Fly Labs and References


A B C D E F G H I J K L M N O P Q R S T U V W X Y Z


  • Walt Eanes Department of Ecology and Evolution, Stony Brook University
    Cogni, R., Kuczynski, K., Koury, S., Lavington, E., Behrman, E. L., O'Brien, K. R., Schmidt, P. S. and Eanes, W. F. (2017). On the long-term stability of clines in some metabolic genes in Drosophila melanogaster. Sci Rep 7: 42766. PubMed ID: 28220806

    Talbert, M. E., Barnett, B., Hoff, R., Amella, M., Kuczynski, K., Lavington, E., Koury, S., Brud, E. and Eanes, W. F. (2015). Genetic perturbation of key central metabolic genes extends lifespan in Drosophila and affects response to dietary restriction. Proc Biol Sci 282. PubMed ID: 26378219

    Cogni, R., Kuczynski, K., Lavington, E., Koury, S., Behrman, E. L., O'Brien, K. R., Schmidt, P. S. and Eanes, W. F. (2015). Variation in Drosophila melanogaster central metabolic genes appears driven by natural selection both within and between populations. Proc Biol Sci 282: 20142688. PubMed ID: 25520361

    Lavington, E., Cogni, R., Kuczynski, C., Koury, S., Behrman, E. L., O'Brien, K. R., Schmidt, P. S. and Eanes, W. F. (2014). A small system--high-resolution study of metabolic adaptation in the central metabolic pathway to temperate climates in Drosophila melanogaster. Mol Biol Evol 31: 2032-2041. PubMed ID: 24770333

  • Bill Earnshaw Wellcome Trust Centre for Cell Biology, University of Edinburgh
    Carmena, M., Lombardia, M. O., Ogawa, H. and Earnshaw, W. C. (2014). Polo kinase regulates the localization and activity of the chromosomal passenger complex in meiosis and mitosis in Drosophila melanogaster. Open Biol 4. PubMed ID: 25376909

  • Suzanne Eaton Max Planck Institute for Cell Biology and Genetics, Dresden
    Ghosh, S., Leng, W., Wilsch-Brauninger, M., Barrera-Velazquez, M., Leopold, P. and Eaton, S. (2022). A local insulin reservoir in Drosophila alpha cell homologs ensures developmental progression under nutrient shortage. Curr Biol 32(8): 1788-1797.e1785. PubMed ID: 35316653

    Nellas, I., Iyer, K. V., Iglesias-Artola, J. M., Pippel, M., Nadler, A., Eaton, S. and Dye, N. A. (2022). Hedgehog signaling can enhance glycolytic ATP production in the Drosophila wing disc. EMBO Rep: e202154025. PubMed ID: 36134875

    Ghosh, S., Leng, W., Wilsch-Brauninger, M., Barrera-Velazquez, M., Leopold, P. and Eaton, S. (2022). A local insulin reservoir in Drosophila alpha cell homologs ensures developmental progression under nutrient shortage. Curr Biol. PubMed ID: 35316653

    Iyer, K. V., Taubenberger, A., Zeidan, S. A., Dye, N. A., Eaton, S. and Julicher, F. (2021). Apico-basal cell compression regulates Lamin A/C levels in epithelial tissues. Nat Commun 12(1): 1756. PubMed ID: 33767161

    Dye, N. A., Popovic, M., Iyer, K. V., Fuhrmann, J., Piscitello-Gomez, R., Eaton, S. and Julicher, F. (2021). Self-organized patterning of cell morphology via mechanosensitive feedback. Elife 10. PubMed ID: 33769281

    Iyer, K. V., Piscitello-Gomez, R., Paijmans, J., Julicher, F. and Eaton, S. (2019). Epithelial viscoelasticity is regulated by mechanosensitive E-cadherin turnover. Curr Biol. PubMed ID: 30744966

    Brankatschk, M., Gutmann, T., Knittelfelder, O., Palladini, A., Prince, E., Grzybek, M., Brankatschk, B., Shevchenko, A., Coskun, U. and Eaton, S. (2018). A temperature-dependent switch in feeding preference improves Drosophila development and survival in the cold. Dev Cell 46(6): 781-793.e784. PubMed ID: 30253170

    Azpurua, J., Mahoney, R. E. and Eaton, B. A. (2018). Transcriptomics of aged Drosophila motor neurons reveals a matrix metalloproteinase that impairs motor function. Aging Cell 17(2). PubMed ID: 29411505

    Dye, N. A., Popovic, M., Spannl, S., Etournay, R., Kainmuller, D., Ghosh, S., Myers, E. W., Julicher, F. and Eaton, S. (2017). Cell dynamics underlying oriented growth of the Drosophila wing imaginal disc. Development 144(23): 4406-4421. PubMed ID: 29038308

    Kreko-Pierce, T. and Eaton, B. A. (2017). The Drosophila LC8 homologue Cut-up specifies the axonal transport of proteasomes. J Cell Sci. PubMed ID: 28808087

  • Dan Eberl Department of Biology, University of Iowa, Iowa City
    Karak, S., Jacobs, J. S., Kittelmann, M., Spalthoff, C., Katana, R., Sivan-Loukianova, E., Schon, M. A., Kernan, M. J., Eberl, D. F. and Gopfert, M. C. (2015). Diverse roles of axonemal dyneins in Drosophila auditory neuron function and mechanical amplification in hearing. Sci Rep 5: 17085. PubMed ID: 26608786

    Chen, J. V., Kao, L. R., Jana, S. C., Sivan-Loukianova, E., Mendonca, S., Cabrera, O. A., Singh, P., Cabernard, C., Eberl, D. F., Bettencourt-Dias, M. and Megraw, T. L. (2015). Rootletin organizes the ciliary rootlet to achieve neuron sensory function in Drosophila. J Cell Biol 211: 435-453. PubMed ID: 26483560

    Kavlie, R. G., Fritz, J. L., Nies, F., Gopfert, M. C., Oliver, D., Albert, J. T. and Eberl, D. F. (2014). Prestin is an anion transporter dispensable for mechanical feedback amplification in Drosophila hearing. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 25412730

    Christie, K. W. and Eberl, D. F. (2014). Noise-induced hearing loss: new animal models. Curr Opin Otolaryngol Head Neck Surg. PubMed ID: 25111054

    Boekhoff-Falk, G. and Eberl, D. F. (2014). The Drosophila auditory system. Wiley Interdiscip Rev Dev Biol 3: 179-191. PubMed ID: 24719289

    Christie, K. W., Sivan-Loukianova, E., Smith, W. C., Aldrich, B. T., Schon, M. A., Roy, M., Lear, B. C. and Eberl, D. F. (2013). Physiological, anatomical, and behavioral changes after acoustic trauma in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 24003166

    Bharadwaj, R., Roy, M., Ohyama, T., Sivan-Loukianova, E., Delannoy, M., Lloyd, T. E., Zlatic, M., Eberl, D. F. and Kolodkin, A. L. (2013). Cbl-associated protein regulates assembly and function of two tension-sensing structures in Drosophila. Development 140: 627-638. PubMed ID: 23293294

    Roy, M., Sivan-Loukianova, E. and Eberl, D. F. (2013). Cell-type-specific roles of Na+/K+ ATPase subunits in Drosophila auditory mechanosensation. Proc Natl Acad Sci U S A 110: 181-186. PubMed ID: 23248276

    Kavlie, R. G., Kernan, M. J. and Eberl, D. F. (2010). Hearing in Drosophila requires TilB, a conserved protein associated with ciliary motility. Genetics 185: 177-188. PubMed ID: 20215474

  • Isaac Edery Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ
    Yang, Y. and Edery, I. (2019). Daywake, an anti-siesta gene linked to a splicing-based thermostat from an adjoining clock gene. Curr Biol 29(10): 1728-1734.e1724. PubMed ID: 31080079

    Yang, Y. and Edery, I. (2018). Parallel clinal variation in the mid-day siesta of Drosophila melanogaster implicates continent-specific targets of natural selection. PLoS Genet 14(9): e1007612. PubMed ID: 30180162

    Zhang, Z., Cao, W. and Edery, I. (2018). The SR protein B52/SRp55 regulates splicing of the period thermosensitive intron and mid-day siesta in Drosophila. Sci Rep 8(1): 1872. PubMed ID: 29382842

    Cao, W. and Edery, I. (2017). Mid-day siesta in natural populations of D. melanogaster from Africa exhibits an altitudinal cline and is regulated by splicing of a thermosensitive intron in the period clock gene. BMC Evol Biol 17(1): 32. PubMed ID: 28114910

    Yildirim, E., Chiu, J. C. and Edery, I. (2015). Identification of light-sensitive phosphorylation sites on PERIOD that regulate the pace of circadian rhythms in Drosophila. Mol Cell Biol. PubMed ID: 26711257

    Kwok, R.S., Li, Y.H., Lei, A.J., Edery, I. and Chiu, J.C. (2015). The catalytic and non-catalytic functions of the Brahma chromatin-remodeling protein collaborate to fine-tune circadian transcription in Drosophila. PLoS Genet 11: e1005307. PubMed ID: 26132408

    Cao, W. and Edery, I. (2014). A novel pathway for sensory-mediated arousal involves splicing of an intron in the clock gene. Sleep. PubMed ID: 25325457

    Lee, E., Jeong, E. H., Jeong, H. J., Yildirim, E., Vanselow, J. T., Ng, F., Liu, Y., Mahesh, G., Kramer, A., Hardin, P. E., Edery, I. and Kim, E. Y. (2014). Phosphorylation of a central clock transcription factor is required for thermal but not photic entrainment. PLoS Genet 10: e1004545. PubMed ID: 25121504

    Mahesh, G., Jeong, E., Ng, F. S., Liu, Y., Gunawardhana, K., Houl, J. H., Yildirim, E., Amunugama, R., Jones, R., Allen, D. L., Edery, I., Kim, E. Y. and Hardin, P. E. (2014). Phosphorylation of the Transcription Activator CLOCK Regulates Progression through a approximately 24-h Feedback Loop to Influence the Circadian Period in Drosophila. J Biol Chem 289: 19681-19693. PubMed ID: 24872414

    Low, K. H., Chen, W. F., Yildirim, E. and Edery, I. (2012). Natural variation in the Drosophila melanogaster clock gene period modulates splicing of its 3'-terminal intron and mid-day siesta. PLoS One 7: e49536. PubMed ID: 23152918

  • Bruce Edgar University of Utah
    Li, Y., Chaurasia, B., Rahman, M. M., Kaddai, V., Maschek, J. A., Berg, J. A., Wilkerson, J. L., Mahmassani, Z. S., Cox, J., Wei, P., Meikle, P. J., Atkinson, D., Wang, L., Poss, A. M., Playdon, M. C., Tippetts, T. S., Mousa, E. M., Nittayaboon, K., Anandh Babu, P. V., Drummond, M. J., Clevers, H., Shayman, J. A., Hirabayashi, Y., Holland, W. L., Rutter, J., Edgar, B. and Summers, S. A. (2023). Ceramides increase fatty acid utilization in intestinal progenitors to enhance stemness and increase tumor risk. Gastroenterology. PubMed ID: 37541526

    Zhang, C., Jin, Y., Marchetti, M., Lewis, M. R., Hammouda, O. T. and Edgar, B. A. (2022). EGFR signaling activates intestinal stem cells by promoting mitochondrial biogenesis and β-oxidation. Curr Biol 32(17): 3704-3719.e3707. PubMed ID: 35896119

    Ovrebø, J. I., Bradley-Gill, M. R., Zielke, N., Kim, M., Marchetti, M., Bohlen, J., Lewis, M., van Straaten, M., Moon, N. S. and Edgar, B. A. (2022). Translational control of E2f1 regulates the Drosophila cell cycle. Proc Natl Acad Sci U S A 119(4). PubMed ID: 35074910

    Tauc, H. M., Rodriguez-Fernandez, I. A., Hackney, J. A., Pawlak, M., Ronnen Oron, T., Korzelius, J., Moussa, H. F., Chaudhuri, S., Modrusan, Z., Edgar, B. A. and Jasper, H. (2021). Age-related changes in polycomb gene regulation disrupt lineage fidelity in intestinal stem cells. Elife 10. PubMed ID: 33724181

    Tamamouna, V., Rahman, M. M., Petersson, M., Charalambous, I., Kux, K., Mainor, H., Bolender, V., Isbilir, B., Edgar, B. A. and Pitsouli, C. (2021). Remodelling of oxygen-transporting tracheoles drives intestinal regeneration and tumorigenesis in Drosophila. Nat Cell Biol 23(5): 497-510. PubMed ID: 33972730

    Zhang, P., Katzaroff, A. J., Buttitta, L. A., Ma, Y., Jiang, H., Nickerson, D. W., Ovrebo, J. I. and Edgar, B. A. (2021). The Kruppel-like factor Cabut has cell cycle regulatory properties similar to E2F1. Proc Natl Acad Sci U S A 118(7). PubMed ID: 33558234

    Ahmed, S. M. H., Maldera, J. A., Krunic, D., Paiva-Silva, G. O., Penalva, C., Teleman, A. A. and Edgar, B. A. (2020). Fitness trade-offs incurred by ovary-to-gut steroid signalling in Drosophila. Nature. PubMed ID: 32641829

    Kwon, Y. V., Zhao, B., Xu, C., Lee, J., Chen, C. L., Vinayagam, A., Edgar, B. A. and Perrimon, N. (2019). The role of translationally controlled tumor protein in proliferation of Drosophila intestinal stem cells. Proc Natl Acad Sci U S A. PubMed ID: 31843907

    Patel, P. H., Penalva, C., Kardorff, M., Roca, M., Pavlovic, B., Thiel, A., Teleman, A. A. and Edgar, B. A. (2019). Damage sensing by a Nox-Ask1-MKK3-p38 signaling pathway mediates regeneration in the adult Drosophila midgut. Nat Commun 10(1): 4365. PubMed ID: 31554796

    Zhang, P., Holowatyj, A. N., Ulrich, C. M. and Edgar, B. A. (2019). Tumor suppressive autophagy in intestinal stem cells controls gut homeostasis. Autophagy: 1-3. PubMed ID: 31213134

    Garcia Del Arco, A., Edgar, B. A. and Erhardt, S. (2018). In vivo analysis of centromeric proteins reveals a stem cell-specific asymmetry and an essential role in differentiated, non-proliferating cells. Cell Rep 22(8): 1982-1993. PubMed ID: 29466727

  • Kevin Edwards Dept. of Biological Sciences, Illinois State University, Normal
    Javeed, N., Tardi, N. J., Maher, M., Singari, S. and Edwards, K. A. (2015). Controlled expression of Drosophila homeobox loci using the Hostile takeover system. Dev Dyn 244: 808-825. PubMed ID: 26017699

    Singari, S., Javeed, N., Tardi, N. J., Marada, S., Carlson, J. C., Kirk, S., Thorn, J. M. and Edwards, K. A. (2013). Inducible Protein Traps with Dominant Phenotypes for Functional Analysis of the Drosophila Genome. Genetics. PubMed ID: 24172131

    Tardi, N. J., Cook, M. E. and Edwards, K. A. (2012). Rapid phenotypic analysis of uncoated Drosophila samples with low-vacuum scanning electron microscopy. Fly (Austin) 6: 184-192. PubMed ID: 22722327

    Tilley, D. M., Evans, C. R., Larson, T. M., Edwards, K. A. and Friesen, J. A. (2008). Identification and characterization of the nuclear isoform of Drosophila melanogaster CTP:phosphocholine cytidylyltransferase. Biochemistry 47: 11838-11846. PubMed ID: 18922025

  • Boris Egger Department of Biology, University of Fribourg, Switzerland
    Baccino-Calace, M., Prieto, D., Cantera, R. and Egger, B. (2020). Compartment and cell-type specific hypoxia responses in the developing Drosophila brain. Biol Open 9(8). PubMed ID: 32816692

    Guillermin, O., Perruchoud, B., Sprecher, S. G. and Egger, B. (2015). Characterization of Tailless functions during drosophila optic lobe formation. Dev Biol. PubMed ID: 26111972

    Perruchoud, B. and Egger, B. (2014). Immunofluorescent labeling of neural stem cells in the Drosophila optic lobe. Methods Mol Biol 1082: 71-78. PubMed ID: 24048927

    Egger, B., van Giesen, L., Moraru, M. and Sprecher, S. G. (2013). In vitro imaging of primary neural cell culture from Drosophila. Nat Protoc 8: 958-965. PubMed ID: 23598446

  • Thomas Eickbush Department of Biology, University of Rochester
    Zhou, J., Sackton, T. B., Martinsen, L., Lemos, B., Eickbush, T. H. and Hartl, D. L. (2012). Y chromosome mediates ribosomal DNA silencing and modulates the chromatin state in Drosophila. Proc Natl Acad Sci U S A 109: 9941-9946. PubMed ID: 22665801

    Eickbush, M. T. and Eickbush, T. H. (2011). Retrotransposition of R2 elements in somatic nuclei during the early development of Drosophila. Mob DNA 2: 11. PubMed ID: 21958913

  • Michael Eisen University of California, Berkeley
    Eisen, M. B.

    Chandler, J. A., Innocent, L. V., Martinez, D. J., Huang, I. L., Yang, J. L., Eisen, M. B. and Ludington, W. B. (2022). Microbiome-by-ethanol interactions impact Drosophila melanogaster fitness, physiology, and behavior. iScience 25(4): 104000. PubMed ID: 35313693

    Berrocal, A., Lammers, N. C., Garcia, H. G. and Eisen, M. B. (2020). Kinetic sculpting of the seven stripes of the Drosophila even-skipped gene. Elife 9. PubMed ID: 33300492

    Bronski, M. J., Martinez, C. C., Weld, H. A. and Eisen, M. B. (2020). Whole genome Sequences of 23 Species from the Drosophila montium Species Group (Diptera: Drosophilidae): A Resource for Testing Evolutionary Hypotheses. G3 (Bethesda). PubMed ID: 32220952

    Mir, M., Stadler, M. R., Ortiz, S. A., Hannon, C. E., Harrison, M. M., Darzacq, X. and Eisen, M. B. (2018). Dynamic multifactor hubs interact transiently with sites of active transcription in Drosophila embryos. Elife 7. PubMed ID: 30589412

    Elya, C., Zhang, V., Ludington, W. B. and Eisen, M. B. (2016). Stable Host Gene Expression in the Gut of Adult Drosophila melanogaster with Different Bacterial Mono-Associations. PLoS One 11(11): e0167357. PubMed ID: 27898741

    Elya, C., Lok, T. C., Spencer, Q. E., McCausland, H., Martinez, C. C. and Eisen, M. (2018). Robust manipulation of the behavior of Drosophila melanogaster by a fungal pathogen in the laboratory. Elife 7. PubMed ID: 30047862

    Haines, J. E. and Eisen, M. B. (2018). Patterns of chromatin accessibility along the anterior-posterior axis in the early Drosophila embryo. PLoS Genet 14(5): e1007367. PubMed ID: 29727464

    Stadler, M. R., Haines, J. E. and Eisen, M. (2017). Convergence of topological domain boundaries, insulators, and polytene interbands revealed by high-resolution mapping of chromatin contacts in the early Drosophila melanogaster embryo. Elife 6. PubMed ID: 29148971

    Mir, M., Reimer, A., Haines, J. E., Li, X. Y., Stadler, M., Garcia, H., Eisen, M. B. and Darzacq, X. (2017). Dense Bicoid hubs accentuate binding along the morphogen gradient. Genes Dev 31(17): 1784-1794. PubMed ID: 28982761

  • Robert Eisenman Fred Hutchinson Cancer Research Center, Seattle, Washington
    Neves, A. and Eisenman, R. N. (2019). Distinct gene-selective roles for a network of core promoter factors in Drosophila neural stem cell identity. Biol Open 8(4). PubMed ID: 30948355

    Li, L., Anderson, S., Secombe, J. and Eisenman, R. N. (2013). The Drosophila ubiquitin-specific protease Puffyeye regulates dMyc-mediated growth. Development 140(23): 4776-4787. PubMed ID: 24173801

    >Li, L., Greer, C., Eisenman, R. N. and Secombe, J. (2010). Essential functions of the histone demethylase lid. PLoS Genet 6(11): e1001221. PubMed ID: 21124823

  • Joel Eissenberg Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine
    Hasanagic, M., van Meel, E., Luan, S., Aurora, R., Kornfeld, S. and Eissenberg, J. C. (2015). The lysosomal enzyme receptor protein (LERP) is not essential, but is implicated in lysosomal function in Drosophila melanogaster. Biol Open. PubMed ID: 26405051

    Herz, H. M., Morgan, M., Gao, X., Jackson, J., Rickels, R., Swanson, S. K., Florens, L., Washburn, M. P., Eissenberg, J. C. and Shilatifard, A. (2014). Histone H3 lysine-to-methionine mutants as a paradigm to study chromatin signaling. Science 345: 1065-1070. PubMed ID: 25170156

    Eissenberg, J. C. and Elgin, S. C. (2014). HP1a: a structural chromosomal protein regulating transcription. Trends Genet. PubMed ID: 24555990

    Hu, D., Smith, E. R., Garruss, A. S., Mohaghegh, N., Varberg, J. M., Lin, C., Jackson, J., Gao, X., Saraf, A., Florens, L., Washburn, M. P., Eissenberg, J. C. and Shilatifard, A. (2013). The Little Elongation Complex Functions at Initiation and Elongation Phases of snRNA Gene Transcription. Mol Cell. PubMed ID: 23932780

    Luan, S., Ilvarsonn, A. M. and Eissenberg, J. C. (2012). The unique GGA clathrin adaptor of Drosophila melanogaster is not essential. PLoS One 7: e45163. PubMed ID: 23028818

    Smith, E. R., Lin, C., Garrett, A. S., Thornton, J., Mohaghegh, N., Hu, D., Jackson, J., Saraf, A., Swanson, S. K., Seidel, C., Florens, L., Washburn, M. P., Eissenberg, J. C. and Shilatifard, A. (2011). The little elongation complex regulates small nuclear RNA transcription. Mol Cell 44: 954-965. PubMed ID: 22195968

    Bhadra, M. P., Horikoshi, N., Pushpavallipvalli, S. N., Sarkar, A., Bag, I., Krishnan, A., Lucchesi, J. C., Kumar, R., Yang, Q., Pandita, R. K., Singh, M., Bhadra, U., Eissenberg, J. C. and Pandita, T. K. (2012). The role of MOF in the ionizing radiation response is conserved in Drosophila melanogaster. Chromosoma 121: 79-90. PubMed ID: 22072291

  • Aki Ejima Department of Applied Biological Chemistry, Kyoto University
    Tanaka, N. K., Hirao, T., Chida, H. and Ejima, A. (2021). A sexually dimorphic olfactory neuron mediates fixed action transition during courtship ritual in Drosophila melanogaster. J Neurosci. PubMed ID: 34649953

    Tachibana, S., Touhara, K. and Ejima, A. (2015). Modification of Male Courtship Motivation by Olfactory Habituation via the GABAA Receptor in Drosophila melanogaster. PLoS One 10: e0135186. PubMed ID: 26252206

    Ejima, A. (2015). Pleiotropic actions of the male pheromone cis-vaccenyl acetate in Drosophila melanogaster. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 26105507

    Trott, A. R., Donelson, N. C., Griffith, L. C. and Ejima, A. (2012). Song choice is modulated by female movement in Drosophila males. PLoS One 7: e46025. PubMed ID: 23049926

  • Felice Elefant Department of Biology, Drexel University, Philadelphia
    Bhatnagar, A., Parmar, V., Barbieri, N., Bearoff, F., Elefant, F. and Kortagere, S. (2023). Novel EAAT2 activators improve motor and cognitive impairment in a transgenic model of Huntington's disease. Front Behav Neurosci 17: 1176777. PubMed ID: 37351153

    Bhatnagar, A., Krick, K., Karisetty, B. C., Armour, E. M., Heller, E. A. and Elefant, F. (2023). Tip60's Novel RNA-Binding Function Modulates Alternative Splicing of Pre-mRNA Targets Implicated in Alzheimer's Disease. J Neurosci 43(13): 2398-2423. PubMed ID: 36849418

    Beaver, M., Bhatnagar, A., Panikker, P., Zhang, H., Snook, R., Parmar, V., Vijayakumar, G., Betini, N., Akhter, S. and Elefant, F. (2020). Disruption of Tip60 HAT mediated neural histone acetylation homeostasis is an early common event in neurodegenerative diseases. Sci Rep 10(1): 18265. PubMed ID: 33106538

    Zhang, H., Karisetty, B. C., Bhatnagar, A., Armour, E. M., Beaver, M., Roach, T. V., Mortazavi, S., Mandloi, S. and Elefant, F. (2020). Tip60 protects against amyloid-&betal-induced transcriptomic alterations via different modes of action in early versus late stages of neurodegeneration. Mol Cell Neurosci 109: 103570. PubMed ID: 33160016

    Panikker, P., Xu, S. J., Zhang, H., Sarthi, J., Beaver, M., Sheth, A., Akhter, S. and Elefant, F. (2018). Restoring Tip60 HAT/HDAC2 balance in the neurodegenerative brain relieves epigenetic transcriptional repression and reinstates cognition. J Neurosci. PubMed ID: 29654189

    Xu, S., Panikker, P., Iqbal, S. and Elefant, F. (2016). Tip60 HAT action mediates environmental enrichment induced cognitive restoration. PLoS One 11: e0159623. PubMed ID: 27454757

    Xu, S. and Elefant, F. (2015). Tip off the HAT- Epigenetic control of learning and memory by Drosophila Tip60. Fly (Austin) 9: 22-28. PubMed ID: 26327426

    Xu, S., Wilf, R., Menon, T., Panikker, P., Sarthi, J. and Elefant, F. (2014). Epigenetic control of learning and memory in Drosophila by Tip60 HAT action. Genetics. PubMed ID: 25326235

    Pirooznia, S. K., Reube, W., Elefant, F. (2013) Increasing Tip60 HAT Levels Rescues Axonal Transport Defects and Associated Behavioral Phenotypes in a Drosophila Alzheimer's Disease Model. J Neurosci 33: 7535-7547. PubMed ID: 23616558

    Pirooznia, S. K., Elefant, F. (2013) A HAT for sleep?: Epigenetic regulation of sleep by Tip60 in Drosophila. Fly (Austin) 7. PubMed ID: 23572111

    Pirooznia, S. K., Chiu, K., Chan, M. T., Zimmerman, J. E. and Elefant, F. (2012). Epigenetic regulation of axonal growth of Drosophila pacemaker cells by histone acetyltransferase tip60 controls sleep. Genetics 192: 1327-1345. PubMed ID: 22982579

  • Ioannis Eleftherianos Department of Biological Sciences, George Washington University, Washington, DC
    Ozakman, Y., Raval, D. and Eleftherianos, I. (2023). Drosophila melanogaster Imd signaling interacts with insulin signaling and alters feeding rate upon parasitic nematode infection. Heliyon 9(5): e16139. PubMed ID: 37251825

    Tafesh-Edwards, G., Kalukin, A., Bunnell, D., Chtarbanova, S. and Eleftherianos, I. (2023). Temperature and sex shape Zika virus pathogenicity in the adult Brat (cheesehead) brain: A Drosophila model for virus-associated neurological diseases. iScience 26(4): 106424. PubMed ID: 37009222

    Tafesh-Edwards, G. and Eleftherianos, I. (2022). Functional role of thioester-containing proteins in the Drosophila anti-pathogen immune response. Dev Comp Immunol 139: 104578. PubMed ID: 36270515

    Ozakman, Y., Raval, D. and Eleftherianos, I. (2021). Activin and BMP Signaling Activity Affects Different Aspects of Host Anti-Nematode Immunity in Drosophila melanogaster. Front Immunol 12: 795331. PubMed ID: 35003118

    Harsh, S. and Eleftherianos, I. (2020). Tumor induction in Drosophila imaginal epithelia triggers modulation of fat body lipid droplets. Biochimie 179: 65-68. PubMed ID: 32946989

    Harsh, S., Fu, Y., Kenney, E., Han, Z. and Eleftherianos, I. (2020). Zika virus non-structural protein NS4A restricts eye growth in Drosophila through regulation of JAK/STAT signaling. Dis Model Mech. PubMed ID: 32152180

    Ozakman, Y. and Eleftherianos, I. (2019). TGF-beta signaling interferes With the Drosophila innate immune and metabolic response to parasitic nematode infection. Front Physiol 10: 716. PubMed ID: 31316388

    Harsh, S., Heryanto, C. and Eleftherianos, I. (2019). Intestinal lipid droplets as novel mediators of host-pathogen interaction in Drosophila. Biol Open. PubMed ID: 31278163

    Patrnogic, J., Heryanto, C., Ozakman, Y. and Eleftherianos, I. (2019). Transcript analysis reveals the involvement of NF-kappaB transcription factors for the activation of TGF-beta signaling in nematode-infected Drosophila. Immunogenetics. PubMed ID: 31147740

    Yadav, S. and Eleftherianos, I. (2019). Participation of the serine protease Jonah66Ci in the Drosophila anti-nematode immune response. Infect Immun. PubMed ID: 31182620

    Patrnogic, J., Castillo, J. C., Shokal, U., Yadav, S., Kenney, E., Heryanto, C., Ozakman, Y. and Eleftherianos, I. (2018). Pre-exposure to non-pathogenic bacteria does not protect Drosophila against the entomopathogenic bacterium Photorhabdus. PLoS One 13(10): e0205256. PubMed ID: 30379824

  • Sarah Elgin Washington University, St. Louis
    Wang, S. H. and Elgin, S. C. R. (2019). The impact of genetic background and cell lineage on the level and pattern of gene expression in position effect variegation. Epigenetics Chromatin 12(1): 70. PubMed ID: 31722719

    Riddle, N. C. and Elgin, S. C. R. (2018). The Drosophila dot chromosome: Where genes flourish amidst repeats. Genetics 210(3): 757-772. PubMed ID: 30401762

    Huisinga, K. L., Riddle, N. C., Leung, W., Shimonovich, S., McDaniel, S., Figueroa-Clarevega, A. and Elgin, S. C. (2015). Targeting of P element reporters to heterochromatic domains by transposable element 1360 in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 26680659

    Eissenberg, J. C. and Elgin, S. C. (2014). HP1a: a structural chromosomal protein regulating transcription. Trends Genet. PubMed ID: 24555990

    Wang, S. H., Nan, R., Accardo, M. C., Sentmanat, M., Dimitri, P. and Elgin, S. C. (2014). A Distinct Type of Heterochromatin at the Telomeric Region of the Drosophila melanogaster Y Chromosome. PLoS One 9: e86451. PubMed ID: 24475122

    Gu, T. and Elgin, S. C. (2013). Maternal Depletion of Piwi, a Component of the RNAi System, Impacts Heterochromatin Formation in Drosophila. PLoS Genet 9: e1003780. PubMed ID: 24068954

    Elgin, S. C. and Reuter, G. (2013). Position-effect variegation, heterochromatin formation, and gene silencing in Drosophila. Cold Spring Harb Perspect Biol 5. PubMed ID: 23906716

    Sentmanat, M. F. and Elgin, S. C. (2012). Ectopic assembly of heterochromatin in Drosophila melanogaster triggered by transposable elements. Proc Natl Acad Sci 109: 14104-14109. PubMed ID: 22891327

    Alekseyenko, A. A., et al. (2012). Sequence-specific targeting of dosage compensation in Drosophila favors an active chromatin context. PLoS Genet. 8(4): e1002646. PubMed Citation: 22570616

  • Chris Elliott Biology, University of York
    Petridi, S., Middleton, C. A., Ugbode, C., Fellgett, A., Covill, L. and Elliott, C. J. H. (2020). In Vivo Visual Screen for Dopaminergic Rab <--> LRRK2-G2019S Interactions in Drosophila Discriminates Rab10 from Rab3. G3 (Bethesda). PubMed ID: 32321836

    West, R. J., Furmston, R., Williams, C. A. and Elliott, C. J. (2015). Neurophysiology of Drosophila models of Parkinson's disease. Parkinsons Dis 2015: 381281. PubMed ID: 25960916

    Hindle, S. J. and Elliott, C. J. (2013). Spread of neuronal degeneration in a dopaminergic, Lrrk-G2019S model of Parkinson disease. Autophagy 9: 936-938. PubMed ID: 23529190

    Hindle, S., Afsari, F., Stark, M., Middleton, C. A., Evans, G. J., Sweeney, S. T. and Elliott, C. J. (2013). Dopaminergic expression of the Parkinsonian gene LRRK2-G2019S leads to non-autonomous visual neurodegeneration, accelerated by increased neural demands for energy. Hum Mol Genet 22: 2129-2140. PubMed ID: 23396536

    Diaper, D. C., Adachi, Y., Sutcliffe, B., Humphrey, D. M., Elliott, C. J., Stepto, A., Ludlow, Z. N., Vanden Broeck, L., Callaerts, P., Dermaut, B., Al-Chalabi, A., Shaw, C. E., Robinson, I. M. and Hirth, F. (2013). Loss and gain of Drosophila TDP-43 impair synaptic efficacy and motor control leading to age-related neurodegeneration by loss-of-function phenotypes. Hum Mol Genet 22: 1539-1557. PubMed ID: 23307927

  • Christopher E Ellison Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ
    Torosin, N. S., Golla, T. R., Lawlor, M. A., Cao, W. and Ellison, C. E. (2022). Mode and tempo of 3D genome evolution in Drosophila. Mol Biol Evol. PubMed ID: 36201625

    Stanek, T. J., Cao, W., Mehra, R. M. and Ellison, C. E. (2022). Sex-specific variation in R-loop formation in Drosophila melanogaster. PLoS Genet 18(6): e1010268. PubMed ID: 35687614

    Lawlor, M. A., Cao, W. and Ellison, C. E. (2021). A transposon expression burst accompanies the activation of Y-chromosome fertility genes during Drosophila spermatogenesis. Nat Commun 12(1): 6854. PubMed ID: 34824217

  • Greg Emery University of Montreal
    Roberto, G. M., Boutet, A., Keil, S. and Emery, G. (2023). Dual regulation of Misshapen by Tao and Rap2l promotes collective cell migration. bioRxiv. PubMed ID: 37503122

    Boutet, A., Zeledon, C. and Emery, G. (2023). ArfGAP1 regulates the endosomal sorting of guidance receptors to promote directed collective cell migration in vivo. iScience 26(8): 107467. PubMed ID: 37599820

    Zeledon, C., Sun, X., Plutoni, C. and Emery, G. (2019). The ArfGAP Drongo promotes actomyosin contractility during collective cell migration by releasing Myosin phosphatase from the trailing edge. Cell Rep 28(12): 3238-3248. PubMed ID: 31533044

    Plutoni, C., Keil, S., Zeledon, C., Delsin, L. E. A., Decelle, B., Roux, P. P., Carreno, S. and Emery, G. (2019). Misshapen coordinates protrusion restriction and actomyosin contractility during collective cell migration. Nat Commun 10(1): 3940. PubMed ID: 31477736

    Colombie, N., Choesmel-Cadamuro, V., Series, J., Emery, G., Wang, X. and Ramel, D. (2017). Non-autonomous role of Cdc42 in cell-cell communication during collective migration. Dev Biol [Epub ahead of print]. PubMed ID: 28143705

    Wang, P., Larouche, M., Normandin, K., Kachaner, D., Mehsen, H., Emery, G. and Archambault, V. (2016). Spatial Regulation of Greatwall by Cdk1 and PP2A-Tws in the Cell Cycle. Cell Cycle: 0. PubMed ID: 26761639

    Laflamme, C. and Emery, G. (2015). In vitro and in vivo characterization of the Rab11-GAP activity of Drosophila Evi5. Methods Mol Biol 1298: 187-194. PubMed ID: 25800843

    Laflamme, C., et al. (2012). Evi5 promotes collective cell migration through its Rab-GAP activity. J. Cell Biol. 198(1): 57-67. PubMed Citation: 22778279

    Assaker, G., Ramel, D., Wculek, S. K., Gonzalez-Gaitan, M. and Emery, G. (2010). Spatial restriction of receptor tyrosine kinase activity through a polarized endocytic cycle controls border cell migration. Proc Natl Acad Sci U S A 107: 22558-22563. PubMed ID: 21149700

  • Patrick Emery University of Massachusetts Medical School, Worcester
    Joshi, R., Cai, Y. D., Xia, Y., Chiu, J. C. and Emery, P. (2022). PERIOD Phosphoclusters Control Temperature Compensation of the Drosophila Circadian Clock. Front Physiol 13: 888262. PubMed ID: 35721569

    Chaturvedi, R., Stork, T., Yuan, C., Freeman, M. R. and Emery, P. (2022). Astrocytic GABA transporter controls sleep by modulating GABAergic signaling in Drosophila circadian neurons. Curr Biol. PubMed ID: 35303417

    Foley, L., Ling, J., Joshi, R., Evantal, N., Kadener, S. and Emery, P. (2019). Drosophila PSI controls circadian period and the phase of circadian behavior under temperature cycle via tim splicing. Elife 8. PubMed ID: 31702555

    Lamba, P., Foley, L. E. and Emery, P. (2018). Neural network interactions modulate CRY-dependent photoresponses in Drosophila. J Neurosci. PubMed ID: 29875268

    Chatterjee, A., Lamaze, A., De, J., Mena, W., Chelot, E., Martin, B., Hardin, P., Kadener, S., Emery, P. and Rouyer, F. (2018). Reconfiguration of a multi-oscillator network by light in the Drosophila circadian clock. Curr Biol. PubMed ID: 29910074

    Fujii, S., Emery, P. and Amrein, H. (2017). SIK3-HDAC4 signaling regulates Drosophila circadian male sex drive rhythm via modulating the DN1 clock neurons. Proc Natl Acad Sci U S A 114(32): E6669-e6677. PubMed ID: 28743754

    Zhang, Y., Lamba, P., Guo, P. and Emery, P. (2016). miR-124 regulates the phase of Drosophila circadian locomotor behavior. J Neurosci 36: 2007-2013. PubMed ID: 26865623

    Tataroglu, O., Zhao, X., Busza, A., Ling, J., O'Neill, J. S. and Emery, P. (2015). Calcium and SOL Protease Mediate Temperature Resetting of Circadian Clocks. Cell 163: 1214-1224. PubMed ID: 26590423

    Tataroglu, O. and Emery, P. (2015). The molecular ticks of the Drosophila circadian clock. Curr Opin Insect Sci 7: 51-57. PubMed ID: 26120561

    Tataroglu, O. and Emery, P. (2014). Studying circadian rhythms in Drosophila melanogaster. Methods. PubMed ID: 24412370

    Karpowicz, P., Zhang, Y., Hogenesch, J. B., Emery, P. and Perrimon, N. (2013). The circadian clock gates the intestinal stem cell regenerative state. Cell Rep 3: 996-1004. PubMed ID: 23583176

  • Kazuo Emoto Neural Morphogenesis Laboratory, National Institute of Genetics, Shizuoka, Japan
    Omamiuda-Ishikawa, N., Sakai, M. and Emoto, K. (2020). A pair of ascending neurons in the subesophageal zone mediates aversive sensory inputs-evoked backward locomotion in Drosophila larvae. PLoS Genet 16(11): e1009120. PubMed ID: 33137117

    Kitatani, Y., Tezuka, A., Hasegawa, E., Yanagi, S., Togashi, K., Tsuji, M., Kondo, S., Parrish, J. Z. and Emoto, K. (2020). Drosophila miR-87 promotes dendrite regeneration by targeting the transcriptional repressor Tramtrack69. PLoS Genet 16(8): e1008942. PubMed ID: 32764744

    Utashiro, N., Williams, C. R., Parrish, J. Z. and Emoto, K. (2018). Prior activity of olfactory receptor neurons is required for proper sensory processing and behavior in Drosophila larvae. Sci Rep 8(1): 8580. PubMed ID: 29872087

    Yoshino, J., Morikawa, R. K., Hasegawa, E. and Emoto, K. (2017). Neural circuitry that evokes escape behavior upon activation of nociceptive sensory neurons in Drosophila larvae. Curr Biol. PubMed ID: 28803873

    Yasunaga, K., Tezuka, A., Ishikawa, N., Dairyo, Y., Togashi, K., Koizumi, H. and Emoto, K. (2015). Adult Drosophila sensory neurons specify dendritic territories independently of dendritic contacts through the Wnt5-Drl signaling pathway. Genes Dev 29: 1763-1775. PubMed ID: 26302791

    Kanamori, T., Yoshino, J., Yasunaga, K., Dairyo, Y. and Emoto, K. (2015). Local endocytosis triggers dendritic thinning and pruning in Drosophila sensory neurons. Nat Commun 6: 6515. PubMed ID: 25761586

    Kanamori, T., Kanai, M. I., Dairyo, Y., Yasunaga, K. I., Morikawa, R. K. and Emoto, K. (2013). Compartmentalized Calcium Transients Trigger Dendrite Pruning in Drosophila Sensory Neurons. Science. PubMed ID: 23722427

    Sakurai, A., Koganezawa, M., Yasunaga, K., Emoto, K. and Yamamoto, D. (2013). Select interneuron clusters determine female sexual receptivity in Drosophila. Nat Commun 4: 1825. PubMed ID: 23652013

    Morikawa, R. K., Kanamori, T., Yasunaga, K. and Emoto, K. (2011). Different levels of the Tripartite motif protein, Anomalies in sensory axon patterning (Asap), regulate distinct axonal projections of Drosophila sensory neurons. Proc Natl Acad Sci U S A 108: 19389-19394. PubMed ID: 22084112

    Yasunaga, K., Kanamori, T., Morikawa, R., Suzuki, E. and Emoto, K. (2010). Dendrite reshaping of adult Drosophila sensory neurons requires matrix metalloproteinase-mediated modification of the basement membranes. Dev Cell 18: 621-632. PubMed ID: 20412776

  • Sharyn Endow Department of Cell Biology, Yale
    Do, K. K., Hoang, K. L. and Endow, S. A. (2014). The kinesin-13 KLP10A motor regulates oocyte spindle length and affects EB1 binding without altering microtubule growth rates. Biol Open. PubMed ID: 24907370

    Endow, S. A. and Hallen, M. A. (2011). Anastral spindle assembly and gamma-tubulin in Drosophila oocytes. BMC Cell Biol 12: 1. PubMed ID: 21208439

    Liang, Z. Y., Hallen, M. A. and Endow, S. A. (2009). Mature Drosophila meiosis I spindles comprise microtubules of mixed polarity. Curr Biol 19: 163-168. PubMed ID: 19167226

  • Ylva Engström Stockholm University
    Zhao, Y., Lindberg, B. G., Esfahani, S. S., Tang, X., Piazza, S. and Engstrom, Y. (2021). Stop codon readthrough alters the activity of a POU/Oct transcription factor during Drosophila development. BMC Biol 19(1): 185. PubMed ID: 34479564

    Duan, J., Zhao, Y., Li, H., Habernig, L., Gordon, M. D., Miao, X., Engstrom, Y. and Buttner, S. (2020). Bab2 Functions as an Ecdysone-Responsive Transcriptional Repressor during Drosophila Development. Cell Rep 32(4): 107972. PubMed ID: 32726635

    Tang, X., Zhao, Y., Buchon, N. and Engstrom, Y. (2018). The POU/Oct transcription factor Nubbin controls the balance of intestinal stem cell maintenance and differentiation by isoform-specific regulation. Stem Cell Reports 10(5):1565-1578. PubMed ID: 29681543

    Lindberg, B. G., Tang, X., Dantoft, W., Gohel, P., Seyedoleslami Esfahani, S., Lindvall, J. M. and Engstrom, Y. (2018). Nubbin isoform antagonism governs Drosophila intestinal immune homeostasis. PLoS Pathog 14(3): e1006936. PubMed ID: 29499056

    Dantoft, W., Lundin, D., Esfahani, S.S. and Engström, Y. (2016). The POU/Oct transcription factor Pdm1/nub is necessary for a beneficial gut microbiota and normal lifespan of Drosophila. J Innate Immun 8: 412-426. PubMed ID: 27231014

    Dantoft, W., Davis, M. M., Lindvall, J. M., Tang, X., Uvell, H., Junell, A., Beskow, A. and Engstrom, Y. (2013). The Oct1 homolog Nubbin is a repressor of NF-kappaB-dependent immune gene expression that increases the tolerance to the gut microbiota. BMC Biol 11: 99. PubMed ID: 24010524

    Davis, M. M. and Engstrom, Y. (2012). Immune response in the barrier epithelia: lessons from the fruit fly Drosophila melanogaster. J Innate Immun 4: 273-283. PubMed ID: 22237424

    Davis, M. M., Alvarez, F. J., Ryman, K., Holm, A. A., Ljungdahl, P. O. and Engstrom, Y. (2011). Wild-type Drosophila melanogaster as a model host to analyze nitrogen source dependent virulence of Candida albicans. PLoS One 6: e27434. PubMed ID: 22110651

    Esfahani, S. S. and Engstrom, Y. (2011). Activation of an innate immune response in large numbers of permeabilized Drosophila embryos. Dev Comp Immunol 35: 263-266. PubMed ID: 21075135

  • Anne Ephrussi European Molecular Biology Laboratory, Heidelberg
    Gaspar, I., Phea, L. J., McClintock, M. A., Heber, S., Bullock, S. L. and Ephrussi, A. (2023). An RNA-based feed-forward mechanism ensures motor switching in oskar mRNA transport. J Cell Biol 222(7). PubMed ID: 37213090

    Cassella, L. and Ephrussi, A. (2022). Subcellular spatial transcriptomics identifies three mechanistically different classes of localizing RNAs. Nat Commun 13(1): 6355. PubMed ID: 36289223

    Bose, M., Lampe, M., Mahamid, J. and Ephrussi, A. (2022). Liquid-to-solid phase transition of oskar ribonucleoprotein granules is essential for their function in Drosophila embryonic development. Cell 185(8): 1308-1324.e1323. PubMed ID: 35325593

    Dimitrova-Paternoga, L., Jagtap, P. K. A., Cyrklaff, A., Vaishali, Lapouge, K., Sehr, P., Perez, K., Heber, S., Low, C., Hennig, J. and Ephrussi, A. (2021). Molecular basis of mRNA transport by a kinesin-1-atypical tropomyosin complex. Genes Dev 35(13-14): 976-991. PubMed ID: 34140355

    Vaishali, Dimitrova-Paternoga, L., Haubrich, K., Sun, M., Ephrussi, A. and Hennig, J. (2021). Validation and classification of RNA binding proteins identified by mRNA interactome capture. Rna. PubMed ID: 34215685

    Hampoelz, B., Schwarz, A., Ronchi, P., Bragulat-Teixidor, H., Tischer, C., Gaspar, I., Ephrussi, A., Schwab, Y. and Beck, M. (2019). Nuclear pores assemble from Nucleoporin condensates during oogenesis. Cell 179(3): 671-686.e617. PubMed ID: 31626769

    Obrdlik, A., Lin, G., Haberman, N., Ule, J. and Ephrussi, A. (2019). The transcriptome-wide landscape and modalities of EJC binding in adult Drosophila. Cell Rep 28(5): 1219-1236.e1211. PubMed ID: 31365866

    Durdevic, Z. and Ephrussi, A. (2019). Germ cell lineage homeostasis in Drosophila requires the Vasa RNA helicase. Genetics. PubMed ID: 31484689

    Heber, S., Gaspar, I., Tants, J. N., Gunther, J., Moya, S. M. F., Janowski, R., Ephrussi, A., Sattler, M. and Niessing, D. (2019). Staufen2-mediated RNA recognition and localization requires combinatorial action of multiple domains. Nat Commun 10(1): 1659. PubMed ID: 30971701

    Durdevic, Z., Pillai, R. S. and Ephrussi, A. (2018). Transposon silencing in the Drosophila female germline is essential for genome stability in progeny embryos. Life Sci Alliance 1(5): e201800179. PubMed ID: 30456388

  • Miklos Erdelyi Biological Research Centre, Hungarian Academy of Sciences, Szeged
    Jankovics, F., Bence, M., Sinka, R., Farago, A., Bodai, L., Pettko-Szandtner, A., Ibrahim, K., Takacs, Z., Szarka-Kovacs, A. B. and Erdelyi, M. (2018). Drosophila small ovary gene is required for transposon silencing and heterochromatin organisation and ensures germline stem cell maintenance and differentiation. Development. PubMed ID: 30389853

    Kristo, I., Bajusz, C., Borsos, B. N., Pankotai, T., Dopie, J., Jankovics, F., Vartiainen, M. K., Erdelyi, M. and Vilmos, P. (2017). The actin binding cytoskeletal protein Moesin is involved in nuclear mRNA export. Biochim Biophys Acta 1864(10): 1589-1604. PubMed ID: 28554770

    Takács, Z., Jankovics, F., Vilmos, P., Lénárt, P., Röper, K. and Erdélyi, M. (2017). The spectraplakin short stop is an essential microtubule regulator involved in epithelial closure in Drosophila. J Cell Sci [Epub ahead of print]. PubMed ID: 28062848

    Vilmos, P., Bujna, A., Szuperak, M., Havelda, Z., Varallyay, E., Szabad, J., Kucerova, L., Somogyi, K., Kristo, I., Lukacsovich, T., Jankovics, F., Henn, L. and Erdelyi, M. (2013). The miR-282 Drosophila melanogaster microRNA Gene Regulates Viability, Longevity, and Egg Production. Genetics. PubMed ID: 23852386

    Muha, V., Horvath, A., Bekesi, A., Pukancsik, M., Hodoscsek, B., Merenyi, G., Rona, G., Batki, J., Kiss, I., Jankovics, F., Vilmos, P., Erdelyi, M. and Vertessy, B. G. (2012). Uracil-containing DNA in Drosophila: stability, stage-specific accumulation, and developmental involvement. PLoS Genet 8: e1002738. PubMed ID: 22685418

    Banreti, A., Lukacsovich, T., Csikos, G., Erdelyi, M. and Sass, M. (2012). PP2A regulates autophagy in two alternative ways in Drosophila. Autophagy 8: 623-636. PubMed ID: 22330894

    Jankovics, F., Henn, L., Bujna, A., Vilmos, P., Kiss, N. and Erdelyi, M. (2011). A functional genomic screen combined with time-lapse microscopy uncovers a novel set of genes involved in dorsal closure of Drosophila embryos. PLoS One 6: e22229. PubMed ID: 21799798

  • Sylvia Erhardt Universität Heidelberg
    Demirdizen, E., Spiller-Becker, M., Fortsch, A., Wilhelm, A., Corless, S., Bade, D., Bergner, A., Hessling, B. and Erhardt, S. (2019). Localization of Drosophila CENP-A to non-centromeric sites depends on the NuRD complex. Nucleic Acids Res. PubMed ID: 31713634

    Pauleau, A. L., Bergner, A., Kajtez, J. and Erhardt, S. (2019). The checkpoint protein Zw10 connects CAL1-dependent CENP-A centromeric loading and mitosis duration in Drosophila cells. PLoS Genet 15(9): e1008380. PubMed ID: 31553715

    Garcia Del Arco, A., Edgar, B. A. and Erhardt, S. (2018). In vivo analysis of centromeric proteins reveals a stem cell-specific asymmetry and an essential role in differentiated, non-proliferating cells. Cell Rep 22(8): 1982-1993. PubMed ID: 29466727

    Rosic, S., Kohler, F. and Erhardt, S. (2014). Repetitive centromeric satellite RNA is essential for kinetochore formation and cell division. J Cell Biol. PubMed ID: 25365994
    Mathew, V., Pauleau, A. L., Steffen, N., Bergner, A., Becker, P. B. and Erhardt, S. (2014). The Histone-Fold Protein CHRAC14 Influences Chromatin Composition in Response to DNA Damage. Cell Rep. PubMed ID: 24703848

    Bade, D., Pauleau, A. L., Wendler, A. and Erhardt, S. (2014). The E3 Ligase CUL3/RDX Controls Centromere Maintenance by Ubiquitylating and Stabilizing CENP-A in a CAL1-Dependent Manner. Dev Cell 28: 508-519. PubMed ID: 24636256

    Bade, D., Pauleau, A. L., Wendler, A. and Erhardt, S. (2014). The E3 Ligase CUL3/RDX Controls Centromere Maintenance by Ubiquitylating and Stabilizing CENP-A in a CAL1-Dependent Manner. Dev Cell 28: 508-519. PubMed ID: 24636256

    Heun, P., Erhardt, S., Blower, M. D., Weiss, S., Skora, A. D. and Karpen, G. H. (2006). Mislocalization of the Drosophila centromere-specific histone CID promotes formation of functional ectopic kinetochores. Dev Cell 10: 303-315. PubMed ID: 16516834

  • James Erickson Biological Sciences, Texas A&M, College Station Texas
    Erickson, J. W. (2016). Primary Sex Determination in Drosophila melanogaster Does Not Rely on the Male-Specific Lethal Complex. Genetics 202(2): 541-549. PubMed ID: 26614741

    Salz, H. K. and Erickson, J. W. (2010). Sex determination in Drosophila: The view from the top. Fly (Austin) 4(1): 60-70. PubMed ID: 20160499

    Gonzalez, A. N., Lu, H. and Erickson, J. W. (2008). A shared enhancer controls a temporal switch between promoters during Drosophila primary sex determination. Proc Natl Acad Sci U S A 105(47): 18436-18441. PubMed ID: 19011108

  • Albert Erives Department of Biology, University of Iowa, Iowa City
    Stroebele, E. and Erives, A. (2016). Integration of orthogonal signaling by the Notch and Dpp pathways in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 26975664

    Rice, C., Beekman, D., Liu, L. and Erives, A. (2015). The nature, extent, and consequences of genetic variation in the opa repeats of Notch in Drosophila. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26362765

    Brittain, A., Stroebele, E. and Erives, A. (2014). Microsatellite repeat instability fuels evolution of embryonic enhancers in hawaiian Drosophila. PLoS One 9: e101177. PubMed ID: 24978198

    Crocker, J., Erives, A. (2013) A Schnurri/Mad/Medea complex attenuates the dorsal-twist gradient readout at vnd. Dev Biol. PubMed ID: 23499655

    Crocker, J., Potter, N. and Erives, A. (2010). Dynamic evolution of precise regulatory encodings creates the clustered site signature of enhancers. Nat Commun 1: 99. PubMed ID: 20981027

  • Ted Erclik Department of Cell and Systems Biology, University of Toronto,
    Valentino, P. and Erclik, T. (2022). Spalt and Disco Define the Dorsal-Ventral Neuroepithelial Compartments of the Developing Drosophila Medulla. Genetics. PubMed ID: 36135799

    Islam, I. M. and Erclik, T. (2022). Imp and Syp mediated temporal patterning of neural stem cells in the developing Drosophila CNS. Genetics 222(1). PubMed ID: 35881070

    Islam, I. M., Ng, J., Valentino, P. and Erclik, T. (2021). Identification of enhancers that drive the spatially restricted expression of Vsx1 and Rx in the outer proliferation center of the developing Drosophila optic lobe. Genome 64(2): 109-117. PubMed ID: 33054400

  • Carlos Estella CBM Severo Ochoa, Universidad Autonoma de Madrid
    Ruiz-Losada, M., González, R., Peropadre, A., Gil-Galvez, A., Tena, J. J., Baonza, A. and Estella, C. (2021). Coordination between cell proliferation and apoptosis after DNA damage in Drosophila. Cell Death Differ. PubMed ID: 34824391

    Ruiz-Losada, M., Perez-Reyes, C. and Estella, C. (2021). Role of the Forkhead Transcription Factors Fd4 and Fd5 During Drosophila Leg Development. Front Cell Dev Biol 9: 723927. PubMed ID: 34409041

    Blom-Dahl, D., Cordoba, S., Gabilondo, H., Carr-Baena, P., Diaz-Benjumea, F. J. and Estella, C. (2020). In vivo analysis of the evolutionary conserved BTD-box domain of Sp1 and Btd during Drosophila development. Dev Biol. PubMed ID: 32738261

    Cordoba, S. and Estella, C. (2018). The transcription factor Dysfusion promotes fold and joint morphogenesis through regulation of Rho1. PLoS Genet 14(8): e1007584. PubMed ID: 30080872

    Requena, D., Alvarez, J. A., Gabilondo, H., Loker, R., Mann, R. S. and Estella, C. (2017). Origins and Specification of the Drosophila Wing. Curr Biol 27(24): 3826-3836.e3825. PubMed ID: 29225023

    Cordoba, S., Requena, D., Jory, A., Saiz, A. and Estella, C. (2016). The evolutionary conserved transcription factor Sp1 controls appendage growth through Notch signaling. Development 143(19):3623-3631. PubMed ID: 27578786

    Bieli, D., Kanca, O., Requena, D., Hamaratoglu, F., Gohl, D., Schedl, P., Affolter, M., Slattery, M., Muller, M. and Estella, C. (2015). Establishment of a developmental compartment requires interactions between three synergistic cis-regulatory modules. PLoS Genet 11: e1005376. PubMed ID: 26468882

    Cordoba, S. and Estella, C. (2014). The bHLH-PAS transcription factor Dysfusion regulates tarsal joint formation in response to Notch activity during Drosophila leg development. PLoS Genet 10: e1004621. PubMed ID: 25329825

    Jory, A., Estella, C., Giorgianni, M. W., Slattery, M., Laverty, T. R., Rubin, G. M. and Mann, R. S. (2012). A survey of 6,300 genomic fragments for cis-regulatory activity in the imaginal discs of Drosophila melanogaster. Cell Rep 2: 1014-1024. PubMed ID: 23063361

    Estella, C. and Mann, R. S. (2010). Non-redundant selector and growth-promoting functions of two sister genes, buttonhead and Sp1, in Drosophila leg development. PLoS Genet 6: e1001001. PubMed ID: 20585625

  • Maksim Erokhin Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia.
    Chetverina, D. A., Gorbenko, F. V., Lomaev, D. V., Georgiev, P. G. and Erokhin, M. M. (2022). Recruitment to Chromatin of (GA)n-Associated Factors GAF and Psq in the Transgenic Model System Depends on the Presence of Architectural Protein Binding Sites. Dokl Biochem Biophys 506(1): 210-214. PubMed ID: 36303054

    Mazina, M. Y., Kovalenko, E. V., Evdokimova, A. A., Erokhin, M., Chetverina, D. and Vorobyeva, N. E. (2022). RNA Polymerase II "Pause" Prepares Promoters for Upcoming Transcription during Drosophila Development. Int J Mol Sci 23(18). PubMed ID: 36142573

    Erokhin, M. M., Shidlovskii, Y. V., Lomaev, D. V., Georgiev, P. G. and Chetverina, D. A. (2021). Sfmbt Co-purifies with Hangover and SWI/SNF-Remodelers in Drosophila melanogaster. Dokl Biochem Biophys 500(1): 304-307. PubMed ID: 34697732

    Vorobyeva, N. E., Erokhin, M., Chetverina, D., Krasnov, A. N. and Mazina, M. Y. (2021). Su(Hw) primes 66D and 7F Drosophila chorion genes loci for amplification through chromatin decondensation. Sci Rep 11(1): 16963. PubMed ID: 34417521

  • Pat Estes Department of Genetics, North Carolina State University
    Long, S. K., Fulkerson, E., Breese, R., Hernandez, G., Davis, C., Melton, M. A., Chandran, R. R., Butler, N., Jiang, L. and Estes, P. (2014). A Comparison of Midline and Tracheal Gene Regulation during Drosophila Development. PLoS One 9: e85518. PubMed ID: 24465586

    Zhang, Y., Wheatley, R., Fulkerson, E., Tapp, A. and Estes, P. A. (2011). Mastermind mutations generate a unique constellation of midline cells within the Drosophila CNS. PLoS One 6(10): e26197. PubMed ID: 22046261

  • Beatriz Estrada Universidad Pablo de Olavide, Sevilla, Andalucia, Spain
    Perez-Moreno, J. J., Santa-Cruz Mateos, C., Martin-Bermudo, M. D. and Estrada, B. (2021). LanB1 Cooperates With Kon-Tiki During Embryonic Muscle Migration in Drosophila. Front Cell Dev Biol 9: 749723. PubMed ID: 35047493

    Dai, J., Estrada, B., Jacobs, S., Sanchez-Sanchez, B. J., Tang, J., Ma, M., Magadan-Corpas, P., Pastor-Pareja, J. C. and Martin-Bermudo, M. D. (2018). Dissection of Nidogen function in Drosophila reveals tissue-specific mechanisms of basement membrane assembly. PLoS Genet 14(9): e1007483. PubMed ID: 30260959

    Molnar, C., Estrada, B. and de Celis, J. F. (2018). Tay bridge and extracellular-regulated kinase activity are required for motoneuron function in the Drosophila neural system. Genes Brain Behav 17(8): e12470. PubMed ID: 29524312

  • Iwan Evans Dept. of Infection, Immunity and Cardiovascular Disease and the Bateson Centre, University of Sheffield, U. K.
    Tardy, O. R., Armitage, E. L., Prince, L. R. and Evans, I. R. (2021). The Epidermal Growth Factor Ligand Spitz Modulates Macrophage Efferocytosis, Wound Responses and Migration Dynamics During Drosophila Embryogenesis. Front Cell Dev Biol 9: 636024. PubMed ID: 33898424

    Coates, J. A., Brooks, E., Brittle, A. L., Armitage, E. L., Zeidler, M. P. and Evans, I. R. (2021). Identification of functionally distinct macrophage subpopulations in Drosophila. Elife 10. PubMed ID: 33885361

    Tardy, O. R., Armitage, E. L., Prince, L. R. and Evans, I. R. (2021). The Epidermal Growth Factor Ligand Spitz Modulates Macrophage Efferocytosis, Wound Responses and Migration Dynamics During Drosophila Embryogenesis. Front Cell Dev Biol 9: 636024. PubMed ID: 33898424

    Armitage, E. L., Roddie, H. G. and Evans, I. R. (2020). Overexposure to apoptosis via disrupted glial specification perturbs Drosophila macrophage function and reveals roles of the CNS during injury. Cell Death Dis 11(8): 627. PubMed ID: 32796812

    Davidson, A. J., Millard, T. H., Evans, I. R. and Wood, W. (2019). Ena orchestrates remodelling within the actin cytoskeleton to drive robust Drosophila macrophage chemotaxis. J Cell Sci 132(5). PubMed ID: 30718364

  • Tim Evans Department of Biological Sciences, University of Arkansas, Fayetteville
    Carranza, A., Howard, L. J., Brown, H. E., Ametepe, A. S. and Evans, T. A. (2023). Slit-independent guidance of longitudinal axons by Drosophila Robo3. bioRxiv. PubMed ID: 37214810

    Hauptman, G., Reichert, M. C., Abdal Rhida, M. A. and Evans, T. A. (2022). Characterization of enhancer fragments in Drosophila robo2. Fly (Austin) 16(1): 312-346. PubMed ID: 36217698

    Howard, L. J., Reichert, M. C. and Evans, T. A. (2021). The Slit-binding Ig1 domain is required for multiple axon guidance activities of Drosophila Robo2. Genesis: e23443. PubMed ID: 34411419

    Daiber, T., VanderZwan-Butler, C. J., Bashaw, G. J. and Evans, T. A. (2021). Conserved and divergent aspects of Robo receptor signaling and regulation between Drosophila Robo1 and C. elegans SAX-3. Genetics 217(3). PubMed ID: 33789352

    Brown, H. E. and Evans, T. A. (2020). Minimal structural elements required for midline repulsive signaling and regulation of Drosophila Robo1. PLoS One 15(10): e0241150. PubMed ID: 33091076

    Brown, H. E., Reichert, M. C. and Evans, T. A. (2018). In Vivo Functional Analysis of Drosophila Robo1 Fibronectin Type-III Repeats. G3 (Bethesda) 8(2): 621-630. PubMed ID: 29217730

    Howard, L. J., Brown, H. E., Wadsworth, B. C. and Evans, T. A. (2019). Midline axon guidance in the Drosophila embryonic central nervous system. Semin Cell Dev Biol 85: 13-25. PubMed ID: 29174915

  • Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia
    Zatsepina, O. G., Chuvakova, L. N., Nikitina, E. A., Rezvykh, A. P., Zakluta, A. S., Sarantseva, S. V., Surina, N. V., Ksenofontov, A. L., Baratova, L. A., Shilova, V. Y. and Evgen'ev, M. B. (2022). Genes Responsible for H2S Production and Metabolism Are Involved in Learning and Memory in Drosophila melanogaster. Biomolecules 12(6). PubMed ID: 35740876

    33829398

    Shaposhnikov, M. V., Zakluta, A. S., Zemskaya, N. V., Guvatova, Z. G., Shilova, V. Y., Yakovleva, D. V., Gorbunova, A. A., Koval, L. A., Ulyasheva, N. S., Evgen'ev, M. B., Zatsepina, O. G. and Moskalev, A. A. (2022). Deletions of the cystathionine-beta-synthase (CBS) and cystathionine-gamma-lyase (CSE) genes, involved in the control of hydrogen sulfide biosynthesis, significantly affect lifespan and fitness components of Drosophila melanogaster. Mech Ageing Dev 203: 111656. PubMed ID: 35247392


  • John Ewer Centro Interdisciplinario de Neurociencia de Valparaíso, Chile
    Palacios-Munoz, A., de Paula Moreira, D., Silva, V., Garcia, I. E., Aboitiz, F., Zarrei, M., Campos, G., Rennie, O., Howe, J. L., Anagnostou, E., Ambrozewic, P., Scherer, S. W., Passos-Bueno, M. R. and Ewer, J. (2022). Mutations in trpgamma, the homologue of TRPC6 autism candidate gene, causes autism-like behavioral deficits in Drosophila. Mol Psychiatry. PubMed ID: 35501408

    Silva, V., Palacios-Munoz, A., Volonte, M., Frenkel, L., Ewer, J. and Ons, S. (2021). Orcokinin neuropeptides regulate reproduction in the fruit fly, Drosophila melanogaster. Insect Biochem Mol Biol 139: 103676. PubMed ID: 34742859

    Mark, B., Bustos-Gonzalez, L., Cascallares, G., Conejera, F. and Ewer, J. (2021). The circadian clock gates Drosophila adult emergence by controlling the timecourse of metamorphosis. Proc Natl Acad Sci U S A 118(27). PubMed ID: 34183412

    Mark, B., Bustos-Gonzalez, L., Cascallares, G., Conejera, F. and Ewer, J. (2021) Silva, V., Palacios-Munoz, A., Okray, Z., Adair, K. L., Waddell, S., Douglas, A. E. and Ewer, J. (2020). The impact of the gut microbiome on memory and sleep in Drosophila. J Exp Biol. PubMed ID: 33376141

    Flaven-Pouchon, J., Alvarez, J. V., Rojas, C. and Ewer, J. (2020). The tanning hormone, bursicon, does not act directly on the epidermis to tan the Drosophila exoskeleton. BMC Biol 18(1): 17. PubMed ID: 32075655

    Palacios-Munoz, A. and Ewer, J. (2018). Calcium and cAMP directly modulate the speed of the Drosophila circadian clock. PLoS Genet 14(6): e1007433. PubMed ID: 29879123

    Selcho, M., Millan, C., Palacios-Munoz, A., Ruf, F., Ubillo, L., Chen, J., Bergmann, G., Ito, C., Silva, V., Wegener, C. and Ewer, J. (2017). Central and peripheral clocks are coupled by a neuropeptide pathway in Drosophila. Nat Commun 8: 15563. PubMed ID: 28555616

    Mena, W., Diegelmann, S., Wegener, C. and Ewer, J. (2016). Stereotyped responses of Drosophila peptidergic neuronal ensemble depend on downstream neuromodulators. Elife 5. PubMed ID: 27976997

    Diao, F., Mena, W., Shi, J., Park, D., Diao, F., Taghert, P., Ewer, J. and White, B. H. (2015). The Splice Isoforms of the Drosophila Ecdysis Triggering Hormone Receptor Have Developmentally Distinct Roles. Genetics. PubMed ID: 26534952

    Kruger, E., Mena, W., Lahr, E. C., Johnson, E. C. and Ewer, J. (2015). Genetic analysis of Eclosion hormone action during Drosophila larval ecdysis. Development. PubMed ID: 26395475

    Diao, F., Ironfield, H., Luan, H., Diao, F., Shropshire, W. C., Ewer, J., Marr, E., Potter, C. J., Landgraf, M. and White, B. H. (2015). Plug-and-play genetic access to drosophila cell types using exchangeable exon cassettes. Cell Rep 10: 1410-1421. PubMed ID: 25732830

  • Andrew Ewing Department of Chemistry and Molecular Biology, University of Gothenburg, Sweden
    Philipsen, M., Gu, C. and Ewing, A. G. (2020). Imaging Mass Spectrometry Shows That Zinc Deficiency Leads to Lipids Changes in Drosophila Brain Similar to Cognitive Impairing Drugs. Chembiochem. PubMed ID: 32402134

    Phan, N. T., Fletcher, J. S. and Ewing, A. G. (2015). Lipid structural effects of oral administration of methylphenidate in Drosophila brain by secondary ion mass spectrometry imaging. Anal Chem 87: 4063-4071. PubMed ID: 25856152

    Berglund, E. C., Makos, M. A., Keighron, J. D., Phan, N. T., Heien, M. L. and Ewing, A. G. (2013). Oral Administration of Methylphenidate Blocks the Effect of Cocaine on Uptake at the Drosophila Dopamine Transporter. ACS Chem Neurosci. PubMed ID: 23402315

    Berglund, C., Kuklinski, N., Karagunduz, E., Ucar, K., Hanrieder, J. and Ewing, A. G. (2013). Freeze-Drying as Sample Preparation for Micellar Electrokinetic Capillary Chromatography - Electrochemical Separations of Neurochemicals in Drosophila Brains. Anal Chem. PubMed ID: 23387977

    Kuklinski, N. J., Berglund, E. C., Engelbrektsson, J. and Ewing, A. G. (2010). Biogenic amines in microdissected brain regions of Drosophila melanogaster measured with micellar electrokinetic capillary chromatography-electrochemical detection. Anal Chem 82: 7729-7735. PubMed ID: 20738098

  • Cassandra Extavour Department of Organismic and Evolutionary Biology, Harvard University
    Church, S. H., Munro, C., Dunn, C. W. and Extavour, C. G. (2023). The evolution of ovary-biased gene expression in Hawaiian Drosophila. PLoS Genet 19(1): e1010607. PubMed ID: 36689550

    Tarikere, S., Ylla, G. and Extavour, C. G. (2021). Distinct gene expression dynamics in germ line and somatic tissue during ovariole morphogenesis in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 34849771

    Blondel, L., Besse, S., Rivard, E. L., Ylla, G. and Extavour, C. G. (2021). Evolution of a cytoplasmic determinant: evidence for the biochemical basis of functional evolution of the novel germ line regulator oskar. Mol Biol Evol. PubMed ID: 34550378

    Kumar, T., Blondel, L. and Extavour, C. G. (2020). Topology-driven protein-protein interaction network analysis detects genetic sub-networks regulating reproductive capacity. Elife 9. PubMed ID: 32901612

    Sarikaya, D. P., Church, S. H., Lagomarsino, L. P., Magnacca, K. N., Montgomery, S. L., Price, D. K., Kaneshiro, K. Y. and Extavour, C. G. (2019). Reproductive capacity evolves in response to ecology through common changes in cell number in Hawaiian Drosophila. Curr Biol 29(11): 1877-1884. PubMed ID: 31130459

    Sarikaya, D.P. and Extavour, C.G. (2015). The Hippo pathway regulates homeostatic growth of stem cell niche precursors in the Drosophila ovary. PLoS Genet 11: e1004962. PubMed ID: 25643260

    Green, D. A. and Extavour, C. G. (2014). Insulin signalling underlies both plasticity and divergence of a reproductive trait in Drosophila. Proc Biol Sci 281: 20132673. PubMed ID: 24500165

    Sharma, P. P., Gupta, T., Schwager, E. E., Wheeler, W. C. and Extavour, C. G. (2014). Subdivision of arthropod cap-n-collar expression domains is restricted to Mandibulata. Evodevo 5: 3. PubMed ID: 24405788

    Ahuja, A. and Extavour, C. G. (2014). Patterns of molecular evolution of the germ line specification gene oskar suggest that a novel domain may contribute to functional divergence in Drosophila. Dev Genes Evol. PubMed ID: 24407548

    Green, D. A., 2nd and Extavour, C. G. (2012). Convergent evolution of a reproductive trait through distinct developmental mechanisms in Drosophila. Dev Biol 372: 120-130. PubMed ID: 23022298


  • Return to The Interactive Fly