1
|
Hassan A, Blakeley G, McGregor AP, Zancolli G. Venom gland organogenesis in the common house spider. Sci Rep 2024; 14:15379. [PMID: 38965282 PMCID: PMC11224297 DOI: 10.1038/s41598-024-65336-2] [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: 03/14/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
Venom is a remarkable innovation found across the animal kingdom, yet the evolutionary origins of venom systems in various groups, including spiders, remain enigmatic. Here, we investigated the organogenesis of the venom apparatus in the common house spider, Parasteatoda tepidariorum. The venom apparatus consists of a pair of secretory glands, each connected to an opening at the fang tip by a duct that runs through the chelicerae. We performed bulk RNA-seq to identify venom gland-specific markers and assayed their expression using RNA in situ hybridisation experiments on whole-mount time-series. These revealed that the gland primordium emerges during embryonic stage 13 at the chelicera tip, progresses proximally by the end of embryonic development and extends into the prosoma post-eclosion. The initiation of expression of an important toxin component in late postembryos marks the activation of venom-secreting cells. Our selected markers also exhibited distinct expression patterns in adult venom glands: sage and the toxin marker were expressed in the secretory epithelium, forkhead and sum-1 in the surrounding muscle layer, while Distal-less was predominantly expressed at the gland extremities. Our study provides the first comprehensive analysis of venom gland morphogenesis in spiders, offering key insights into their evolution and development.
Collapse
Affiliation(s)
- Afrah Hassan
- Department of Ecology and Evolution, University of Lausanne, Lausanne, 1015, Switzerland
| | - Grace Blakeley
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | | | - Giulia Zancolli
- Department of Ecology and Evolution, University of Lausanne, Lausanne, 1015, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland.
| |
Collapse
|
2
|
Matthew J, Vishwakarma V, Le TP, Agsunod RA, Chung S. Coordination of cell cycle and morphogenesis during organ formation. eLife 2024; 13:e95830. [PMID: 38275142 PMCID: PMC10869137 DOI: 10.7554/elife.95830] [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: 01/07/2024] [Accepted: 01/21/2024] [Indexed: 01/27/2024] Open
Abstract
Organ formation requires precise regulation of cell cycle and morphogenetic events. Using the Drosophila embryonic salivary gland (SG) as a model, we uncover the role of the SP1/KLF transcription factor Huckebein (Hkb) in coordinating cell cycle regulation and morphogenesis. The hkb mutant SG exhibits defects in invagination positioning and organ size due to the abnormal death of SG cells. Normal SG development involves distal-to-proximal progression of endoreplication (endocycle), whereas hkb mutant SG cells undergo abnormal cell division, leading to cell death. Hkb represses the expression of key cell cycle and pro-apoptotic genes in the SG. Knockdown of cyclin E or cyclin-dependent kinase 1, or overexpression of fizzy-related rescues most of the morphogenetic defects observed in the hkb mutant SG. These results indicate that Hkb plays a critical role in controlling endoreplication by regulating the transcription of key cell cycle effectors to ensure proper organ formation.
Collapse
Affiliation(s)
- Jeffrey Matthew
- Department of Biological Sciences, Louisiana State UniversityBaton RougeUnited States
| | - Vishakha Vishwakarma
- Department of Biological Sciences, Louisiana State UniversityBaton RougeUnited States
| | - Thao Phuong Le
- Department of Biological Sciences, Louisiana State UniversityBaton RougeUnited States
| | - Ryan A Agsunod
- Department of Biological Sciences, Louisiana State UniversityBaton RougeUnited States
| | - SeYeon Chung
- Department of Biological Sciences, Louisiana State UniversityBaton RougeUnited States
| |
Collapse
|
3
|
Loganathan R, Levings DC, Kim JH, Wells MB, Chiu H, Wu Y, Slattery M, Andrew DJ. Ribbon boosts ribosomal protein gene expression to coordinate organ form and function. J Cell Biol 2022; 221:213030. [PMID: 35195669 PMCID: PMC9237840 DOI: 10.1083/jcb.202110073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/19/2021] [Accepted: 01/24/2022] [Indexed: 11/22/2022] Open
Abstract
Cell growth is well defined for late (postembryonic) stages of development, but evidence for early (embryonic) cell growth during postmitotic morphogenesis is limited. Here, we report early cell growth as a key characteristic of tubulogenesis in the Drosophila embryonic salivary gland (SG) and trachea. A BTB/POZ domain nuclear factor, Ribbon (Rib), mediates this early cell growth. Rib binds the transcription start site of nearly every SG-expressed ribosomal protein gene (RPG) and is required for full expression of all RPGs tested. Rib binding to RPG promoters in vitro is weak and not sequence specific, suggesting that specificity is achieved through cofactor interactions. Accordingly, we demonstrate Rib’s ability to physically interact with each of the three known regulators of RPG transcription. Surprisingly, Rib-dependent early cell growth in another tubular organ, the embryonic trachea, is not mediated by direct RPG transcription. These findings support a model of early cell growth customized by transcriptional regulatory networks to coordinate organ form and function.
Collapse
Affiliation(s)
| | - Daniel C Levings
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN
| | - Ji Hoon Kim
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD
| | - Michael B Wells
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD
| | - Hannah Chiu
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD
| | - Yifan Wu
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD
| | - Matthew Slattery
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN
| | - Deborah J Andrew
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
4
|
Taracena M, Hunt C, Pennington P, Andrew D, Jacobs-Lorena M, Dotson E, Wells M. Effective Oral RNA Interference (RNAi) Administration to Adult Anopheles gambiae Mosquitoes. J Vis Exp 2022:10.3791/63266. [PMID: 35311819 PMCID: PMC9810275 DOI: 10.3791/63266] [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] [Indexed: 01/07/2023] Open
Abstract
RNA interference has been a heavily utilized tool for reverse genetic analysis for two decades. In adult mosquitoes, double-stranded RNA (dsRNA) administration has been accomplished primarily via injection, which requires significant time and is not suitable for field applications. To overcome these limitations, here we present a more efficient method for robust activation of RNAi by oral delivery of dsRNA to adult Anopheles gambiae. Long dsRNAs were produced in Escherichia coli strain HT115 (DE3), and a concentrated suspension of heat-killed dsRNA-containing bacteria in 10% sucrose was offered on cotton balls ad-libitum to adult mosquitoes. Cotton balls were replaced every 2 days for the duration of the treatment. Use of this method to target doublesex (a gene involved in sex differentiation) or fork head (which encodes a salivary gland transcription factor) resulted in reduced target gene expression and/or protein immunofluorescence signal, as measured by quantitative Real-Time PCR (qRT-PCR) or fluorescence confocal microscopy, respectively. Defects in salivary gland morphology were also observed. This highly flexible, user-friendly, low-cost, time-efficient method of dsRNA delivery could be broadly applicable to target genes important for insect vector physiology and beyond.
Collapse
Affiliation(s)
- Mabel Taracena
- Division of Parasitic Diseases and Malaria, Entomology Branch, Centers for Disease Control and Prevention; Department of Entomology, Cornell University;
| | - Catherine Hunt
- Division of Parasitic Diseases and Malaria, Entomology Branch, Centers for Disease Control and Prevention
| | - Pamela Pennington
- Centro de Estudios en Biotecnologia, Universidad del Valle de Guatemala
| | - Deborah Andrew
- Department of Cell Biology, Johns Hopkins School of Medicine; Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health
| | - Marcelo Jacobs-Lorena
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health and Malaria Research Institute
| | - Ellen Dotson
- Division of Parasitic Diseases and Malaria, Entomology Branch, Centers for Disease Control and Prevention
| | - Michael Wells
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health; Department of Cell Biology, Johns Hopkins School of Medicine; Biomedical Sciences Department, Idaho College of Osteopathic Medicine;
| |
Collapse
|
5
|
Rust K, Wodarz A. Transcriptional Control of Apical-Basal Polarity Regulators. Int J Mol Sci 2021; 22:ijms222212340. [PMID: 34830224 PMCID: PMC8624420 DOI: 10.3390/ijms222212340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/06/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022] Open
Abstract
Cell polarity is essential for many functions of cells and tissues including the initial establishment and subsequent maintenance of epithelial tissues, asymmetric cell division, and morphogenetic movements. Cell polarity along the apical-basal axis is controlled by three protein complexes that interact with and co-regulate each other: The Par-, Crumbs-, and Scrib-complexes. The localization and activity of the components of these complexes is predominantly controlled by protein-protein interactions and protein phosphorylation status. Increasing evidence accumulates that, besides the regulation at the protein level, the precise expression control of polarity determinants contributes substantially to cell polarity regulation. Here we review how gene expression regulation influences processes that depend on the induction, maintenance, or abolishment of cell polarity with a special focus on epithelial to mesenchymal transition and asymmetric stem cell division. We conclude that gene expression control is an important and often neglected mechanism in the control of cell polarity.
Collapse
Affiliation(s)
- Katja Rust
- Department of Molecular Cell Physiology, Institute of Physiology and Pathophysiology, Philipps-University, 35037 Marburg, Germany
- Correspondence: (K.R.); (A.W.)
| | - Andreas Wodarz
- Department of Molecular Cell Biology, Institute I for Anatomy, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cluster of Excellence—Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
- Correspondence: (K.R.); (A.W.)
| |
Collapse
|
6
|
Loganathan R, Kim JH, Wells MB, Andrew DJ. Secrets of secretion-How studies of the Drosophila salivary gland have informed our understanding of the cellular networks underlying secretory organ form and function. Curr Top Dev Biol 2020; 143:1-36. [PMID: 33820619 DOI: 10.1016/bs.ctdb.2020.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Secretory organs are critical for organismal survival. Yet, the transcriptional regulatory mechanisms governing their development and maintenance remain unclear for most model secretory organs. The Drosophila embryonic salivary gland (SG) remedies this deficiency as one of the few organs wherein direct connections from the expression of the early patterning genes to cell specification to organ architecture and functional specialization can be made. Few other models of secretion can be accorded this distinction. Studies from the past three decades have made enormous strides in parsing out the roles of distinct transcription factors (TFs) that direct major steps in furnishing this secretory organ. In the first step of specifying the salivary gland, the activity of the Hox factors Sex combs reduced, Extradenticle, and Homothorax activate expression of fork head (fkh), sage, and CrebA, which code for the major suite of TFs that carry forward the task of organ building and maintenance. Then, in the second key step of building the SG, the program for cell fate maintenance and morphogenesis is deployed. Fkh maintains the secretory cell fate by regulating its own expression and that of sage and CrebA. Fkh and Sage maintain secretory cell viability by actively blocking apoptotic cell death. Fkh, along with two other TFs, Hkb and Rib, also coordinates organ morphogenesis, transforming two plates of precursor cells on the embryo surface into elongated internalized epithelial tubes. Acquisition of functional specialization, the third key step, is mediated by CrebA and Fkh working in concert with Sage and yet another TF, Sens. CrebA directly upregulates expression of all of the components of the secretory machinery as well as other genes (e.g., Xbp1) necessary for managing the physiological stress that inexorably accompanies high secretory load. Secretory cargo specificity is controlled by Sage and Sens in collaboration with Fkh. Investigations have also uncovered roles for various signaling pathways, e.g., Dpp signaling, EGF signaling, GPCR signaling, and cytoskeletal signaling, and their interactions within the gene regulatory networks that specify, build, and specialize the SG. Collectively, studies of the SG have expanded our knowledge of secretory dynamics, cell polarity, and cytoskeletal mechanics in the context of organ development and function. Notably, the embryonic SG has made the singular contribution as a model system that revealed the core function of CrebA in scaling up secretory capacity, thus, serving as the pioneer system in which the conserved roles of the mammalian Creb3/3L-family orthologues were first discovered.
Collapse
Affiliation(s)
- Rajprasad Loganathan
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ji Hoon Kim
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael B Wells
- Idaho College of Osteopathic Medicine, Meridian, ID, United States
| | - Deborah J Andrew
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
7
|
Hu Q, Zhu Z, Zhao D, Zeng B, Zheng S, Song Q, Deng H, Feng Q. Bombyx mori transcription factors FoxA and SAGE divergently regulate the expression of wing cuticle protein gene 4 during metamorphosis. J Biol Chem 2018; 294:632-643. [PMID: 30429222 DOI: 10.1074/jbc.ra118.004395] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/07/2018] [Indexed: 01/05/2023] Open
Abstract
Stage-specific gene expression governs metamorphosis of the silkworm, Bombyx mori. B. mori wing cuticle protein gene 4 (BmWCP4) is an essential gene for wing disc development expressed specifically during pupation. BmWCP4 transcription is suppressed at the larval stage by unknown mechanisms, which we sought to elucidate here. Bioinformatics analysis predicted seven potential Forkhead box (Fox) cis-regulatory elements (CREs) in the BmWCP4 promoter region, and we found that Fox CRE6 contributes to suppression of BmWCP4 expression. Electrophoretic mobility shift (EMSA) and DNA pull-down assays revealed that BmFoxA suppressed activity at the BmWCP4 promoter by specifically binding to the Fox CRE6. The expression level of BmFoxA in the wing discs was higher during the larval stage than at the pupal stage. In contrast, expression of another transcription factor, BmSAGE, increased over the course of development. Of note, the hormone 20-hydroxyecdysone (20E), which governs molting in insects, suppressed BmFoxA expression in the wing discs and up-regulated that of BmSage EMSA and cell co-transfection assays indicated that BmSAGE interacted with BmFoxA and suppressed its binding to the Fox CRE6, thereby releasing BmFoxA-mediated suppression of BmWCP4 In summary, higher BmFoxA expression during the larval stage suppresses BmWCP4 expression by binding to the Fox CRE6 on the BmWCP4 promoter. During metamorphosis, BmSAGE forms a complex with BmFoxA to relieve this repression, initiating BmWCP4 expression. Taken together, this study reveals a switchlike role for BmFoxA in regulating BmWCP4 expression and provides new insights into the regulatory regulation of wing disc development in insects.
Collapse
Affiliation(s)
- Qihao Hu
- From the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China and
| | - Zidan Zhu
- From the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China and
| | - Danhui Zhao
- From the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China and
| | - Baojuan Zeng
- From the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China and
| | - Sichun Zheng
- From the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China and
| | - Qisheng Song
- the Division of Plant Sciences, University of Missouri, Columbia, Missouri 65211
| | - Huimin Deng
- From the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China and
| | - Qili Feng
- From the Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou 510631, China and
| |
Collapse
|
8
|
Vuolo F, Kierzkowski D, Runions A, Hajheidari M, Mentink RA, Gupta MD, Zhang Z, Vlad D, Wang Y, Pecinka A, Gan X, Hay A, Huijser P, Tsiantis M. LMI1 homeodomain protein regulates organ proportions by spatial modulation of endoreduplication. Genes Dev 2018; 32:1361-1366. [PMID: 30366902 PMCID: PMC6217736 DOI: 10.1101/gad.318212.118] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022]
Abstract
Here, Vuolo et al. investigated the mechanisms controlling the relative size of leaves compared with their lateral appendages (stipules). Using genetics, live imaging, and modeling, they demonstrate that the LATE MERISTEM IDENTITY1 (LMI1) homeodomain protein regulates stipule proportions via an endoreduplication-dependent trade-off that limits tissue size despite increasing cell growth. How the interplay between cell- and tissue-level processes produces correctly proportioned organs is a key problem in biology. In plants, the relative size of leaves compared with their lateral appendages, called stipules, varies tremendously throughout development and evolution, yet relevant mechanisms remain unknown. Here we use genetics, live imaging, and modeling to show that in Arabidopsis leaves, the LATE MERISTEM IDENTITY1 (LMI1) homeodomain protein regulates stipule proportions via an endoreduplication-dependent trade-off that limits tissue size despite increasing cell growth. LM1 acts through directly activating the conserved mitosis blocker WEE1, which is sufficient to bypass the LMI1 requirement for leaf proportionality.
Collapse
Affiliation(s)
- Francesco Vuolo
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Daniel Kierzkowski
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Adam Runions
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Mohsen Hajheidari
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Remco A Mentink
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Mainak Das Gupta
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Zhongjuan Zhang
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Daniela Vlad
- Department of Plant Sciences, University of Oxford, Oxford OX1 3RB, United Kingdom
| | - Yi Wang
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Ales Pecinka
- Department of Plant Breeding Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Xiangchao Gan
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Angela Hay
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Peter Huijser
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| | - Miltos Tsiantis
- Deparment of Comparative Development and Genetics, Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany
| |
Collapse
|
9
|
Hanlon CD, Andrew DJ. Drosophila FoxL1 non-autonomously coordinates organ placement during embryonic development. Dev Biol 2016; 419:273-284. [PMID: 27618755 DOI: 10.1016/j.ydbio.2016.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 12/13/2022]
Abstract
Determining how organs attain precise positioning within an organism is a crucial facet of developmental biology. The Fox family winged-helix transcription factors are known to play key roles in development of multiple organs. Drosophila FoxL1 (aka Fd64A) is dynamically expressed in embryos but its function is completely uncharacterized. FoxL1 is expressed in a single group of body wall - muscles in the 2nd and 3rd thoracic segments, in homologous abdominal muscles at earlier stages, and in the hindgut mesoderm from early through late embryogenesis. We show that FoxL1 expression in T2 and T3 is in VIS5, which is not a single muscle spanning the entire thorax, as previously published, but is, instead, three individual muscles, each spanning a single thoracic segment. We generate mutations in foxL1 and show that, surprisingly, none of the tissues that express FoxL1 are affected by its loss. Instead, loss of foxL1 results in defects in salivary gland positioning and morphology, as well as defects in the migration of hemocytes, germ cells and Malpighian tubules. We also show that FoxL1-dependent expression of secreted Sema2a in T3 VIS5 is required for normal salivary gland positioning. Altogether, these findings suggest that Drosophila FoxL1 functions like its mammalian counterpart in non-autonomously orchestrating the behaviors of surrounding tissues.
Collapse
Affiliation(s)
- Caitlin D Hanlon
- Department of Cell Biology, The Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205-2196, United States
| | - Deborah J Andrew
- Department of Cell Biology, The Johns Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD 21205-2196, United States.
| |
Collapse
|
10
|
Liu ZY, Yu Q, Yang CH, Meng M, Ren CJ, Mu ZM, Cui WZ, Liu QX. Transcription factor SGF1 is critical for the neurodevelopment in the silkworm, Bombyx mori. Gene 2016; 587:70-5. [PMID: 27106119 DOI: 10.1016/j.gene.2016.04.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/06/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
FoxA transcription factors play vital roles in regulating the expression of organ-specific genes. BmSGF1, the sole FoxA family member in Bombyx mori, is required for development of the silk gland. However, the function of BmSGF1 in development of the nervous system in the silkworm remains unknown. Here, we show that the amino acids sequence of BmSGF1 is evolutionarily conserved in its middle region from Trichoplax adhaerens to human and diverged from the homologues in most other species in its N-terminal region. BmSGF1 expresses in the nervous system at the embryonic stage. Knockdown of Bmsgf1 by RNA interference (RNAi) results in abnormal development of axons. Therefore, our results demonstrate that BmSGF1 is an indispensable regulator for neurodevelopment.
Collapse
Affiliation(s)
- Zhao-Yang Liu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Qi Yu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Chun-Hong Yang
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Miao Meng
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Chun-Jiu Ren
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Zhi-Mei Mu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Wei-Zheng Cui
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Qing-Xin Liu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China.
| |
Collapse
|
11
|
Sun L, Tang XJ, Luo FM. Forkhead box protein A2 and T helper type 2-mediated pulmonary inflammation. World J Methodol 2015; 5:223-229. [PMID: 26713283 PMCID: PMC4686420 DOI: 10.5662/wjm.v5.i4.223] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/24/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
The transcription factor forkhead box protein A2 (FOXA2, also known as hepatocyte nuclear factor 3β or transcription factor 3β), has been found to play pivotal roles in multiple phases of mammalian life, from the early development to the organofaction, and subsequently in homeostasis and metabolism in the adult. In the embryonic development period, FOXA2 is require d for the formation of the primitive node and notochord, and its absence results in embryonic lethality. Moreover, FOXA2 plays an important role not only in lung development, but also in T helper type 2 (Th2)-mediated pulmonary inflammation and goblet cell hyperplasia. In this article, the role of FOXA2 in lung development and Th2-mediated pulmonary inflammation, as well as in goblet cell hyperplasia, is reviewed. FOXA2 deletion in airway epithelium results into Th2-mediated pulmonary inflammation and goblet cell hyperplasia in developing lung. Leukotriene pathway and signal transducers and activators of transcription 6 pathway may mediate this inflammation through recruitment and activation of denditric cell during lung developments. FOXA2 is a potential treatment target for lung diseases with Th2 inflammation and goblet cell hyperplasia, such as asthma and chronic obstructive pulmonary disease.
Collapse
|
12
|
Loganathan R, Lee JS, Wells MB, Grevengoed E, Slattery M, Andrew DJ. Ribbon regulates morphogenesis of the Drosophila embryonic salivary gland through transcriptional activation and repression. Dev Biol 2015; 409:234-250. [PMID: 26477561 DOI: 10.1016/j.ydbio.2015.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 01/08/2023]
Abstract
Transcription factors affect spatiotemporal patterns of gene expression often regulating multiple aspects of tissue morphogenesis, including cell-type specification, cell proliferation, cell death, cell polarity, cell shape, cell arrangement and cell migration. In this work, we describe a distinct role for Ribbon (Rib) in controlling cell shape/volume increases during elongation of the Drosophila salivary gland (SG). Notably, the morphogenetic changes in rib mutants occurred without effects on general SG cell attributes such as specification, proliferation and apoptosis. Moreover, the changes in cell shape/volume in rib mutants occurred without compromising epithelial-specific morphological attributes such as apicobasal polarity and junctional integrity. To identify the genes regulated by Rib, we performed ChIP-seq analysis in embryos driving expression of GFP-tagged Rib specifically in the SGs. To learn if the Rib binding sites identified in the ChIP-seq analysis were linked to changes in gene expression, we performed microarray analysis comparing RNA samples from age-matched wild-type and rib null embryos. From the superposed ChIP-seq and microarray gene expression data, we identified 60 genomic sites bound by Rib likely to regulate SG-specific gene expression. We confirmed several of the identified Rib targets by qRT-pCR and/or in situ hybridization. Our results indicate that Rib regulates cell growth and tissue shape in the Drosophila salivary gland via a diverse array of targets through both transcriptional activation and repression. Furthermore, our results suggest that autoregulation of rib expression may be a key component of the SG morphogenetic gene network.
Collapse
Affiliation(s)
- Rajprasad Loganathan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Joslynn S Lee
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Michael B Wells
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Elizabeth Grevengoed
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Matthew Slattery
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Deborah J Andrew
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
13
|
Chung S, Hanlon CD, Andrew DJ. Building and specializing epithelial tubular organs: the Drosophila salivary gland as a model system for revealing how epithelial organs are specified, form and specialize. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:281-300. [PMID: 25208491 DOI: 10.1002/wdev.140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/02/2014] [Accepted: 04/15/2014] [Indexed: 12/28/2022]
Abstract
The past two decades have witnessed incredible progress toward understanding the genetic and cellular mechanisms of organogenesis. Among the organs that have provided key insight into how patterning information is integrated to specify and build functional body parts is the Drosophila salivary gland, a relatively simple epithelial organ specialized for the synthesis and secretion of high levels of protein. Here, we discuss what the past couple of decades of research have revealed about organ specification, development, specialization, and death, and what general principles emerge from these studies.
Collapse
Affiliation(s)
- SeYeon Chung
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Caitlin D Hanlon
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Deborah J Andrew
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Girdler GC, Röper K. Controlling cell shape changes during salivary gland tube formation in Drosophila. Semin Cell Dev Biol 2014; 31:74-81. [PMID: 24685610 DOI: 10.1016/j.semcdb.2014.03.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 03/18/2014] [Indexed: 12/23/2022]
Abstract
Any type of tubulogenesis is a process that is highly coordinated between large numbers of cells. Like other morphogenetic processes, it is driven to a great extent by complex cell shape changes and cell rearrangements. The formation of the salivary glands in the fly embryo provides an ideal model system to study these changes and rearrangements, because upon specification of the cells that are destined to form the tube, there is no further cell division or cell death. Thus, morphogenesis of the salivary gland tubes is entirely driven by cell shape changes and rearrangements. In this review, we will discuss and distill from the literature what is known about the control of cell shape during the early invagination process and whilst the tubes extend in the fly embryo at later stages.
Collapse
Affiliation(s)
- Gemma C Girdler
- MRC-Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Katja Röper
- MRC-Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
15
|
Sánchez-Herrero E. Hox targets and cellular functions. SCIENTIFICA 2013; 2013:738257. [PMID: 24490109 PMCID: PMC3892749 DOI: 10.1155/2013/738257] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/20/2013] [Indexed: 06/03/2023]
Abstract
Hox genes are a group of genes that specify structures along the anteroposterior axis in bilaterians. Although in many cases they do so by modifying a homologous structure with a different (or no) Hox input, there are also examples of Hox genes constructing new organs with no homology in other regions of the body. Hox genes determine structures though the regulation of targets implementing cellular functions and by coordinating cell behavior. The genetic organization to construct or modify a certain organ involves both a genetic cascade through intermediate transcription factors and a direct regulation of targets carrying out cellular functions. In this review I discuss new data from genome-wide techniques, as well as previous genetic and developmental information, to describe some examples of Hox regulation of different cell functions. I also discuss the organization of genetic cascades leading to the development of new organs, mainly using Drosophila melanogaster as the model to analyze Hox function.
Collapse
Affiliation(s)
- Ernesto Sánchez-Herrero
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
16
|
Fox RM, Vaishnavi A, Maruyama R, Andrew DJ. Organ-specific gene expression: the bHLH protein Sage provides tissue specificity to Drosophila FoxA. Development 2013; 140:2160-71. [PMID: 23578928 DOI: 10.1242/dev.092924] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
FoxA transcription factors play major roles in organ-specific gene expression, regulating, for example, glucagon expression in the pancreas, GLUT2 expression in the liver, and tyrosine hydroxylase expression in dopaminergic neurons. Organ-specific gene regulation by FoxA proteins is achieved through cooperative regulation with a broad array of transcription factors with more limited expression domains. Fork head (Fkh), the sole Drosophila FoxA family member, is required for the development of multiple distinct organs, yet little is known regarding how Fkh regulates tissue-specific gene expression. Here, we characterize Sage, a bHLH transcription factor expressed exclusively in the Drosophila salivary gland (SG). We show that Sage is required for late SG survival and normal tube morphology. We find that many Sage targets, identified by microarray analysis, encode SG-specific secreted cargo, transmembrane proteins, and the enzymes that modify these proteins. We show that both Sage and Fkh are required for the expression of Sage target genes, and that co-expression of Sage and Fkh is sufficient to drive target gene expression in multiple cell types. Sage and Fkh drive expression of the bZip transcription factor Senseless (Sens), which boosts expression of Sage-Fkh targets, and Sage, Fkh and Sens colocalize on SG chromosomes. Importantly, expression of Sage-Fkh target genes appears to simply add to the tissue-specific gene expression programs already established in other cell types, and Sage and Fkh cannot alter the fate of most embryonic cell types even when expressed early and continuously.
Collapse
Affiliation(s)
- Rebecca M Fox
- Department of Cell Biology, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205-2196, USA
| | | | | | | |
Collapse
|
17
|
Maruyama R, Andrew DJ. Drosophila as a model for epithelial tube formation. Dev Dyn 2011; 241:119-35. [PMID: 22083894 DOI: 10.1002/dvdy.22775] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2011] [Indexed: 12/17/2022] Open
Abstract
Epithelial tubular organs are essential for life in higher organisms and include the pancreas and other secretory organs that function as biological factories for the synthesis and delivery of secreted enzymes, hormones, and nutrients essential for tissue homeostasis and viability. The lungs, which are necessary for gas exchange, vocalization, and maintaining blood pH, are organized as highly branched tubular epithelia. Tubular organs include arteries, veins, and lymphatics, high-speed passageways for delivery and uptake of nutrients, liquids, gases, and immune cells. The kidneys and components of the reproductive system are also epithelial tubes. Both the heart and central nervous system of many vertebrates begin as epithelial tubes. Thus, it is not surprising that defects in tube formation and maintenance underlie many human diseases. Accordingly, a thorough understanding how tubes form and are maintained is essential to developing better diagnostics and therapeutics. Among the best-characterized tubular organs are the Drosophila salivary gland and trachea, organs whose relative simplicity have allowed for in depth analysis of gene function, yielding key mechanistic insight into tube initiation, remodeling and maintenance. Here, we review our current understanding of salivary gland and trachea formation - highlighting recent discoveries into how these organs attain their final form and function.
Collapse
Affiliation(s)
- Rika Maruyama
- The Johns Hopkins University School of Medicine, Department of Cell Biology, Baltimore, Maryland 21205-2196, USA
| | | |
Collapse
|