1
|
Tahami MS, Vargas-Chavez C, Poikela N, Coronado-Zamora M, González J, Kankare M. Transposable elements in Drosophila montana from harsh cold environments. Mob DNA 2024; 15:18. [PMID: 39354634 PMCID: PMC11445987 DOI: 10.1186/s13100-024-00328-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/17/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Substantial discoveries during the past century have revealed that transposable elements (TEs) can play a crucial role in genome evolution by affecting gene expression and inducing genetic rearrangements, among other molecular and structural effects. Yet, our knowledge on the role of TEs in adaptation to extreme climates is still at its infancy. The availability of long-read sequencing has opened up the possibility to identify and study potential functional effects of TEs with higher precision. In this work, we used Drosophila montana as a model for cold-adapted organisms to study the association between TEs and adaptation to harsh climates. RESULTS Using the PacBio long-read sequencing technique, we de novo identified and manually curated TE sequences in five Drosophila montana genomes from eco-geographically distinct populations. We identified 489 new TE consensus sequences which represented 92% of the total TE consensus in D. montana. Overall, 11-13% of the D. montana genome is occupied by TEs, which as expected are non-randomly distributed across the genome. We identified five potentially active TE families, most of them from the retrotransposon class of TEs. Additionally, we found TEs present in the five analyzed genomes that were located nearby previously identified cold tolerant genes. Some of these TEs contain promoter elements and transcription binding sites. Finally, we detected TEs nearby fixed and polymorphic inversion breakpoints. CONCLUSIONS Our research revealed a significant number of newly identified TE consensus sequences in the genome of D. montana, suggesting that non-model species should be studied to get a comprehensive view of the TE repertoire in Drosophila species and beyond. Genome annotations with the new D. montana library allowed us to identify TEs located nearby cold tolerant genes, and present at high population frequencies, that contain regulatory regions and are thus good candidates to play a role in D. montana cold stress response. Finally, our annotations also allow us to identify for the first time TEs present in the breakpoints of three D. montana inversions.
Collapse
Affiliation(s)
- Mohadeseh S Tahami
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
| | | | - Noora Poikela
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland
- Centre for Biological Diversity, University of St Andrews, St Andrews, UK
| | - Marta Coronado-Zamora
- Institute of Evolutionary Biology, CSIC, UPF, Barcelona, Spain
- Institut Botànic de Barcelona (IBB), CSIC-CMCNB, Barcelona, 08038, Catalonia, Spain
| | - Josefa González
- Institute of Evolutionary Biology, CSIC, UPF, Barcelona, Spain.
- Institut Botànic de Barcelona (IBB), CSIC-CMCNB, Barcelona, 08038, Catalonia, Spain.
| | - Maaria Kankare
- Department of Biological and Environmental Science, University of Jyväskylä, Jyväskylä, Finland.
| |
Collapse
|
2
|
Bollepogu Raja KK, Yeung K, Shim YK, Mardon G. Integrative genomic analyses reveal putative cell type-specific targets of the Drosophila ets transcription factor Pointed. BMC Genomics 2024; 25:103. [PMID: 38262913 PMCID: PMC10807358 DOI: 10.1186/s12864-024-10017-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/15/2024] [Indexed: 01/25/2024] Open
Abstract
The Ets domain transcription factors direct diverse biological processes throughout all metazoans and are implicated in development as well as in tumor initiation, progression and metastasis. The Drosophila Ets transcription factor Pointed (Pnt) is the downstream effector of the Epidermal growth factor receptor (Egfr) pathway and is required for cell cycle progression, specification, and differentiation of most cell types in the larval eye disc. Despite its critical role in development, very few targets of Pnt have been reported previously. Here, we employed an integrated approach by combining genome-wide single cell and bulk data to identify putative cell type-specific Pnt targets. First, we used chromatin immunoprecipitation with high-throughput sequencing (ChIP-seq) to determine the genome-wide occupancy of Pnt in late larval eye discs. We identified enriched regions that mapped to an average of 6,941 genes, the vast majority of which are novel putative Pnt targets. Next, we integrated ChIP-seq data with two other larval eye single cell genomics datasets (scRNA-seq and snATAC-seq) to reveal 157 putative cell type-specific Pnt targets that may help mediate unique cell type responses upon Egfr-induced differentiation. Finally, our integrated data also predicts cell type-specific functional enhancers that were not reported previously. Together, our study provides a greatly expanded list of putative cell type-specific Pnt targets in the eye and is a resource for future studies that will allow mechanistic insights into complex developmental processes regulated by Egfr signaling.
Collapse
Affiliation(s)
- Komal Kumar Bollepogu Raja
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kelvin Yeung
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yoon-Kyung Shim
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Graeme Mardon
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Viets K, Sauria MEG, Chernoff C, Rodriguez Viales R, Echterling M, Anderson C, Tran S, Dove A, Goyal R, Voortman L, Gordus A, Furlong EEM, Taylor J, Johnston RJ. Characterization of Button Loci that Promote Homologous Chromosome Pairing and Cell-Type-Specific Interchromosomal Gene Regulation. Dev Cell 2019; 51:341-356.e7. [PMID: 31607649 DOI: 10.1016/j.devcel.2019.09.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 06/07/2019] [Accepted: 09/09/2019] [Indexed: 01/05/2023]
Abstract
Homologous chromosomes colocalize to regulate gene expression in processes including genomic imprinting, X-inactivation, and transvection. In Drosophila, homologous chromosomes pair throughout development, promoting transvection. The "button" model of pairing proposes that specific regions along chromosomes pair with high affinity. Here, we identify buttons interspersed across the fly genome that pair with their homologous sequences, even when relocated to multiple positions in the genome. A majority of transgenes that span a full topologically associating domain (TAD) function as buttons, but not all buttons contain TADs. Additionally, buttons are enriched for insulator protein clusters. Fragments of buttons do not pair, suggesting that combinations of elements within a button are required for pairing. Pairing is necessary but not sufficient for transvection. Additionally, pairing and transvection are stronger in some cell types than in others, suggesting that pairing strength regulates transvection efficiency between cell types. Thus, buttons pair homologous chromosomes to facilitate cell-type-specific interchromosomal gene regulation.
Collapse
Affiliation(s)
- Kayla Viets
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Michael E G Sauria
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Chaim Chernoff
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Max Echterling
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Caitlin Anderson
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sang Tran
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Abigail Dove
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Raghav Goyal
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lukas Voortman
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Andrew Gordus
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eileen E M Furlong
- European Molecular Biology Laboratory, Department of Genome Biology, Heidelberg 69117, Germany
| | - James Taylor
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
4
|
miR-665 promotes hepatocellular carcinoma cell migration, invasion, and proliferation by decreasing Hippo signaling through targeting PTPRB. Cell Death Dis 2018; 9:954. [PMID: 30237408 PMCID: PMC6148030 DOI: 10.1038/s41419-018-0978-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 12/20/2022]
Abstract
Growing evidence suggests that aberrant microRNA (miRNA) expression contributes to hepatocellular carcinoma (HCC) development and progression. However, the potential role and mechanism of miR-665 in the progression of liver cancer remains largely unknown. Our current study showed that miR-665 expression was upregulated in HCC cells and tissues. High expression of miR-665 exhibited more severe tumor size, vascular invasion and Edmondson grading in HCC patients. Gain- or loss-of-function assays demonstrated that miR-665 promoted cell proliferation, migration, invasion, and the epithelial-mesenchymal transition (EMT) of HCC cells in vitro and in vivo. Tyrosine phosphatase receptor type B (PTPRB) was downregulated in HCC tissues, and was negatively correlated with miR-665 expression. Through western blotting and luciferase reporter assay, PTPRB was identified as a direct downstream target of miR-665. Restoration of PTPRB reverses the effects of miR-665 on HCC migration, invasion, and cell proliferation. A mechanistic study showed that PTPTRB mediated the functional role of miR-665 through regulation of the Hippo signaling pathway. In conclusion, our results suggested that miR-665 was a negative regulator of the PTPRB and could promote tumor proliferation and metastasis in HCC through decreasing Hippo signaling pathway activity, which can be a potential target for HCC treatment.
Collapse
|
5
|
Kim W, Khan SK, Liu Y, Xu R, Park O, He Y, Cha B, Gao B, Yang Y. Hepatic Hippo signaling inhibits protumoural microenvironment to suppress hepatocellular carcinoma. Gut 2018; 67:1692-1703. [PMID: 28866620 PMCID: PMC6592016 DOI: 10.1136/gutjnl-2017-314061] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/30/2017] [Accepted: 06/09/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hippo signalling is a recently identified major oncosuppressive pathway that plays critical roles in inhibiting hepatocyte proliferation, survival and hepatocellular carcinoma (HCC) formation. Hippo kinase (Mst1 and Mst2) inhibits HCC proliferation by suppressing Yap/Taz transcription activities. As human HCC is mainly driven by chronic liver inflammation, it is not clear whether Hippo signalling inhibits HCC by shaping its inflammatory microenvironment. DESIGN We have established a genetic HCC model by deleting Mst1 and Mst2 in hepatocytes. Functions of inflammatory responses in this model were characterised by molecular, cellular and FACS analysis, immunohistochemistry and genetic deletion of monocyte chemoattractant protein-1 (Mcp1) or Yap. Human HCC databases and human HCC samples were analysed by immunohistochemistry. RESULTS Genetic deletion of Mst1 and Mst2 in hepatocytes (DKO) led to HCC development, highly upregulated Mcp1 expression and massive infiltration of macrophages with mixed M1 and M2 phenotypes. Macrophage ablation or deletion of Mcp1 in DKO mice markedly reduced hepatic inflammation and HCC development. Moreover, Yap removal abolished induction of Mcp1 expression and restored normal liver growth in the Mst1/Mst2 DKO mice. Finally, we showed that MCP1 is a direct transcription target of YAP in hepatocytes and identified a strong gene expression correlation between YAP targets and MCP-1 in human HCCs. CONCLUSIONS Hippo signalling in hepatocytes maintains normal liver growth by suppressing macrophage infiltration during protumoural microenvironment formation through the inhibition of Yap-dependent Mcp1 expression, providing new targets and strategies to treat HCCs.
Collapse
Affiliation(s)
- Wantae Kim
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02215, USA.,Genetic Disease Research Branch, National Human Genome Research Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - Sanjoy Kumar Khan
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02215, USA.,Genetic Disease Research Branch, National Human Genome Research Institute, National Institute of Health, Bethesda, MD 20892, USA
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02215, USA
| | - Ruoshi Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02215, USA.,West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ogyi Park
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong He
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Boksik Cha
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.,Correspondence to:Yingzi Yang, PhD, Professor of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Avenue, Boston, MA, 02115, USA, Tel: 617–432–8304, Fax: 617–432–3246, , Bin Gao, MD, PhD, Senior Investigator, Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2S-33, Bethesda, MD 20892, Tel: 301.443.3998; Fax: 301 480.0257,
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02215, USA.,Genetic Disease Research Branch, National Human Genome Research Institute, National Institute of Health, Bethesda, MD 20892, USA.,Correspondence to:Yingzi Yang, PhD, Professor of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Avenue, Boston, MA, 02115, USA, Tel: 617–432–8304, Fax: 617–432–3246, , Bin Gao, MD, PhD, Senior Investigator, Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2S-33, Bethesda, MD 20892, Tel: 301.443.3998; Fax: 301 480.0257,
| |
Collapse
|
6
|
Wigerius M, Quinn D, Diab A, Clattenburg L, Kolar A, Qi J, Krueger SR, Fawcett JP. The polarity protein Angiomotin p130 controls dendritic spine maturation. J Cell Biol 2018; 217:715-730. [PMID: 29317530 PMCID: PMC5800806 DOI: 10.1083/jcb.201705184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/31/2017] [Accepted: 11/30/2017] [Indexed: 01/06/2023] Open
Abstract
Wigerius et al. identify the polarity protein AMOT-130 as vital for dendritic spine morphogenesis. They show that reduced Lats1 kinase activity in the neonatal brain is required for the recruitment of AMOT-130 to postsynaptic compartments to stabilize dendritic spines. The actin cytoskeleton is essential for the structural changes in dendritic spines that lead to the formation of new synapses. Although the molecular mechanisms underlying spine formation are well characterized, the events that drive spine maturation during development are largely unknown. In this study, we demonstrate that Angiomotin (AMOT-130) is necessary for spine stabilization. AMOT-130 is enriched in mature dendritic spines and functions to stabilize the actin cytoskeleton by coupling F-actin to postsynaptic protein scaffolds. These functions of AMOT are transiently restricted during postnatal development by phosphorylation imposed by the kinase Lats1. Our study proposes that AMOT-130 is essential for normal spine morphogenesis and identifies Lats1 as an upstream regulator in this process. Moreover, our findings may link AMOT-130 loss and the related spine defects to neurological disorders.
Collapse
Affiliation(s)
- Michael Wigerius
- Department of Pharmacology, Dalhousie University, Halifax, Canada
| | - Dylan Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Antonios Diab
- Department of Pharmacology, Dalhousie University, Halifax, Canada
| | | | - Annette Kolar
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Jiansong Qi
- Department of Pharmacology, Dalhousie University, Halifax, Canada
| | - Stefan R Krueger
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - James P Fawcett
- Department of Pharmacology, Dalhousie University, Halifax, Canada .,Department of Surgery, Dalhousie University, Halifax, Canada
| |
Collapse
|
7
|
Anderson C, Reiss I, Zhou C, Cho A, Siddiqi H, Mormann B, Avelis CM, Deford P, Bergland A, Roberts E, Taylor J, Vasiliauskas D, Johnston RJ. Natural variation in stochastic photoreceptor specification and color preference in Drosophila. eLife 2017; 6:29593. [PMID: 29251595 PMCID: PMC5745083 DOI: 10.7554/elife.29593] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 12/15/2017] [Indexed: 11/25/2022] Open
Abstract
Each individual perceives the world in a unique way, but little is known about the genetic basis of variation in sensory perception. In the fly eye, the random mosaic of color-detecting R7 photoreceptor subtypes is determined by stochastic on/off expression of the transcription factor Spineless (Ss). In a genome-wide association study, we identified a naturally occurring insertion in a regulatory DNA element in ss that lowers the ratio of SsON to SsOFF cells. This change in photoreceptor fates shifts the innate color preference of flies from green to blue. The genetic variant increases the binding affinity for Klumpfuss (Klu), a zinc finger transcriptional repressor that regulates ss expression. Klu is expressed at intermediate levels to determine the normal ratio of SsON to SsOFF cells. Thus, binding site affinity and transcription factor levels are finely tuned to regulate stochastic expression, setting the ratio of alternative fates and ultimately determining color preference.
Collapse
Affiliation(s)
- Caitlin Anderson
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - India Reiss
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Cyrus Zhou
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Annie Cho
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Haziq Siddiqi
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Benjamin Mormann
- Center for Developmental Genetics, Department of Biology, New York University, New York, United States
| | - Cameron M Avelis
- Department of Biophysics, Johns Hopkins University, Baltimore, United States
| | - Peter Deford
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Alan Bergland
- Department of Biology, University of Virginia, Charlottesville, United States
| | - Elijah Roberts
- Department of Biophysics, Johns Hopkins University, Baltimore, United States
| | - James Taylor
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Daniel Vasiliauskas
- Paris-Saclay Institute of Neuroscience, Université Paris Sud, Centre National de la Recherche Scientifque, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, Baltimore, United States
| |
Collapse
|
8
|
Yan J, Anderson C, Viets K, Tran S, Goldberg G, Small S, Johnston RJ. Regulatory logic driving stable levels of defective proventriculus expression during terminal photoreceptor specification in flies. Development 2017; 144:844-855. [PMID: 28126841 DOI: 10.1242/dev.144030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/02/2017] [Indexed: 12/13/2022]
Abstract
How differential levels of gene expression are controlled in post-mitotic neurons is poorly understood. In the Drosophila retina, expression of the transcription factor Defective Proventriculus (Dve) at distinct cell type-specific levels is required for terminal differentiation of color- and motion-detecting photoreceptors. Here, we find that the activities of two cis-regulatory enhancers are coordinated to drive dve expression in the fly eye. Three transcription factors act on these enhancers to determine cell-type specificity. Negative autoregulation by Dve maintains expression from each enhancer at distinct homeostatic levels. One enhancer acts as an inducible backup ('dark' shadow enhancer) that is normally repressed but becomes active in the absence of the other enhancer. Thus, two enhancers integrate combinatorial transcription factor input, feedback and redundancy to generate cell type-specific levels of dve expression and stable photoreceptor fate. This regulatory logic may represent a general paradigm for how precise levels of gene expression are established and maintained in post-mitotic neurons.
Collapse
Affiliation(s)
- Jenny Yan
- Department of Biology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218-2685, USA
| | - Caitlin Anderson
- Department of Biology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218-2685, USA
| | - Kayla Viets
- Department of Biology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218-2685, USA
| | - Sang Tran
- Department of Biology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218-2685, USA
| | - Gregory Goldberg
- Center for Developmental Genetics, Department of Biology, New York University, 100 Washington Square East, New York, NY 10003-6688, USA
| | - Stephen Small
- Center for Developmental Genetics, Department of Biology, New York University, 100 Washington Square East, New York, NY 10003-6688, USA
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218-2685, USA
| |
Collapse
|