gu
- Department of Plant Biosecurity and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, P. R. China
| | - Meizhen Yin
- State Key Lab of Chemical Resource Engineering, Beijing Lab of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Xiangge Du
- Department of Plant Biosecurity and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, P. R. China
| | - Jie Shen
- Department of Plant Biosecurity and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, P. R. China
| | - Shuo Yan
- Department of Plant Biosecurity and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
21
|
Cheng P, Liao HY, Zhang HH. The role of Wnt/mTOR signaling in spinal cord injury. J Clin Orthop Trauma 2022; 25:101760. [PMID: 35070684 PMCID: PMC8762069 DOI: 10.1016/j.jcot.2022.101760] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 01/07/2023] Open
Abstract
Spinal cord injury (SCI) is the most common disabling spinal injury, a complex pathologic process that can eventually lead to severe neurological dysfunction. The Wnt/mTOR signaling pathway is a pervasive signaling cascade that regulates a wide range of physiological processes during embryonic development, from stem cell pluripotency to cell fate. Numerous studies have reported that Wnt/mTOR signaling pathway plays an important role in neural development, synaptogenesis, neuron growth, differentiation and survival after the central nervous system (CNS) is damaged. Wnt/mTOR also plays an important role in regulating various pathophysiological processes after spinal cord injury (SCI). After SCI, Wnt/mTOR signal regulates the physiological and pathological processes of neural stem cell proliferation and differentiation, neuronal axon regeneration, neuroinflammation and pain through multiple pathways. Due to the characteristics of the Wnt signal in SCI make it a potential therapeutic target of SCI. In this paper, the characteristics of Wnt/mTOR signal, the role of Wnt/mTOR pathway on SCI and related mechanisms are reviewed, and some unsolved problems are discussed. It is hoped to provide reference value for the research field of the role of Wnt/mTOR pathway in SCI, and provide a theoretical basis for biological therapy of SCI.
Collapse
Affiliation(s)
- Peng Cheng
- Department of Spine Surgery, LanZhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, PR China
| | - Hai-Yang Liao
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, 342800, PR China
| | - Hai-Hong Zhang
- Department of Spine Surgery, LanZhou University Second Hospital, 82 Cuiying Men, Lanzhou, 730000, PR China
| |
Collapse
|
22
|
Ledru M, Clark CA, Brown J, Verghese S, Ferrara S, Goodspeed A, Su TT. Differential gene expression analysis identified determinants of cell fate plasticity during radiation-induced regeneration in Drosophila. PLoS Genet 2022; 18:e1009989. [PMID: 34990447 PMCID: PMC8769364 DOI: 10.1371/journal.pgen.1009989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/19/2022] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Ionizing radiation (IR) is used to treat half of all cancer patients because of its ability to kill cells. IR, however, can induce stem cell-like properties in non-stem cancer cells, potentiating tumor regrowth and reduced therapeutic success. We identified previously a subpopulation of cells in Drosophila larval wing discs that exhibit IR-induced stem cell-like properties. These cells reside in the future wing hinge, are resistant to IR-induced apoptosis, and are capable of translocating, changing fate, and participating in regenerating the pouch that suffers more IR-induced apoptosis. We used here a combination of lineage tracing, FACS-sorting of cells that change fate, genome-wide RNAseq, and functional testing of 42 genes, to identify two key changes that are required cell-autonomously for IR-induced hinge-to-pouch fate change: (1) repression of hinge determinants Wg (Drosophila Wnt1) and conserved zinc-finger transcription factor Zfh2 and (2) upregulation of three ribosome biogenesis factors. Additional data indicate a role for Myc, a transcriptional activator of ribosome biogenesis genes, in the process. These results provide a molecular understanding of IR-induced cell fate plasticity that may be leveraged to improve radiation therapy. Ionizing radiation (IR) is used to treat half of all cancer patients because of its ability to kill cells but treatment failures are common because tumors grow back (regenerate). Here, we asked which changes in the properties of cells facilitate regeneration in Drosophila (fruit flies) after exposure to radiation. We identified six genes whose products increase or decrease the regenerative potential of cells. These results help us understand how tissues regenerate after IR damage and will aid in designing better therapies that involve radiation.
Collapse
Affiliation(s)
- Michelle Ledru
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Caitlin A. Clark
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Jeremy Brown
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Shilpi Verghese
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
| | - Sarah Ferrara
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Andrew Goodspeed
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Tin Tin Su
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, United States of America
- University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
23
|
Agrawal N, Lawler K, Davidson CM, Keogh JM, Legg R, INTERVAL, Barroso I, Farooqi IS, Brand AH. Predicting novel candidate human obesity genes and their site of action by systematic functional screening in Drosophila. PLoS Biol 2021; 19:e3001255. [PMID: 34748544 PMCID: PMC8575313 DOI: 10.1371/journal.pbio.3001255] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
The discovery of human obesity-associated genes can reveal new mechanisms to target for weight loss therapy. Genetic studies of obese individuals and the analysis of rare genetic variants can identify novel obesity-associated genes. However, establishing a functional relationship between these candidate genes and adiposity remains a significant challenge. We uncovered a large number of rare homozygous gene variants by exome sequencing of severely obese children, including those from consanguineous families. By assessing the function of these genes in vivo in Drosophila, we identified 4 genes, not previously linked to human obesity, that regulate adiposity (itpr, dachsous, calpA, and sdk). Dachsous is a transmembrane protein upstream of the Hippo signalling pathway. We found that 3 further members of the Hippo pathway, fat, four-jointed, and hippo, also regulate adiposity and that they act in neurons, rather than in adipose tissue (fat body). Screening Hippo pathway genes in larger human cohorts revealed rare variants in TAOK2 associated with human obesity. Knockdown of Drosophila tao increased adiposity in vivo demonstrating the strength of our approach in predicting novel human obesity genes and signalling pathways and their site of action.
Collapse
Affiliation(s)
- Neha Agrawal
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Catherine M. Davidson
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Julia M. Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Robert Legg
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | | | - Inês Barroso
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - I. Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Andrea H. Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
24
|
Huang Y, Li D, Qiao L, Liu Y, Peng Q, Wu S, Zhang M, Yang Y, Tan J, Xu S, Jin L, Wang S, Tang K, Grünewald S. A genome-wide association study of facial morphology identifies novel genetic loci in Han Chinese. J Genet Genomics 2021; 48:198-207. [PMID: 33593615 DOI: 10.1016/j.jgg.2020.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 10/23/2022]
Abstract
The human face is a heritable surface with many complex sensory organs. In recent years, many genetic loci associated with facial features have been reported in different populations, yet there is a lack of studies on the Han Chinese population. Here, we report a genome-wide association study of 3D normal human faces of 2,659 Han Chinese with autosegment phenotypes of facial morphology. We identify single-nucleotide polymorphisms (SNPs) encompassing four genomic regions showing significant associations with different facial regions, including SNPs in DENND1B associated with the chin, SNPs among PISRT1 associated with eyes, SNPs between DCHS2 and SFRP2 associated with the nose, and SNPs in VPS13B associated with the nose. We replicate 24 SNPs from previously reported genetic loci in different populations, whose candidate genes are DCHS2, SUPT3H, HOXD1, SOX9, PAX3, and EDAR. These results provide a more comprehensive understanding of the genetic basis of variation in human facial morphology.
Collapse
Affiliation(s)
- Yin Huang
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China
| | - Dan Li
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China; DeepBlue Technology (Shanghai) Co., Ltd, Shanghai 200336, China
| | - Lu Qiao
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China
| | - Yu Liu
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China
| | - Qianqian Peng
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China
| | - Sijie Wu
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Manfei Zhang
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yajun Yang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan-Taizhou Institute of Health Sciences, Taizhou 225300, China
| | - Jingze Tan
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan-Taizhou Institute of Health Sciences, Taizhou 225300, China
| | - Shuhua Xu
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China; Collaborative Innovation Center of Genetics and Development, Shanghai 200438, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Li Jin
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China; State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan-Taizhou Institute of Health Sciences, Taizhou 225300, China; Collaborative Innovation Center of Genetics and Development, Shanghai 200438, China
| | - Sijia Wang
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China; Collaborative Innovation Center of Genetics and Development, Shanghai 200438, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Kun Tang
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China; DeepBlue Technology (Shanghai) Co., Ltd, Shanghai 200336, China.
| | - Stefan Grünewald
- Chinese Academy of Sciences Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, CAS, Shanghai 200031, China.
| |
Collapse
|
25
|
Strutt H, Strutt D. How do the Fat-Dachsous and core planar polarity pathways act together and independently to coordinate polarized cell behaviours? Open Biol 2021; 11:200356. [PMID: 33561385 PMCID: PMC8061702 DOI: 10.1098/rsob.200356] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Planar polarity describes the coordinated polarization of cells within the plane of a tissue. This is controlled by two main pathways in Drosophila: the Frizzled-dependent core planar polarity pathway and the Fat–Dachsous pathway. Components of both of these pathways become asymmetrically localized within cells in response to long-range upstream cues, and form intercellular complexes that link polarity between neighbouring cells. This review examines if and when the two pathways are coupled, focusing on the Drosophila wing, eye and abdomen. There is strong evidence that the pathways are molecularly coupled in tissues that express a specific isoform of the core protein Prickle, namely Spiny-legs. However, in other contexts, the linkages between the pathways are indirect. We discuss how the two pathways act together and independently to mediate a diverse range of effects on polarization of cell structures and behaviours.
Collapse
Affiliation(s)
- Helen Strutt
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - David Strutt
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
26
|
Wu ZL, Xie QQ, Liu TC, Yang X, Zhang GZ, Zhang HH. Role of the Wnt pathway in the formation, development, and degeneration of intervertebral discs. Pathol Res Pract 2021; 220:153366. [PMID: 33647863 DOI: 10.1016/j.prp.2021.153366] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/13/2022]
Abstract
Intervertebral disc degeneration (IVDD) is an age-related degenerative disease that is the main cause of low back pain. It seriously affects the quality of life of patients and places a heavy economic burden on families and society. The Wnt pathway plays an important role in the growth, development, and degeneration of intervertebral discs (IVDs). In the embryonic stage, the Wnt pathway participates in the growth and development of IVD by promoting the transformation of progenitor cells into notochord cells and the extension of the notochord. However, the activation of the Wnt pathway after birth promotes IVD cell senescence, apoptosis, and degradation of the extracellular matrix and induces the production of inflammatory factors, thereby accelerating the IVDD process. This article reviews the relationship between the Wnt pathway and IVD, emphasizing its influence on IVD growth, development, and degeneration. Targeting this pathway may become an effective strategy for the treatment of IVDD.
Collapse
Affiliation(s)
- Zuo-Long Wu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Qi-Qi Xie
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Tai-Cong Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Xing Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Guang-Zhi Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China.
| |
Collapse
|
27
|
DeAngelis MW, Coolon JD, Johnson RI. Comparative transcriptome analyses of the Drosophila pupal eye. G3-GENES GENOMES GENETICS 2021; 11:5995320. [PMID: 33561221 PMCID: PMC8043229 DOI: 10.1093/g3journal/jkaa003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/08/2020] [Indexed: 12/04/2022]
Abstract
Tissue function is dependent on correct cellular organization and behavior. As a result, the identification and study of genes that contribute to tissue morphogenesis is of paramount importance to the fields of cell and developmental biology. Many of the genes required for tissue patterning and organization are highly conserved between phyla. This has led to the emergence of several model organisms and developmental systems that are used to study tissue morphogenesis. One such model is the Drosophila melanogaster pupal eye that has a highly stereotyped arrangement of cells. In addition, the pupal eye is postmitotic that allows for the study of tissue morphogenesis independent from any effects of proliferation. While the changes in cell morphology and organization that occur throughout pupal eye development are well documented, less is known about the corresponding transcriptional changes that choreograph these processes. To identify these transcriptional changes, we dissected wild-type Canton S pupal eyes and performed RNA-sequencing. Our analyses identified differential expression of many loci that are documented regulators of pupal eye morphogenesis and contribute to multiple biological processes including signaling, axon projection, adhesion, and cell survival. We also identified differential expression of genes not previously implicated in pupal eye morphogenesis such as components of the Toll pathway, several non-classical cadherins, and components of the muscle sarcomere, which could suggest these loci function as novel patterning factors.
Collapse
Affiliation(s)
- Miles W DeAngelis
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Joseph D Coolon
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| | - Ruth I Johnson
- Department of Biology, Wesleyan University, 52 Lawn Avenue, Middletown, CT 06459, USA
| |
Collapse
|
28
|
Yu JJS, Maugarny-Calès A, Pelletier S, Alexandre C, Bellaiche Y, Vincent JP, McGough IJ. Frizzled-Dependent Planar Cell Polarity without Secreted Wnt Ligands. Dev Cell 2020; 54:583-592.e5. [PMID: 32888416 PMCID: PMC7497783 DOI: 10.1016/j.devcel.2020.08.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/07/2020] [Accepted: 08/10/2020] [Indexed: 12/26/2022]
Abstract
Planar cell polarity (PCP) organizes the orientation of cellular protrusions and migratory activity within the tissue plane. PCP establishment involves the subcellular polarization of core PCP components. It has been suggested that Wnt gradients could provide a global cue that coordinates local PCP with tissue axes. Here, we dissect the role of Wnt ligands in the orientation of hairs of Drosophila wings, an established system for the study of PCP. We found that PCP was normal in quintuple mutant wings that rely solely on the membrane-tethered Wingless for Wnt signaling, suggesting that a Wnt gradient is not required. We then used a nanobody-based approach to trap Wntless in the endoplasmic reticulum, and hence prevent all Wnt secretion, specifically during the period of PCP establishment. PCP was still established. We conclude that, even though Wnt ligands could contribute to PCP, they are not essential, and another global cue must exist for tissue-wide polarization.
Collapse
Affiliation(s)
| | - Aude Maugarny-Calès
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | - Stéphane Pelletier
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | | | - Yohanns Bellaiche
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, 75248 Paris Cedex 05, France; Sorbonne University, CNRS UMR 3215, INSERM U934, 75005 Paris, France
| | | | | |
Collapse
|
29
|
Abstract
Planar cell polarization, PCP, describes a form of organization where every cell within a group acquires the same planar characteristics, whether it is orientation of cell division, direction of migration, or localization of a cellular structure. PCP is essential for correct organization of cells into tissues and building a proper body plan. Here we use Hydra, an organism with a single axis of symmetry and a very simple body plan to investigate the function of the cell adhesion molecules Fat-like and Dachsous. We show that Hydra Fat-like and Dachsous are planar polarized, providing a demonstration of planar polarization of proteins in a nonbilaterian organism. We also discover roles for Hydra Fat-like in cell adhesion, spindle orientation, and tissue organization. Fat, Fat-like, and Dachsous family cadherins are giant proteins that regulate planar cell polarity (PCP) and cell adhesion in bilaterians. Their evolutionary origin can be traced back to prebilaterian species, but their ancestral function(s) are unknown. We identified Fat-like and Dachsous cadherins in Hydra, a member of phylum Cnidaria a sister group of bilaterian. We found Hydra does not possess a true Fat homolog, but has homologs of Fat-like (HyFatl) and Dachsous (HyDs) that localize at the apical membrane of ectodermal epithelial cells and are planar polarized perpendicular to the oral–aboral axis of the animal. Using a knockdown approach we found that HyFatl is involved in local cell alignment and cell–cell adhesion, and that reduction of HyFatl leads to defects in tissue organization in the body column. Overexpression and knockdown experiments indicate that the intracellular domain (ICD) of HyFatl affects actin organization through proline-rich repeats. Thus, planar polarization of Fat-like and Dachsous cadherins has ancient, prebilaterian origins, and Fat-like cadherins have ancient roles in cell adhesion, spindle orientation, and tissue organization.
Collapse
|
30
|
Role of Notch Signaling in Leg Development in Drosophila melanogaster. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:103-127. [PMID: 32060874 DOI: 10.1007/978-3-030-34436-8_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Notch pathway plays diverse and fundamental roles during animal development. One of the most relevant, which arises directly from its unique mode of activation, is the specification of cell fates and tissue boundaries. The development of the leg of Drosophila melanogaster is a fine example of this Notch function, as it is required to specify the fate of the cells that will eventually form the leg joints, the flexible structures that separate the different segments of the adult leg. Notch activity is accurately activated and maintained at the distal end of each segment in response to the proximo-distal patterning gene network of the developing leg. Region-specific downstream targets of Notch in turn regulate the formation of the different types of joints. We discuss recent findings that shed light on the molecular and cellular mechanisms that are ultimately governed by Notch to achieve epithelial fold and joint morphogenesis. Finally, we briefly summarize the role that Notch plays in inducing the nonautonomous growth of the leg. Overall, this book chapter aims to highlight leg development as a useful model to study how patterning information is translated into specific cell behaviors that shape the final form of an adult organ.
Collapse
|
31
|
Kumar A, Rizvi MS, Athilingam T, Parihar SS, Sinha P. Heterophilic cell-cell adhesion of atypical cadherins Fat and Dachsous regulate epithelial cell size dynamics during Drosophila thorax morphogenesis. Mol Biol Cell 2019; 31:546-560. [PMID: 31877063 PMCID: PMC7202070 DOI: 10.1091/mbc.e19-08-0468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Spatiotemporal changes in epithelial cell sizes-or epithelial cell size dynamics (ECD)-during morphogenesis entail interplay between two opposing forces: cell contraction via actomyosin cytoskeleton and cell expansion via cell-cell adhesion. Cell-cell adhesion-based ECD, however, has not yet been clearly demonstrated. For instance, changing levels of homophilic E-cadherin-based cell-cell adhesion induce cell sorting, but not ECD. Here we show that cell-expansive forces of heterophilic cell-cell adhesion regulate ECD: higher cell-cell adhesion results in cell size enlargement. Thus, ECD during morphogenesis in the heminotal epithelia of Drosophila pupae leading to thorax closure corresponds with spatiotemporal gradients of two heterophilic atypical cadherins-Fat (Ft) and Dachsous (Ds)-and the levels of Ft-Ds heterodimers formed concomitantly. Our mathematical modeling and genetic tests validate this mechanism of dynamic heterophilic cell-cell adhesion-based regulation of ECD. Conservation of these atypical cadherins suggests a wider prevalence of heterophilic cell-cell adhesion-based ECD regulation during animal morphogenesis.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Mohd Suhail Rizvi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Thamarailingam Athilingam
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Saurabh Singh Parihar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Pradip Sinha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
32
|
Mangione F, Martín-Blanco E. The Dachsous/Fat/Four-Jointed Pathway Directs the Uniform Axial Orientation of Epithelial Cells in the Drosophila Abdomen. Cell Rep 2019; 25:2836-2850.e4. [PMID: 30517870 DOI: 10.1016/j.celrep.2018.11.036] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 10/05/2018] [Accepted: 11/06/2018] [Indexed: 12/19/2022] Open
Abstract
The achievement of the final form of an individual requires not only the control of cell size and differentiation but also integrative directional cues to instruct cell movements, positions, and orientations. In Drosophila, the adult epidermis of the abdomen is created de novo by histoblasts. As these expand and fuse, they uniformly orient along the anteroposterior axis. We found that the Dachsous/Fat/Four-jointed (Ds/Ft/Fj) pathway is key for their alignment. The refinement of the tissue-wide expression of the atypical cadherins Ds and Ft result in their polarization and directional adhesiveness. Mechanistically, the axially oriented changes in histoblasts respond to the redesign of the epithelial field. We suggest that the role of Ds/Ft/Fj in long-range oriented cell alignment is a general function and that the regulation of the expression of its components will be crucial in other morphogenetic models or during tissue repair.
Collapse
Affiliation(s)
- Federica Mangione
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Científic de Barcelona, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Enrique Martín-Blanco
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Científic de Barcelona, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| |
Collapse
|
33
|
Arbouzova NI, Fulford AD, Zhang H, McNeill H. Fat regulates expression of four-jointed reporters in vivo through a 20 bp element independently of the Hippo pathway. Dev Biol 2019; 450:23-33. [DOI: 10.1016/j.ydbio.2019.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/07/2019] [Accepted: 03/07/2019] [Indexed: 01/15/2023]
|
34
|
Zhou Z, Alégot H, Irvine KD. Oriented Cell Divisions Are Not Required for Drosophila Wing Shape. Curr Biol 2019; 29:856-864.e3. [PMID: 30799243 DOI: 10.1016/j.cub.2019.01.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/21/2018] [Accepted: 01/15/2019] [Indexed: 12/19/2022]
Abstract
Formation of correctly shaped organs is vital for normal function. The Drosophila wing has an elongated shape, which has been attributed in part to a preferential orientation of mitotic spindles along the proximal-distal axis [1, 2]. Orientation of mitotic spindles is believed to be a fundamental morphogenetic mechanism in multicellular organisms [3-6]. A contribution of spindle orientation to wing shape was inferred from observations that mutation of Dachsous-Fat pathway genes results in both rounder wings and loss of the normal proximal-distal bias in spindle orientation [1, 2, 7]. To directly evaluate the potential contribution of spindle orientation to wing morphogenesis, we assessed the consequences of loss of the Drosophila NuMA homolog Mud, which interacts with the dynein complex and has a conserved role in spindle orientation [8, 9]. Loss of Mud randomizes spindle orientation but does not alter wing shape. Analysis of growth and cell dynamics in developing discs and in ex vivo culture suggests that the absence of oriented cell divisions is compensated for by an increased contribution of cell rearrangements to wing shape. Our results indicate that oriented cell divisions are not required for wing morphogenesis, nor are they required for the morphogenesis of other Drosophila appendages. Moreover, our results suggest that normal organ shape is not achieved through locally specifying and then summing up individual cell behaviors, like oriented cell division. Instead, wing shape might be specified through tissue-wide stresses that dictate an overall arrangement of cells without specifying the individual cell behaviors needed to achieve it.
Collapse
Affiliation(s)
- Zhenru Zhou
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Herve Alégot
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
35
|
Fisher KH, Strutt D. A theoretical framework for planar polarity establishment through interpretation of graded cues by molecular bridges. Development 2019; 146:146/3/dev168955. [PMID: 30709912 PMCID: PMC6382004 DOI: 10.1242/dev.168955] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Planar polarity is a widespread phenomenon found in many tissues, allowing cells to coordinate morphogenetic movements and function. A common feature of animal planar polarity systems is the formation of molecular bridges between cells, which become polarised along a tissue axis. We propose that these bridges provide a general mechanism by which cells interpret different forms of tissue gradients to coordinate directional information. We illustrate this using a generalised and consistent modelling framework, providing a conceptual basis for understanding how different mechanisms of gradient function can generate planar polarity. We make testable predictions of how different gradient mechanisms can influence polarity direction. Summary: This Hypothesis uses a theoretical framework to explore how molecular bridges provide a general mechanism to interpret different forms of tissue gradients to establish planar polarity.
Collapse
Affiliation(s)
- Katherine H Fisher
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - David Strutt
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
36
|
Early girl is a novel component of the Fat signaling pathway. PLoS Genet 2019; 15:e1007955. [PMID: 30699121 PMCID: PMC6370246 DOI: 10.1371/journal.pgen.1007955] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2019] [Accepted: 01/11/2019] [Indexed: 01/05/2023] Open
Abstract
The Drosophila protocadherins Dachsous and Fat regulate growth and tissue polarity by modulating the levels, membrane localization and polarity of the atypical myosin Dachs. Localization to the apical junctional membrane is critical for Dachs function, and the adapter protein Vamana/Dlish and palmitoyl transferase Approximated are required for Dachs membrane localization. However, how Dachs levels are regulated is poorly understood. Here we identify the early girl gene as playing an essential role in Fat signaling by limiting the levels of Dachs protein. early girl mutants display overgrowth of the wings and reduced cross vein spacing, hallmark features of mutations affecting Fat signaling. Genetic experiments reveal that it functions in parallel with Fat to regulate Dachs. early girl encodes an E3 ubiquitin ligase, physically interacts with Dachs, and regulates its protein stability. Concomitant loss of early girl and approximated results in accumulation of Dachs and Vamana in cytoplasmic punctae, suggesting that it also regulates their trafficking to the apical membrane. Our findings establish a crucial role for early girl in Fat signaling, involving regulation of Dachs and Vamana, two key downstream effectors of this pathway. During development, organs grow to achieve a consistent final size. The evolutionarily conserved Hippo signaling network plays a central role in organ size control, and when dysregulated can be associated with cancer and other diseases. Fat signaling is one of several upstream pathways that impinge on Hippo signaling to regulate organ growth. We describe here identification of the Drosophila early girl gene as a new component of the Fat signaling pathway. We show that Early girl controls Fat signaling by regulating the levels of the Dachs protein. However Early girl differs from other Fat signaling regulators in that it doesn’t influence planar cell polarity or control the polarity of Dachs localization. early girl encodes a conserved protein that is predicted to influence protein stability, and it can physically associate with Dachs. We also discovered that Early girl acts together with another protein, called Approximated, to regulate the sub-cellular localization of Dachs and a Dachs-interacting protein called Vamana. Altogether, our observations establish Early girl as an essential component of Fat signaling that acts to regulate the levels and localization of Dachs and Vamana.
Collapse
|
37
|
Tiwari P, Mrigwani A, Kaur H, Kaila P, Kumar R, Guptasarma P. Structural-Mechanical and Biochemical Functions of Classical Cadherins at Cellular Junctions: A Review and Some Hypotheses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1112:107-138. [DOI: 10.1007/978-981-13-3065-0_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
38
|
Lawrence PA, Casal J. Planar cell polarity: two genetic systems use one mechanism to read gradients. Development 2018; 145:145/23/dev168229. [DOI: 10.1242/dev.168229] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
ABSTRACT
Our aim in this short Primer is to explain the principles of planar cell polarity (PCP) in animal development. The literature in this small field is complex and specialized, but we have extracted a simple and central story from it. We explain our hypothesis that polarity, initially cued by the direction of slope of a multicellular gradient, is interpreted at the cellular level so that each cell becomes molecularly polarised. The mechanism involves a comparison between a cell and its neighbours. To achieve this comparison there are (at least) two disparate and independent molecular systems, each depending on molecular bridges that span between neighbouring cells. Even though the two systems are made up of different molecules, we argue that both systems function in a logically equivalent way.
Collapse
Affiliation(s)
- Peter A. Lawrence
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - José Casal
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| |
Collapse
|
39
|
Chen J, Castelvecchi GD, Li-Villarreal N, Raught B, Krezel AM, McNeill H, Solnica-Krezel L. Atypical Cadherin Dachsous1b Interacts with Ttc28 and Aurora B to Control Microtubule Dynamics in Embryonic Cleavages. Dev Cell 2018; 45:376-391.e5. [PMID: 29738714 PMCID: PMC5983389 DOI: 10.1016/j.devcel.2018.04.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 02/22/2018] [Accepted: 04/09/2018] [Indexed: 01/15/2023]
Abstract
Atypical cadherin Dachsous (Dchs) is a conserved regulator of planar cell polarity, morphogenesis, and tissue growth during animal development. Dchs functions in part by regulating microtubules by unknown molecular mechanisms. Here we show that maternal zygotic (MZ) dchs1b zebrafish mutants exhibit cleavage furrow progression defects and impaired midzone microtubule assembly associated with decreased microtubule turnover. Mechanistically, Dchs1b interacts via a conserved motif in its intracellular domain with the tetratricopeptide motifs of Ttc28 and regulates its subcellular distribution. Excess Ttc28 impairs cleavages and decreases microtubule turnover, while ttc28 inactivation increases turnover. Moreover, ttc28 deficiency in dchs1b mutants suppresses the microtubule dynamics and midzone microtubule assembly defects. Dchs1b also binds to Aurora B, a known regulator of cleavages and microtubules. Embryonic cleavages in MZdchs1b mutants exhibit increased, and in MZttc28 mutants decreased, sensitivity to Aurora B inhibition. Thus, Dchs1b regulates microtubule dynamics and embryonic cleavages by interacting with Ttc28 and Aurora B.
Collapse
Affiliation(s)
- Jiakun Chen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gina D Castelvecchi
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nanbing Li-Villarreal
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Andrzej M Krezel
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Genetics, University of Toronto, Toronto, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
The Molecular and Genetic Characterization of Second Chromosome Balancers in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2018; 8:1161-1171. [PMID: 29420191 PMCID: PMC5873907 DOI: 10.1534/g3.118.200021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Balancer chromosomes are multiply inverted and rearranged chromosomes used in Drosophila melanogaster for many tasks, such as maintaining mutant alleles in stock and complex stock construction. Balancers were created before molecular characterization of their breakpoints was possible, so the precise locations of many of these breakpoints are unknown. Here, we report or confirm the positions of the 14 euchromatic breakpoints on the 2nd chromosome balancers SM1, SM5, CyO, and SM6a This total includes three breakpoints involved in a complex rearrangement on SM5 that is associated with the duplication of two genomic regions. Unbiased sequencing of several balancers allowed us to identify stocks with incorrectly identified balancers as well as single and double crossover events that had occurred between 2nd chromosome balancers and their homologs. The confirmed crossover events that we recovered were at least 2 Mb from the closest inversion breakpoint, consistent with observations from other balancer chromosomes. Balancer chromosomes differ from one another both by large tracts of sequence diversity generated by recombination and by small differences, such as single nucleotide polymorphisms (SNPs). Therefore, we also report loss-of-function mutations carried by these chromosomes and unique SNP and InDel polymorphisms present on only single balancers. These findings provide valuable information about the structure of commonly used 2nd chromosome balancers and extend recent work examining the structure of X and 3rd chromosome balancers. Finally, these observations provide new insights into how the sequences of individual balancers have diverged over time.
Collapse
|
41
|
Cusanovich DA, Reddington JP, Garfield DA, Daza RM, Aghamirzaie D, Marco-Ferreres R, Pliner HA, Christiansen L, Qiu X, Steemers FJ, Trapnell C, Shendure J, Furlong EEM. The cis-regulatory dynamics of embryonic development at single-cell resolution. Nature 2018. [PMID: 29539636 PMCID: PMC5866720 DOI: 10.1038/nature25981] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding how gene regulatory networks control the progressive restriction of cell fates is a long-standing challenge. Recent advances in measuring single cell gene expression are providing new insights into lineage commitment. However, the regulatory events underlying these changes remain elusive. Here we investigate the dynamics of chromatin regulatory landscapes during embryogenesis at single cell resolution. Using single cell combinatorial indexing assay for transposase accessible chromatin (sci-ATAC-seq)1, we profiled chromatin accessibility in over 20,000 single nuclei from fixed Drosophila embryos spanning three landmark embryonic stages: 2-4 hours (hrs) after egg laying (predominantly stage 5 blastoderm nuclei), when each embryo comprises ~6,000 multipotent cells; 6-8hrs (predominantly stage 10-11), to capture a midpoint in embryonic development when major lineages in the mesoderm and ectoderm are specified; and 10-12hrs (predominantly stage 13), when each of the embryo’s >20,000 cells are undergoing terminal differentiation. Our results reveal spatial heterogeneity in the usage of the regulatory genome prior to gastrulation, a feature that aligns with future cell fate, and nuclei can be temporally ordered along developmental trajectories. During mid-embryogenesis, tissue granularity emerges such that individual cell types can be inferred by their chromatin accessibility, while maintaining a signature of their germ layer of origin. The data reveal overlapping usage of regulatory elements between cells of the endoderm and non-myogenic mesoderm, suggesting a common developmental program reminiscent of the mesendoderm lineage in other species2–4. Altogether, we identify over 30,000 distal regulatory elements exhibiting tissue-specific accessibility. We validated the germ layer specificity of a subset of these predicted enhancers in transgenic embryos, achieving 90% accuracy. Overall, our results demonstrate the power of shotgun single cell profiling of embryos to resolve dynamic changes in the chromatin landscape during development, and to uncover the cis-regulatory programs of metazoan germ layers and cell types.
Collapse
Affiliation(s)
- Darren A Cusanovich
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - James P Reddington
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - David A Garfield
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Delasa Aghamirzaie
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Raquel Marco-Ferreres
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Hannah A Pliner
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Xiaojie Qiu
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA.,Howard Hughes Medical Institute, Seattle, Washington, USA
| | - Eileen E M Furlong
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| |
Collapse
|
42
|
Hust J, Lavine MD, Worthington AM, Zinna R, Gotoh H, Niimi T, Lavine L. The Fat-Dachsous signaling pathway regulates growth of horns in Trypoxylus dichotomus, but does not affect horn allometry. JOURNAL OF INSECT PHYSIOLOGY 2018; 105:85-94. [PMID: 29366850 DOI: 10.1016/j.jinsphys.2018.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 06/07/2023]
Abstract
Males of the Asian rhinoceros beetle, Trypoxylus dichotomus, possess exaggerated head and thoracic horns that scale dramatically out of proportion to body size. While studies of insulin signaling suggest that this pathway regulates nutrition-dependent growth including exaggerated horns, what regulates disproportionate growth has yet to be identified. The Fat signaling pathway is a potential candidate for regulating disproportionate growth of sexually-selected traits, a hypothesis we advanced in a previous paper (Gotoh et al., 2015). To investigate the role of Fat signaling in the growth and scaling of the sexually dimorphic, condition-dependent traits of the in the Asian rhinoceros beetle T. dichotomus, we used RNA interference to knock down expression of fat and its co-receptor dachsous. Knockdown of fat, and to a lesser degree dachsous, caused shortening and widening of appendages, including the head and thoracic horns. However, scaling of horns to body size was not affected. Our results show that Fat signaling regulates horn growth in T. dichotomus as it does in appendage growth in other insects. However, we provide evidence that Fat signaling does not mediate the disproportionate, positive allometric growth of horns in T. dichotomus.
Collapse
Affiliation(s)
- James Hust
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Mark D Lavine
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Amy M Worthington
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Robert Zinna
- Department of Entomology, Washington State University, Pullman, WA 99164, United States
| | - Hiroki Gotoh
- Department of Entomology, Washington State University, Pullman, WA 99164, United States; Lab of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya 464-8601, Japan
| | - T Niimi
- Lab of Sericulture and Entomoresources, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa, Nagoya 464-8601, Japan; Division of Evolutionary Developmental Biology, National Institute for Basic Biology, Okazaki, Aichi 444-8585, Japan
| | - Laura Lavine
- Department of Entomology, Washington State University, Pullman, WA 99164, United States.
| |
Collapse
|
43
|
Dye NA, Popović M, Spannl S, Etournay R, Kainmüller D, Ghosh S, Myers EW, Jülicher F, Eaton S. Cell dynamics underlying oriented growth of the Drosophila wing imaginal disc. Development 2017; 144:4406-4421. [PMID: 29038308 DOI: 10.1242/dev.155069] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022]
Abstract
Quantitative analysis of the dynamic cellular mechanisms shaping the Drosophila wing during its larval growth phase has been limited, impeding our ability to understand how morphogen patterns regulate tissue shape. Such analysis requires explants to be imaged under conditions that maintain both growth and patterning, as well as methods to quantify how much cellular behaviors change tissue shape. Here, we demonstrate a key requirement for the steroid hormone 20-hydroxyecdysone (20E) in the maintenance of numerous patterning systems in vivo and in explant culture. We find that low concentrations of 20E support prolonged proliferation in explanted wing discs in the absence of insulin, incidentally providing novel insight into the hormonal regulation of imaginal growth. We use 20E-containing media to observe growth directly and to apply recently developed methods for quantitatively decomposing tissue shape changes into cellular contributions. We discover that whereas cell divisions drive tissue expansion along one axis, their contribution to expansion along the orthogonal axis is cancelled by cell rearrangements and cell shape changes. This finding raises the possibility that anisotropic mechanical constraints contribute to growth orientation in the wing disc.
Collapse
Affiliation(s)
- Natalie A Dye
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Marko Popović
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187 Dresden, Germany
| | - Stephanie Spannl
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Raphaël Etournay
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany.,Unité de Génétique et Physiologie de l'Audition UMRS 1120, Département de Neurosciences, Institut Pasteur, 75015 Paris, France
| | - Dagmar Kainmüller
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany.,Janelia Farm Research Campus, 19700 Helix Dr, Ashburn, VA 20147, USA
| | - Suhrid Ghosh
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Eugene W Myers
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany.,Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Nöthnitzer Strasse 38, 01187 Dresden, Germany .,Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01309 Dresden, Germany
| | - Suzanne Eaton
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01309 Dresden, Germany .,Biotechnologisches Zentrum, Technische Universität Dresden, Tatzberg 47/49, 01309 Dresden, Germany
| |
Collapse
|
44
|
Difference in Dachsous Levels between Migrating Cells Coordinates the Direction of Collective Cell Migration. Dev Cell 2017; 42:479-497.e10. [DOI: 10.1016/j.devcel.2017.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/19/2017] [Accepted: 07/31/2017] [Indexed: 12/21/2022]
|
45
|
Ebnet K, Kummer D, Steinbacher T, Singh A, Nakayama M, Matis M. Regulation of cell polarity by cell adhesion receptors. Semin Cell Dev Biol 2017; 81:2-12. [PMID: 28739340 DOI: 10.1016/j.semcdb.2017.07.032] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/12/2017] [Accepted: 07/20/2017] [Indexed: 01/01/2023]
Abstract
The ability of cells to polarize is an intrinsic property of almost all cells and is required for the devlopment of most multicellular organisms. To develop cell polarity, cells integrate various signals derived from intrinsic as well as extrinsic sources. In the recent years, cell-cell adhesion receptors have turned out as important regulators of cellular polarization. By interacting with conserved cell polarity proteins, they regulate the recruitment of polarity complexes to specific sites of cell-cell adhesion. By initiating intracellular signaling cascades at those sites, they trigger their specific subcellular activation. Not surprisingly, cell-cell adhesion receptors regulate diverse aspects of cell polarity, including apico-basal polarity in epithelial and endothelial cells, front-to-rear polarity in collectively migrating cells, and planar cell polarity during organ development. Here, we review the recent developments highlighting the central roles of cell-cell adhesion molecules in the development of cell polarity.
Collapse
Affiliation(s)
- Klaus Ebnet
- Institute-associated Research Group: Cell adhesion and cell polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Germany; Interdisciplinary Clinical Research Center (IZKF), University of Münster, Germany; Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany.
| | - Daniel Kummer
- Institute-associated Research Group: Cell adhesion and cell polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Germany; Interdisciplinary Clinical Research Center (IZKF), University of Münster, Germany
| | - Tim Steinbacher
- Institute-associated Research Group: Cell adhesion and cell polarity, Institute of Medical Biochemistry, ZMBE, University of Münster, Germany; Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany
| | - Amrita Singh
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany; Institute of Cell Biology, ZMBE, University of Münster, Germany
| | - Masanori Nakayama
- Laboratory for Cell Polarity and Organogenesis, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Maja Matis
- Cells-In-Motion Cluster of Excellence (EXC1003-CiM), University of Münster, Germany; Institute of Cell Biology, ZMBE, University of Münster, Germany.
| |
Collapse
|
46
|
Wortman JC, Nahmad M, Zhang PC, Lander AD, Yu CC. Expanding signaling-molecule wavefront model of cell polarization in the Drosophila wing primordium. PLoS Comput Biol 2017; 13:e1005610. [PMID: 28671940 PMCID: PMC5515495 DOI: 10.1371/journal.pcbi.1005610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 07/18/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
In developing tissues, cell polarization and proliferation are regulated by morphogens and signaling pathways. Cells throughout the Drosophila wing primordium typically show subcellular localization of the unconventional myosin Dachs on the distal side of cells (nearest the center of the disc). Dachs localization depends on the spatial distribution of bonds between the protocadherins Fat (Ft) and Dachsous (Ds), which form heterodimers between adjacent cells; and the Golgi kinase Four-jointed (Fj), which affects the binding affinities of Ft and Ds. The Fj concentration forms a linear gradient while the Ds concentration is roughly uniform throughout most of the wing pouch with a steep transition region that propagates from the center to the edge of the pouch during the third larval instar. Although the Fj gradient is an important cue for polarization, it is unclear how the polarization is affected by cell division and the expanding Ds transition region, both of which can alter the distribution of Ft-Ds heterodimers around the cell periphery. We have developed a computational model to address these questions. In our model, the binding affinity of Ft and Ds depends on phosphorylation by Fj. We assume that the asymmetry of the Ft-Ds bond distribution around the cell periphery defines the polarization, with greater asymmetry promoting cell proliferation. Our model predicts that this asymmetry is greatest in the radially-expanding transition region that leaves polarized cells in its wake. These cells naturally retain their bond distribution asymmetry after division by rapidly replenishing Ft-Ds bonds at new cell-cell interfaces. Thus we predict that the distal localization of Dachs in cells throughout the pouch requires the movement of the Ds transition region and the simple presence, rather than any specific spatial pattern, of Fj. In the tissues of a developing organism, specialized proteins can control cell growth and give cells a sense of direction, e.g., which way is the head or the tail, by having their concentration vary throughout the tissue. In cells of the developing fruit fly wing, a protein called Dachs localizes on the side of the cell closest to the center of the tissue, indicating a directionality. The localization of Dachs is determined by the spatial distribution, around the periphery of a cell, of intercellular bonds of the proteins Fat and Dachsous between adjacent cells. Here we asked how this cell directionality is affected when cells divide and when the concentration of Dachsous changes over time. We use a computational model to show that as the circular step-up region of the Dachsous concentration profile sweeps radially outward, like rings radiating outward from where a pebble was dropped in a pond, it leaves polarized cells in its wake. Our model also shows how cells can naturally recover their directionality after cell division.
Collapse
Affiliation(s)
- Juliana C. Wortman
- Department of Physics and Astronomy, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
| | - Marcos Nahmad
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
| | - Peng Cheng Zhang
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
| | - Arthur D. Lander
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
| | - Clare C. Yu
- Department of Physics and Astronomy, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Configuring a robust nervous system with Fat cadherins. Semin Cell Dev Biol 2017; 69:91-101. [PMID: 28603077 DOI: 10.1016/j.semcdb.2017.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/26/2017] [Accepted: 06/07/2017] [Indexed: 01/14/2023]
Abstract
Atypical Fat cadherins represent a small but versatile group of signaling molecules that influence proliferation and tissue polarity. With huge extracellular domains and intracellular domains harboring many independent protein interaction sites, Fat cadherins are poised to translate local cell adhesion events into a variety of cell behaviors. The need for such global coordination is particularly prominent in the nervous system, where millions of morphologically diverse neurons are organized into functional networks. As we learn more about their biological functions and molecular properties, increasing evidence suggests that Fat cadherins mediate contact-induced changes that ultimately impose a structure to developing neuronal circuits.
Collapse
|
48
|
Keira Y, Wada M, Ishikawa HO. Regulation of Drosophila Development by the Golgi Kinase Four-Jointed. Curr Top Dev Biol 2017; 123:143-179. [DOI: 10.1016/bs.ctdb.2016.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
49
|
Misra JR, Irvine KD. Vamana Couples Fat Signaling to the Hippo Pathway. Dev Cell 2016; 39:254-266. [PMID: 27746048 DOI: 10.1016/j.devcel.2016.09.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 08/09/2016] [Accepted: 09/15/2016] [Indexed: 01/08/2023]
Abstract
The protocadherins Dachsous and Fat initiate a signaling pathway that controls growth and planar cell polarity by regulating the membrane localization of the atypical myosin Dachs. How Dachs is regulated by Fat signaling has remained unclear. Here we identify the vamana gene as playing a crucial role in regulating membrane localization of Dachs and in linking Fat and Dachsous to Dachs regulation. Vamana, an SH3-domain-containing protein, physically associates with and co-localizes with Dachs and promotes its membrane localization. Vamana also associates with the Dachsous intracellular domain and with a region of the Fat intracellular domain that is essential for controlling Hippo signaling and levels of Dachs. Epistasis experiments, structure-function analysis, and physical interaction experiments argue that Fat negatively regulates Dachs in a Vamana-dependent process. Our findings establish Vamana as a crucial component of the Dachsous-Fat pathway that transmits Fat signaling by regulating Dachs.
Collapse
Affiliation(s)
- Jyoti R Misra
- Department of Molecular Biology and Biochemistry, Howard Hughes Medical Institute, Waksman Institute, Rutgers University, Piscataway NJ 08854, USA
| | - Kenneth D Irvine
- Department of Molecular Biology and Biochemistry, Howard Hughes Medical Institute, Waksman Institute, Rutgers University, Piscataway NJ 08854, USA.
| |
Collapse
|
50
|
Clarke DN, Miller PW, Lowe CJ, Weis WI, Nelson WJ. Characterization of the Cadherin-Catenin Complex of the Sea Anemone Nematostella vectensis and Implications for the Evolution of Metazoan Cell-Cell Adhesion. Mol Biol Evol 2016; 33:2016-29. [PMID: 27189570 PMCID: PMC4948710 DOI: 10.1093/molbev/msw084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cadherin-catenin complex (CCC) mediates cell-cell adhesion in bilaterian animals by linking extracellular cadherin-based adhesions to the actin cytoskeleton. However, it is unknown whether the basic organization of the complex is conserved across all metazoans. We tested whether protein interactions and actin-binding properties of the CCC are conserved in a nonbilaterian animal, the sea anemone Nematostella vectensis We demonstrated that N. vectensis has a complete repertoire of cadherin-catenin proteins, including two classical cadherins, one α-catenin, and one β-catenin. Using size-exclusion chromatography and multi-angle light scattering, we showed that α-catenin and β-catenin formed a heterodimer that bound N. vectensis Cadherin-1 and -2. Nematostella vectensis α-catenin bound F-actin with equivalent affinity as either a monomer or an α/β-catenin heterodimer, and its affinity for F-actin was, in part, regulated by a novel insert between the N- and C-terminal domains. Nematostella vectensis α-catenin inhibited Arp2/3 complex-mediated nucleation of actin filaments, a regulatory property previously thought to be unique to mammalian αE-catenin. Thus, despite significant differences in sequence, the key interactions of the CCC are conserved between bilaterians and cnidarians, indicating that the core function of the CCC as a link between cell adhesions and the actin cytoskeleton is ancestral in the eumetazoans.
Collapse
Affiliation(s)
| | - Phillip W Miller
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine
| | | | - William I Weis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine Department of Structural Biology, Stanford University School of Medicine
| | - William James Nelson
- Department of Biology, Stanford University Department of Molecular and Cellular Physiology, Stanford University School of Medicine
| |
Collapse
|