Fly Labs and References

Tweet

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


  • Uri Abdu Life Sciences, Ben-Gurion University of the Negev
    Aharoni, S., Proskorovski-Ohayon, R., Krishnan, R. K., Yogev, Y., Wormser, O., Hadar, N., Bakhrat, A., Alshafee, I., Gombosh, M., Agam, N., Gradstein, L., Shorer, Z., Zarivach, R., Eskin-Schwartz, M., Abdu, U. and Birk, O. S. (2022). PSMC1 variant causes a novel neurological syndrome. Clin Genet 102(4): 324-332. PubMed ID: 35861243
    Krishnan, R. K., Baskar, R., Anna, B., Elia, N., Boermel, M., Bausch, A. R. and Abdu, U. (2021). Recapitulating Actin Module Organization in the Drosophila Oocyte Reveals New Roles for Bristle-Actin-Modulating Proteins. Int J Mol Sci 22(8). PubMed ID: 33924532

    Melkov, A., Baskar, R., Shachal, R., Alcalay, Y. and Abdu, U. (2019). The organization of Golgi in Drosophila bristles requires microtubule motor protein function and a properly organized microtubule array. PLoS One 14(10): e0223174. PubMed ID: 31577833

    Baskar, R., Bahkrat, A., Otani, T., Wada, H., Davidov, G., Pandey, H., Gheber, L., Zarivach, R., Hayashi, S. and Abdu, U. (2019). The plus-tip tracking and microtubule stabilizing activities of Javelin-like regulate microtubule organization and cell polarity. Febs j. PubMed ID: 31152621

    Baskar, R., Bakrhat, A. and Abdu, U. (2019). GFP-Forked, a genetic reporter for studying Drosophila oocyte polarity. Biol Open 8(1). PubMed ID: 30598482

    Melkov, A., Baskar, R., Alcalay, Y. and Abdu, U. (2016). New mode of mitochondrial transport and polarized sorting regulation by Dynein, Milton and Miro. Development [Epub ahead of print]. PubMed ID: 27707795

    Melkov, A., Simchoni, Y., Alcalay, Y. and Abdu, U. (2015). Dynamic microtubule organization and mitochondrial transport are regulated by distinct Kinesin-1 pathways. Biol Open [Epub ahead of print]. PubMed ID: 26581590

    Otani, T., Oshima, K., Kimpara, A., Takeda, M., Abdu, U. and Hayashi, S. (2015). A transport and retention mechanism for the sustained distal localization of Spn-F-IKKepsilon during Drosophila bristle elongation. Development 142: 2338-2351. PubMed ID: 26092846

    Amsalem, S., Bakrhat, A., Otani, T., Hayashi, S., Goldstein, B. and Uri, A. (2013). Drosophila oocyte polarity and cytoskeleton organization requires regulation of Ik2 activity by Spn-F and Javelin-like. Mol Cell Biol. PubMed ID: 24019068

    Marzuk, O., Peretz, G., Bakhrat, A., Abdu, U. (2013) The Drosophila CIAPIN1 homologue is required for follicle cell proliferation and survival. Dev Dyn. PubMed ID: 23444317

  • Hermann Aberle. Heinrich-Heine-Universität Düsseldorf
    Kinold, J. C., Brenner, M. and Aberle, H. (2021). Misregulation of Drosophila Sidestep Leads to Uncontrolled Wiring of the Adult Neuromuscular System and Severe Locomotion Defects. Front Neural Circuits 15: 658791. PubMed ID: 34149366

    Kinold, J. C., Pfarr, C. and Aberle, H. (2018). Sidestep-induced neuromuscular miswiring causes severe locomotion defects in Drosophila larvae. Development 145(17). PubMed ID: 30166331

    Niehues, S., Bussmann, J., Steffes, G., Erdmann, I., Kohrer, C., Sun, L., Wagner, M., Schafer, K., Wang, G., Koerdt, S. N., Stum, M., Jaiswal, S., RajBhandary, U. L., Thomas, U., Aberle, H., Burgess, R. W., Yang, X. L., Dieterich, D. and Storkebaum, E. (2015). Impaired protein translation in Drosophila models for Charcot-Marie-Tooth neuropathy caused by mutant tRNA synthetases. Nat Commun 6: 7520. PubMed ID: 26138142

    Gundner, A. L., Hahn, I., Sendscheid, O., Aberle, H. and Hoch, M. (2014). The PIKE homolog Centaurin gamma regulates developmental timing in Drosophila. PLoS One 9(5): e97332. PubMed ID: 24845618

  • Elizabeth Ables East Carolina University, Department of Biology, Greenville, NC
    Beachum, A. N., Hinnant, T. D., Williams, A. E., Powell, A. M. and Ables, E. T. (2023). Beta-importin Tnpo-SR promotes germline stem cell maintenance and oocyte differentiation in female Drosophila. Dev Biol 494: 1-12. PubMed ID: 36450333

    Finger, D. S., Williams, A. E., Holt, V. V. and Ables, E. T. (2022). Novel roles for RNA binding proteins squid, hephaesteus, and Hrb27C in Drosophila oogenesis. Dev Dyn. PubMed ID: 36308715

    Beachum, A. N., Whitehead, K. M., McDonald, S. I., Phipps, D. N., Berghout, H. E. and Ables, E. T. (2021). Orphan nuclear receptor ftz-f1 (NR5A3) promotes egg chamber survival in the Drosophila ovary. G3 (Bethesda) 11(2). PubMed ID: 33693603

    McDonald, S. I., Beachum, A. N., Hinnant, T. D., Blake, A. J., Bynum, T., Hickman, E. P., Barnes, J., Churchill, K. L., Roberts, T. S., Zangwill, D. E. and Ables, E. T. (2019). Novel cis-regulatory regions in ecdysone responsive genes are sufficient to promote gene expression in Drosophila ovarian cells. Gene Expr Patterns: 119074. PubMed ID: 31563631

    Hinnant, T. D., Alvarez, A. A. and Ables, E. T. (2017). Temporal remodeling of the cell cycle accompanies differentiation in the Drosophila germline. Dev Biol 429(1): 118-131. PubMed ID: 28711427

    Blake, A. J., Finger, D. S., Hardy, V. L. and Ables, E. T. (2017). RNAi-Based Techniques for the Analysis of Gene Function in Drosophila Germline Stem Cells. Methods Mol Biol 1622: 161-184. PubMed ID: 28674809

    Ables, E. T. and Drummond-Barbosa, D. (2017). Steroid Hormones and the Physiological Regulation of Tissue-Resident Stem Cells: Lessons from the Drosophila Ovary. Curr Stem Cell Rep 3(1): 9-18. PubMed ID: 28458991

  • Susan Abmayr Department of Anatomy & Cell Biology, The University of Kansas School of Medicine
    Soffers, J. H. M., Li, X., Saraf, A., Seidel, C. W., Florens, L., Washburn, M. P., Abmayr, S. M. and Workman, J. L. (2019). Characterization of a metazoan ADA acetyltransferase complex. Nucleic Acids Res. PubMed ID: 30715476

    Huang, F., Saraf, A., Florens, L., Kusch, T., Swanson, S. K., Szerszen, L. T., Li, G., Dutta, A., Washburn, M. P., Abmayr, S. M. and Workman, J. L. (2016). The Enok acetyltransferase complex interacts with Elg1 and negatively regulates PCNA unloading to promote the G1/S transition. Genes Dev 30: 1198-1210. PubMed ID: 27198229

    Haralalka, S. and Abmayr, S. M. (2015). Tracing myoblast fusion in Drosophila embryos by fluorescent actin probes. Methods Mol Biol 1313: 149-164. PubMed ID: 25947663

    Kumar, R.P., Dobi, K.C., Baylies, M.K. and Abmayr, S.M. (2015). Muscle cell fate choice requires the T-Box transcription factor Midline in Drosophila. Genetic [Epub ahead of print]. PubMed ID: 25614583

    Huang, F., Paulson, A., Dutta, A., Venkatesh, S., Smolle, M., Abmayr, S. M. and Workman, J. L. (2014). Histone acetyltransferase Enok regulates oocyte polarization by promoting expression of the actin nucleation factor spire. Genes Dev 28: 2750-2763. PubMed ID: 25512562

    Mohan, R. D., Workman, J. L. and Abmayr, S. M. (2014). Drosophila models reveal novel insights into mechanisms underlying neurodegeneration. Fly (Austin) 8: 148-152. PubMed ID: 25483136

    Haralalka, S., Shelton, C., Cartwright, H. N., Guo, F., Trimble, R., Kumar, R. P. and Abmayr, S. M. (2014). Live imaging provides new insights on dynamic F-Actin filopodia and differential endocytosis during myoblast fusion in Drosophila. PLoS One 9: e114126. PubMed ID: 25474591

    Mohan, R. D., Dialynas, G., Weake, V. M., Liu, J., Martin-Brown, S., Florens, L., Washburn, M. P., Workman, J. L. and Abmayr, S. M. (2014). Loss of Drosophila Ataxin-7, a SAGA subunit, reduces H2B ubiquitination and leads to neural and retinal degeneration. Genes Dev 28: 259-272. PubMed ID: 24493646

    Kaipa, B. R., Shao, H., Schafer, G., Trinkewitz, T., Groth, V., Liu, J., Beck, L., Abmayr, S. M. and Onel, S. F. (2012). Dock mediates Scar- and WASp-dependent actin polymerization through interaction with cell adhesion molecules in founder cells and fusion-competent myoblasts. J Cell Sci. PubMed ID: 22992459

    Lin, C. H., Paulson, A., Abmayr, S. M. and Workman, J. L. (2012). HP1a targets the Drosophila KDM4A demethylase to a subset of heterochromatic genes to regulate H3K36me3 levels. PLoS One 7: e39758. PubMed ID: 22761891

  • John Abrams Cell Biology, Southwest Medical University, Dallas
    Wylie, A., Jones, A. E., Das, S., Lu, W. J. and Abrams, J. M. (2022). Distinct p53 isoforms code for opposing transcriptional outcomes. Dev Cell 57(15): 1833-1846.e1836. PubMed ID: 35820415

    Chen, P., Visokay, S. and Abrams, J. M. (2020). Drosophila GFAT1 and GFAT2 enzymes encode obligate developmental functions. Fly (Austin): 1-7. PubMed ID: 32615907

    Tiwari, B., Kurtz, P., Jones, A. E., Wylie, A., Amatruda, J. F., Boggupalli, D. P., Gonsalvez, G. B. and Abrams, J. M. (2017). Retrotransposons mimic germ plasm determinants to promote transgenerational inheritance. Curr Biol 27(19): 3010-3016.e3013. PubMed ID: 28966088

    Garcia-Hughes, G., Link, N., Ghosh, A. B. and Abrams, J. M. (2015). Hid arbitrates collective cell death in the Drosophila wing. Mech Dev. PubMed ID: 26226435

    Vaisnav, M., Xing, C., Ku, H. C., Hwang, D., Stojadinovic, S., Pertsemlidis, A. and Abrams, J. M. (2014). Genome-Wide Association Analysis of Radiation Resistance in Drosophila melanogaster. PLoS One 9: e104858. PubMed ID: 25121966

    Wylie, A., Lu, W. J., D'Brot, A., Buszczak, M. and Abrams, J. M. (2014). p53 activity is selectively licensed in the Drosophila stem cell compartment. Elife 3: e01530. PubMed ID: 24618896

    Link, N., Kurtz, P., O'Neal, M., Garcia-Hughes, G. and Abrams, J. M. (2013). A p53 enhancer region regulates target genes through chromatin conformations in cis and in trans. Genes Dev 27: 2433-2438. PubMed ID: 24240233

    D'Brot, A., Chen, P., Vaishnav, M., Yuan, S., Akey, C. W. and Abrams, J. M. (2013). Tango7 directs cellular remodeling by the Drosophila apoptosome. Genes Dev 27: 1650-1655. PubMed ID: 23913920

    Chen, P., Tu, X., Akdemir, F., Chew, S. K., Rothenfluh, A. and Abrams, J. M. (2012). Effectors of alcohol-induced cell killing in Drosophila. Cell Death Differ 19: 1655-1663. PubMed ID: 22539005

    Chew, S. K., Chen, P., Link, N., Galindo, K. A., Pogue, K. and Abrams, J. M. (2009). Genome-wide silencing in Drosophila captures conserved apoptotic effectors. Nature 460: 123-127. PubMed ID: 19483676

  • Jairaj Acharya Center for Cancer Research, National Cancer Institute, Frederick, MD
    Kunduri, G., Turner-Evans, D., Konya, Y., Izumi, Y., Nagashima, K., Lockett, S., Holthuis, J., Bamba, T., Acharya, U. and Acharya, J. K. (2018). Defective cortex glia plasma membrane structure underlies light-induced epilepsy in cpes mutants. Proc Natl Acad Sci U S A 115(38): E8919-e8928. PubMed ID: 30185559

    Kunduri, G., Turner-Evans, D., Konya, Y., Izumi, Y., Nagashima, K., Lockett, S., Holthuis, J., Bamba, T., Acharya, U. and Acharya, J. K. (2018). Defective cortex glia plasma membrane structure underlies light-induced epilepsy in cpes mutants. Proc Natl Acad Sci U S A. PubMed ID: 30185559

    Kunduri, G., Yuan, C., Parthibane, V., Nyswaner, K. M., Kanwar, R., Nagashima, K., Britt, S. G., Mehta, N., Kotu, V., Porterfield, M., Tiemeyer, M., Dolph, P. J., Acharya, U. and Acharya, J. K. (2014). Phosphatidic acid phospholipase A1 mediates ER-Golgi transit of a family of G protein-coupled receptors. J Cell Biol 206: 79-95. PubMed ID: 25002678

    Rahman, M., Nirala, N. K., Singh, A., Zhu, L. J., Taguchi, K., Bamba, T., Fukusaki, E., Shaw, L. M., Lambright, D. G., Acharya, J. K. and Acharya, U. R. (2014). Drosophila Sirt2/mammalian SIRT3 deacetylates ATP synthase beta and regulates complex V activity. J Cell Biol 206: 289-305. PubMed ID: 25023514

    Yuan, C., Rao, R. P., Jesmin, N., Bamba, T., Nagashima, K., Pascual, A., Preat, T., Fukusaki, E., Acharya, U. and Acharya, J. K. (2011). CDase is a pan-ceramidase in Drosophila. Mol Biol Cell 22: 33-43. PubMed ID: 21148295

  • Takashi Adachi-Yamada Department of Life Science, Gakushuin University
    Taniguchi, K., Kokuryo, A., Imano, T., Nakagoshi, H. and Adachi-Yamada, T. (2018). Binucleation of accessory gland lobe contributes to effective ejection of seminal fluid in Drosophila melanogaster. Zoolog Sci 35(5): 446-458. PubMed ID: 30298781

    Kubo, A., Matsuka, M., Minami, R., Kimura, F., Sakata-Niitsu, R., Kokuryo, A., Taniguchi, K., Adachi-Yamada, T. and Nakagoshi, H. (2018). Nutrient conditions sensed by the reproductive organ during development optimize male fecundity in Drosophila. Genes Cells. PubMed ID: 29846027

    Takeda, K., Okumura, T., Terahata, M., Yamaguchi, M., Taniguchi, K. and Adachi-Yamada, T. (2018). Drosophila Peptide Hormones Allatostatin A and Diuretic Hormone 31 Exhibiting Complementary Gradient Distribution in Posterior Midgut Antagonistically Regulate Midgut Senescence and Adult Lifespan. Zoolog Sci 35(1): 75-85. PubMed ID: 29417892

    Taniguchi, K., Kokuryo, A., Imano, T., Minami, R., Nakagoshi, H. and Adachi-Yamada, T. (2014). Isoform-specific functions of Mud/NuMA mediate binucleation of Drosophila male accessory gland cells. BMC Dev Biol 14: 46. PubMed ID: 25527079

    Okumura, T., Takeda, K., Taniguchi, K. and Adachi-Yamada, T. (2014). betanu Integrin Inhibits Chronic and High Level Activation of JNK to Repress Senescence Phenotypes in Drosophila Adult Midgut. PLoS One 9: e89387. PubMed ID: 24586740

    Kuwamura, M., Maeda, K. and Adachi-Yamada, T. (2012). Mathematical modelling and experiments for the proliferation and differentiation of Drosophila intestinal stem cells II. J Biol Dyn 6: 267-276. PubMed ID: 22873590

    Minami, R., Wakabayashi, M., Sugimori, S., Taniguchi, K., Kokuryo, A., Imano, T., Adachi-Yamada, T., Watanabe, N. and Nakagoshi, H. (2012). The homeodomain protein defective proventriculus is essential for male accessory gland development to enhance fecundity in Drosophila. PLoS One 7: e32302. PubMed ID: 22427829

  • Michael E. Adams Cell Biology and Neuroscience, U. C. Riverside
    Lee, S. S. and Adams, M. E. (2021). Regulation of Drosophila Long-Term Courtship Memory by Ecdysis Triggering Hormone. Front Neurosci 15: 670322. PubMed ID: 33967686

    Deshpande, S. A., Meiselman, M., Hice, R. H., Arensburger, P., Rivera-Perez, C., Kim, D. H., Croft, R. L., Noriega, F. G. and Adams, M. E. (2018). Ecdysis Triggering Hormone Receptors Regulate Male Courtship Behavior via Antennal Lobe Interneurons in Drosophila. Gen Comp Endocrinol. PubMed ID: 30543770

    Kim, D. H., Kim, Y. J. and Adams, M. E. (2018). Endocrine regulation of airway clearance in Drosophila. Proc Natl Acad Sci U S A 115(7): 1535-1540. PubMed ID: 29386394

    Meiselman, M. R., Kingan, T. G. and Adams, M. E. (2018). Stress-induced reproductive arrest in Drosophila occurs through ETH deficiency-mediated suppression of oogenesis and ovulation. BMC Biol 16(1): 18. PubMed ID: 29382341

    Lee, S. S., Ding, Y., Karapetians, N., Rivera-Perez, C., Noriega, F. G. and Adams, M. E. (2017). Hormonal signaling cascade during an early-adult critical period required for courtship memory retention in Drosophila. Curr Biol 27(18): 2798-2809.e2793. PubMed ID: 28918947

    Meiselman, M., Lee, S. S., Tran, R. T., Dai, H., Ding, Y., Rivera-Perez, C., Wijesekera, T. P., Dauwalder, B., Noriega, F. G. and Adams, M. E. (2017). Endocrine network essential for reproductive success in Drosophila melanogaster. Proc Natl Acad Sci U S A 114(19): E3849-E3858. PubMed ID: 28439025

    Tong, M. Q., Hasan, M. M., Lee, S. S., Haque, M. R., Kim, D. H., Islam, M. S., Adams, M. E. and Park, B. H. (2017). OCT intensity and phase fluctuations correlated with activity-dependent neuronal calcium dynamics in the Drosophila CNS [Invited]. Biomed Opt Express 8(2): 726-735. PubMed ID: 28270979

    Cho, K. H., Daubnerova, I., Park, Y., Zitnan, D. and Adams, M. E. (2014). Secretory competence in a gateway endocrine cell conferred by the nuclear receptor betaFTZ-F1 enables stage-specific ecdysone responses throughout development in Drosophila. Dev Biol 385(2): 253-262. PubMed ID: 24247008

  • Paul Adler Department of Biology, University of Virginia, Charlottesville
    Wang, Y., Naturale, V.F. and Adler, P.N. (2017). Planar cell polarity effector Fritz interacts with Dishevelled and has multiple functions in regulating PCP. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 28258110Sobala, L.F. and Adler, P.N. (2016). The gene expression program for the formation of wing cuticle in Drosophila. PLoS Genet 12: e1006100. PubMed ID: 27232182

    Sobala, L. F., Wang, Y. and Adler, P. N. (2015). ChtVis-Tomato, a genetic reporter for in vivo visualization of chitin deposition in Drosophila. Development. PubMed ID: 26395478

    Lu, Q., Schafer, D. A. and Adler, P. N. (2015). The Drosophila planar polarity gene multiple wing hairs directly regulates the actin cytoskeleton. Development 142: 2478-2486. PubMed ID: 26153232

    Lu, Q. and Adler, P. N. (2015). The diaphanous gene of Drosophila interacts antagonistically with multiple wing hairs and plays a key role in wing hair morphogenesis. PLoS One 10: e0115623. PubMed ID: 25730111

    Wang, Y., Yan, J., Lee, H., Lu, Q. and Adler, P. N. (2014). The proteins encoded by the Drosophila Planar Polarity Effector genes inturned, fuzzy and fritz interact physically and can re-pattern the accumulation of "upstream" Planar Cell Polarity proteins. Dev Biol. PubMed ID: 25072625

    Adler, P. N., Sobala, L. F., Thom, D., Nagaraj, R. (2013) dusky-like is required to maintain the integrity and planar cell polarity of hairs during the development of the Drosophila wing. Dev Biol. PubMed ID: 23623898

    Adler, P. N. (2012). The frizzled/stan pathway and planar cell polarity in the Drosophila wing. Curr Top Dev Biol 101: 1-31. PubMed ID: 23140623

    Nagaraj, R. and Adler, P. N. (2012). Dusky-like functions as a Rab11 effector for the deposition of cuticle during Drosophila bristle development. Development 139: 906-916. PubMed ID: 22278919

    Fang, X., Lu, Q., Emoto, K. and Adler, P. N. (2010). The Drosophila Fry protein interacts with Trc and is highly mobile in vivo. BMC Dev Biol 10: 40. PubMed ID: 20406475

  • Christof Aegerter University of Zurich, Physics Institute
    Jiang, J. and Aegerter, C. M. (2023). An integrated vertex model of the mesoderm invagination during the embryonic development of Drosophila. J Theor Biol 572: 111581. PubMed ID: 37481232

    Selvaggi, L., Ackermann, M., Pasakarnis, L., Brunner, D. and Aegerter, C. M. (2021). Force measurements of Myosin II waves at the yolk surface during Drosophila dorsal closure. Biophys J. PubMed ID: 34971619

    Schluck, T., Nienhaus, U., Aegerter-Wilmsen, T. and Aegerter, C. M. (2013). Mechanical control of organ size in the development of the Drosophila wing disc. PLoS One 8: e76171. PubMed ID: 24204600

    Nienhaus, U., Aegerter-Wilmsen, T. and Aegerter, C. M. (2012). In-vivo imaging of the Drosophila wing imaginal disc over time: novel insights on growth and boundary formation. PLoS One 7: e47594. PubMed ID: 23091633

    Schluck, T. and Aegerter, C. M. (2010). Photo-elastic properties of the wing imaginal disc of Drosophila. Eur Phys J E Soft Matter 33: 111-115. PubMed ID: 20563622

  • Stein Aerts Laboratory of Computational Biology, Katholieke Universiteit Leuven, Belgium
    Kowalczyk, W., Romanelli, L., Atkins, M., Hillen, H., Bravo Gonzalez-Blas, C., Jacobs, J., Xie, J., Soheily, S., Verboven, E., Moya, I. M., Verhulst, S., de Waegeneer, M., Sansores-Garcia, L., van Huffel, L., Johnson, R. L., van Grunsven, L. A., Aerts, S. and Halder, G. (2022). Hippo signaling instructs ectopic but not normal organ growth. Science 378(6621): eabg3679. PubMed ID: 36395225

    Janssens, J., Aibar, S., Taskiran, II, Ismail, J. N., Gomez, A. E., Aughey, G., Spanier, K. I., De Rop, F. V., González-Blas, C. B., Dionne, M., Grimes, K., Quan, X. J., Papasokrati, D., Hulselmans, G., Makhzami, S., De Waegeneer, M., Christiaens, V., Southall, T. and Aerts, S. (2022). Decoding gene regulation in the fly brain. Nature 601(7894): 630-636. PubMed ID: 34987221

    Ravenscroft, T. A., Janssens, J., Lee, P. T., Tepe, B., Marcogliese, P. C., Makhzami, S., Holmes, T. C., Aerts, S. and Bellen, H. J. (2020). Drosophila voltage-gated sodium channels are only expressed in active neurons and are localized to distal axonal initial segment-like domains. J Neurosci. PubMed ID: 32928889

    Bravo Gonzalez-Blas, C., Quan, X. J., Duran-Romana, R., Taskiran, II, Koldere, D., Davie, K., Christiaens, V., Makhzami, S., Hulselmans, G., de Waegeneer, M., Mauduit, D., Poovathingal, S., Aibar, S. and Aerts, S. (2020). Identification of genomic enhancers through spatial integration of single-cell transcriptomics and epigenomics. Mol Syst Biol 16(5): e9438. PubMed ID: 32431014

    Cabasso, O., Paul, S., Dorot, O., Maor, G., Krivoruk, O., Pasmanik-Chor, M., Mirzaian, M., Ferraz, M., Aerts, J. and Horowitz, M. (2019). Drosophila melanogaster mutated in its GBA1b ortholog recapitulates Neuronopathic Gaucher disease. J Clin Med 8(9). PubMed ID: 31505865

    Davie, K., Janssens, J., Koldere, D., De Waegeneer, M., Pech, U., Kreft, L., Aibar, S., Makhzami, S., Christiaens, V., Bravo Gonzalez-Blas, C., Poovathingal, S., Hulselmans, G., Spanier, K. I., Moerman, T., Vanspauwen, B., Geurs, S., Voet, T., Lammertyn, J., Thienpont, B., Liu, S., Konstantinides, N., Fiers, M., Verstreken, P. and Aerts, S. (2018). A single-cell transcriptome atlas of the aging Drosophila brain. Cell. PubMed ID: 29909982

    Jacobs, J., Atkins, M., Davie, K., Imrichova, H., Romanelli, L., Christiaens, V., Hulselmans, G., Potier, D., Wouters, J., Taskiran, II, Paciello, G., Gonzalez-Blas, C. B., Koldere, D., Aibar, S., Halder, G. and Aerts, S. (2018). The transcription factor Grainy head primes epithelial enhancers for spatiotemporal activation by displacing nucleosomes. Nat Genet. PubMed ID: 29867222

    Pascual, J., Jacobs, J., Sansores-Garcia, L., Natarajan, M., Zeitlinger, J., Aerts, S., Halder, G. and Hamaratoglu, F. (2017). Hippo Reprograms the Transcriptional Response to Ras Signaling. Dev Cell 42(6): 667-680.e664. PubMed ID: 28950103

    Neto, M., Naval-Sanchez, M., Potier, D., Pereira, P. S., Geerts, D., Aerts, S. and Casares, F. (2017). Nuclear receptors connect progenitor transcription factors to cell cycle control. Sci Rep 7(1): 4845. PubMed ID: 28687780

    Atkins, M., Potier, D., Romanelli, L., Jacobs, J., Mach, J., Hamaratoglu, F., Aerts, S. and Halder, G. (2016). An ectopic network of transcription factors regulated by Hippo signaling drives growth and invasion of a malignant tumor model. Curr Biol [Epub ahead of print]. PubMed ID: 27476594

  • Markus Affolter Biozentrum, University of Basel
    Bauer, M., Aguilar, G., Wharton, K. A., Matsuda, S. and Affolter, M. (2023). Heterodimerization-dependent secretion of bone morphogenetic proteins in Drosophila. Dev Cell. PubMed ID: 37054707

    Lepeta, K., Roubinet, C., Bauer, M., Vigano, M. A., Aguilar, G., Kanca, O., Ochoa-Espinosa, A., Bieli, D., Cabernard, C., Caussinus, E. and Affolter, M. (2022). Engineered kinases as a tool for phosphorylation of selected targets in vivo. J Cell Biol 221(10). PubMed ID: 36102907

    Mesrouze, Y., Aguilar, G., Meyerhofer, M., Bokhovchuk, F., Zimmermann, C., Fontana, P., Vissieres, A., Voshol, H., Erdmann, D., Affolter, M. and Chene, P. (2022). The role of lysine palmitoylation/myristoylation in the function of the TEAD transcription factors. Sci Rep 12(1): 4984. PubMed ID: 35322151

    Matsuda, S., Aguilar, G., Vigano, M. A. and Affolter, M. (2022). Nanobody-Based GFP Traps to Study Protein Localization and Function in Developmental Biology. Methods Mol Biol 2446: 581-593. PubMed ID: 35157295

    Matsuda, S., Schaefer, J. V., Mii, Y., Hori, Y., Bieli, D., Taira, M., Pluckthun, A. and Affolter, M. (2021). Asymmetric requirement of Dpp/BMP morphogen dispersal in the Drosophila wing disc. Nat Commun 12(1): 6435. PubMed ID: 34750371

    Roubinet, C., Tsankova, A., Pham, T. T., Monnard, A., Caussinus, E., Affolter, M. and Cabernard, C. (2017). Spatio-temporally separated cortical flows and spindle geometry establish physical asymmetry in fly neural stem cells. Nat Commun 8(1): 1383. PubMed ID: 29123099

    Ochoa-Espinosa, A., Harmansa, S., Caussinus, E. and Affolter, M. (2017). Myosin II is not required for Drosophila tracheal branch elongation and cell intercalation. Development 144(16): 2961-2968. PubMed ID: 28811312

    Matsuda, S. and Affolter, M. (2017). Dpp from the anterior stripe of cells is crucial for the growth of the Drosophila wing disc. Elife 6. PubMed ID: 28675373

    Harmansa, S., Alborelli, I., Bieli, D., Caussinus, E. and Affolter, M. (2017). A nanobody-based toolset to investigate the role of protein localization and dispersal in Drosophila. Elife 6. PubMed ID: 28395731

    Harmansa, S., Hamaratoglu, F., Affolter, M. and Caussinus, E. (2015). Dpp spreading is required for medial but not for lateral wing disc growth. Nature 527: 317-322. PubMed ID: 26550827

    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

  • Aneil Agrawal Department of Ecology & Evolutionary Biology, University of Toronto
    Singh, A. and Agrawal, N. (2022). Progressive transcriptional changes in metabolic genes and altered fatbody homeostasis in Drosophila model of Huntington's disease. Metab Brain Dis. PubMed ID: 36121619

    Singh, A. and Agrawal, A. F. (2022). Sex-Specific Variance in Fitness and the Efficacy of Selection. Am Nat 199(5): 587-602. PubMed ID: 35472021

    Yun, L., Bayoumi, M., Yang, S., Chen, P. J., Rundle, H. D. and Agrawal, A. F. (2019). Testing for local adaptation in adult male and female fitness among populations evolved under different mate competition regimes. Evolution. PubMed ID: 31206649

    Huang, Y. and Agrawal, A. F. (2016). Experimental evolution of gene expression and plasticity in alternative selective regimes. PLoS Genet 12: e1006336. PubMed ID: 27661078

    Sharp, N.P. and Agrawal, A.F. (2016). Low genetic quality alters key dimensions of the mutational spectrum. PLoS Biol 14: e1002419. PubMed ID: 27015430

    Sharp, N. P. and Agrawal, A. F. (2015). The decline in fitness with inbreeding: evidence for negative dominance-by-dominance epistasis in Drosophila melanogaster. J Evol Biol. PubMed ID: 26709722

    Huang, Y., Stinchcombe, J. R. and Agrawal, A. F. (2015). Quantitative genetic variance in experimental fly populations evolving with or without environmental heterogeneity. Evolution [Epub ahead of print]. PubMed ID: 26362112

    Yun, L. and Agrawal, A. F. (2014). Variation in the strength of inbreeding depression across environments: Effects of stress and density dependence. Evolution. PubMed ID: 25213285

    Huang, Y., Wright, S. I. and Agrawal, A. F. (2014). Genome-Wide Patterns of Genetic Variation within and among Alternative Selective Regimes. PLoS Genet 10: e1004527. PubMed ID: 25101783

    Arbuthnott, D., Agrawal, A. F. and Rundle, H. D. (2014). Remating and Sperm Competition in Replicate Populations of Drosophila melanogaster Adapted to Alternative Environments. PLoS One 9: e90207. PubMed ID: 24587283

  • Montserrat Aguadé Department of Genetics, University of Barcelona
    Orengo, D. J., Puerma, E., Cereijo, U. and Aguade, M. (2019). The molecular genealogy of sequential overlapping inversions implies both homologous chromosomes of a heterokaryotype in an inversion origin. Sci Rep 9(1): 17009. PubMed ID: 31740730

    Orengo, D. J., Puerma, E. and Aguade, M. (2019). The molecular characterization of fixed inversions breakpoints unveils the ancestral character of the Drosophila guanche chromosomal arrangements. Sci Rep 9(1): 1706. PubMed ID: 30737415

    Orengo, D. J., Aguade, M. and Juan, E. (2017). Characterization of dFOXO binding sites upstream of the Insulin Receptor P2 promoter across the Drosophila phylogeny. PLoS One 12(12): e0188357. PubMed ID: 29200426

    Puerma, E., Orengo, D. J. and Aguade, M. (2016). The origin of chromosomal inversions as a source of segmental duplications in the Sophophora subgenus of Drosophila. Sci Rep 6: 30715. PubMed ID: 27470196

    Puerma, E., Orengo, D. J. and Aguade, M. (2014). Evidence for a gene involved in multiple and diverse rearrangements in the Drosophila genus. Mol Biol Evol. PubMed ID: 25135946

    Guirao-Rico, S., Aguade, M. (2013) Patterns of Nucleotide Diversity at the Regions Encompassing the Drosophila Insulin-Like Peptide (dilp) Genes: Demography vs. Positive Selection in Drosophila melanogaster. PLoS One 8: e53593. PubMed ID: 23308258

    Papaceit, M., Segarra, C. and Aguade, M. (2013). Structure and population genetics of the breakpoints of a polymorphic inversion in Drosophila subobscura. Evolution 67: 66-79. PubMed ID: 23289562

    Khadem, M., Munte, A., Camacho, R., Aguade, M. and Segarra, C. (2012). Multilocus analysis of nucleotide variation in Drosophila madeirensis, an endemic species of the Laurisilva forest in Madeira. J Evol Biol 25: 726-739. PubMed ID: 22320146

  • Kami Ahmad Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School
    Khyzha, N., Henikoff, S. and Ahmad, K. (2022). Profiling RNA at chromatin targets in situ by antibody-targeted tagmentation. Nat Methods 19(11): 1383-1392. PubMed ID: 36192462

    Kump, A. J., Panta, M., Schwab, K. R., Inlow, M. H. and Ahmad, S. M. (2021). The Drosophila Forkhead/Fox transcription factor Jumeau mediates specific cardiac progenitor cell divisions by regulating expression of the kinesin Nebbish. Sci Rep 11(1): 3221. PubMed ID: 33547352

    Orsi, G. A., Kasinathan, S., Hughes, K. T., Saminadin-Peter, S., Henikoff, S. and Ahmad, K. (2014). High-resolution mapping defines the cooperative architecture of Polycomb response elements. Genome Res. PubMed ID: 24668908

    Schneiderman, J. I., Orsi, G. A., Hughes, K. T., Loppin, B. and Ahmad, K. (2012). Nucleosome-depleted chromatin gaps recruit assembly factors for the H3.3 histone variant. Proc Natl Acad Sci U S A 109: 19721-19726. PubMed ID: 23150573

    Greil, F. and Ahmad, K. (2012). Nucleolar dominance of the Y chromosome in Drosophila melanogaster. Genetics 191: 1119-1128. PubMed ID: 22649076

    Schneiderman, J. I., Goldstein, S. and Ahmad, K. (2010). Perturbation analysis of heterochromatin-mediated gene silencing and somatic inheritance. PLoS Genet 6. PubMed ID: 20838586

  • Syed_Tariq Ahmad Colby College, Waterville, Maine
    Tremblay, S., Zeng, Y., Yue, A., Chabot, K., Mynahan, A., Desrochers, S., Bridges, S. and Ahmad, S. T. (2022). Caffeine Delays Ethanol-Induced Sedation in Drosophila. Biology (Basel) 12(1). PubMed ID: 36671755

    Gupta, S., Verma, A. K. and Ahmad, S. (2020). Feature Selection for Topological Proximity Prediction of Single-Cell Transcriptomic Profiles in Drosophila Embryo Using Genetic Algorithm. Genes (Basel) 12(1). PubMed ID: 33379262

    Wilson, C., Kavaler, J. and Ahmad, S. T. (2019). Expression of a human variant of CHMP2B linked to neurodegeneration in Drosophila external sensory organs leads to cell fate transformations associated with increased Notch activity. Dev Neurobiol. PubMed ID: 31587468

    Liao, J., Seggio, J. A. and Ahmad, S. T. (2016). Mutations in the circadian gene period alter behavioral and biochemical responses to ethanol in Drosophila. Behav Brain Res 302: 213-219. PubMed ID: 26802726

    Liao, J., Morin, L. W. and Ahmad, S. T. (2014). Methods to Characterize Spontaneous and Startle-induced Locomotion in a Rotenone-induced Parkinson's Disease Model of Drosophila. J Vis Exp. PubMed ID: 25178101

    Cheruiyot, A., Lee, J. A., Gao, F. B. and Ahmad, S. T. (2013). Expression of mutant CHMP2B, an ESCRT-III component involved in frontotemporal dementia, causes eye deformities due to Notch misregulation in Drosophila. FASEB J. PubMed ID: 24158394

    St Laurent, R., O'Brien, L. M. and Ahmad, S. T. (2013). Sodium butyrate improves locomotor impairment and early mortality in a rotenone-induced Drosophila model of Parkinson's disease. Neuroscience 246: 382-390. PubMed ID: 23623990

    Seggio, J. A., Possidente, B. and Ahmad, S. T. (2012). Larval ethanol exposure alters adult circadian free-running locomotor activity rhythm in Drosophila melanogaster. Chronobiol Int 29: 75-81. PubMed ID: 22217104

  • Yashi Ahmed Department of Genetics, Dartmouth Medical School
    Taylor, M. J., Thompson, A. M., Alhajlah, S., Tuxworth, R. I. and Ahmed, Z. (2022). Inhibition of Chk2 promotes neuroprotection, axon regeneration, and functional recovery after CNS injury. Sci Adv 8(37): eabq2611. PubMed ID: 36103534

    Tian, A., Duwadi, D., Benchabane, H. and Ahmed, Y. (2019). Essential long-range action of Wingless/Wnt in adult intestinal compartmentalization. PLoS Genet 15(6): e1008111. PubMed ID: 31194729

    Wang, Z., Tacchelly-Benites, O., Noble, G. P., Johnson, M. K., Gagne, J. P., Poirier, G. G. and Ahmed, Y. (2018). A context-dependent role for the RNF146 ubiquitin ligase in Wingless/Wnt signaling in Drosophila. Genetics. PubMed ID: 30593492

    Wang, Z., Tacchelly-Benites, O., Noble, G. P., Johnson, M. K., Gagne, J. P., Poirier, G. G. and Ahmed, Y. (2018). A Context-Dependent Role for the RNF146 Ubiquitin Ligase in Wingless/Wnt Signaling in Drosophila. Genetics. PubMed ID: 30593492

    Tian, A., Benchabane, H., Wang, Z., Zimmerman, C., Xin, N., Perochon, J., Kalna, G., Sansom, O. J., Cheng, C., Cordero, J. B. and Ahmed, Y. (2017). Intestinal stem cell overproliferation resulting from inactivation of the APC tumor suppressor requires the transcription cofactors Earthbound and Erect wing. PLoS Genet 13(7): e1006870. PubMed ID: 28708826

    Wang, Z., Tacchelly-Benites, O., Yang, E. and Ahmed, Y. (2016). Dual roles for membrane association of Drosophila Axin in Wnt signaling. PLoS Genet 12: e1006494. PubMed ID: 27959917

    Yang, E., Tacchelly-Benites, O., Wang, Z., Randall, M. P., Tian, A., Benchabane, H., Freemantle, S., Pikielny, C., Tolwinski, N. S., Lee, E. and Ahmed, Y. (2016). Wnt pathway activation by ADP-ribosylation. Nat Commun 7: 11430. PubMed ID: 27138857

    Wang, Z., Tacchelly-Benites, O., Yang, E., Thorne, C. A., Nojima, H., Lee, E. and Ahmed, Y. (2016). Wnt/Wingless pathway activation is promoted by a critical threshold of Axin maintained by the tumor suppressor APC and the ADP-ribose polymerase Tankyrase. Genetics [Epub ahead of print]. PubMed ID: 26975665

    Tian, A., Benchabane, H., Wang, Z. and Ahmed, Y. (2016). Regulation of stem cell proliferation and cell fate specification by Wingless/Wnt signaling gradients enriched at adult intestinal compartment boundaries. PLoS Genet 12: e1005822. PubMed ID: 26845150

    Xin, N., Benchabane, H., Tian, A., Nguyen, K., Klofas, L. and Ahmed, Y. (2011). Erect Wing facilitates context-dependent Wnt/Wingless signaling by recruiting the cell-specific Armadillo-TCF adaptor Earthbound to chromatin. Development 138: 4955-4967. PubMed ID: 22028028

  • Toshiro Aigaki Department of Biological Sciences, Tokyo Metropolitan University
    Ohnuma, K., Kishita, Y., Nyuzuki, H., Kohda, M., Ohtsu, Y., Takeo, S., Asano, T., Sato-Miyata, Y., Ohtake, A., Murayama, K., Okazaki, Y. and Aigaki, T. (2020). Ski3/TTC37 deficiency associated with trichohepatoenteric syndrome causes mitochondrial dysfunction in Drosophila. FEBS Lett. PubMed ID: 32294252

    Funakoshi, M., Tsuda, M., Muramatsu, K., Hatsuda, H., Morishita, S. and Aigaki, T. (2018). Overexpression of Larp4B downregulates dMyc and reduces cell and organ sizes in Drosophila. Biochem Biophys Res Commun 497(2): 762-768. PubMed ID: 29462618

    Quan, X., Sato-Miyata, Y., Tsuda, M., Muramatsu, K., Asano, T., Takeo, S. and Aigaki, T. (2016). Deficiency of succinyl-CoA synthetase alpha subunit delays development, impairs locomotor activity and reduces survival under starvation in Drosophila. Biochem Biophys Res Commun. PubMed ID: 28017724

    Tsuda, M. and Aigaki, T. (2016). Evolution of sex-peptide in Drosophila. Fly (Austin): 0. PubMed ID: 27230918

    Seong, K.H., Tsuda, M., Tsuda-Sakurai, K. and Aigaki, T. (2015). The plant homeodomain finger protein MESR4 is essential for embryonic development in Drosophila. Genesis [Epub ahead of print]. PubMed ID: 26467775

    Tsuda, M., Peyre, J. B., Asano, T. and Aigaki, T. (2015). Visualizing molecular functions and cross-species activity of Sex-peptide in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 26022240

    Sato-Miyata, Y., Muramatsu, K., Funakoshi, M., Tsuda, M. and Aigaki, T. (2014). Overexpression of dilp2 causes nutrient-dependent semi-lethality in Drosophila. Front Physiol 5: 147. PubMed ID: 24795642

    Cheng, Z., Tsuda, M., Kishita, Y., Sato, Y., Aigaki, T. (2013) Impaired energy metabolism in a Drosophila model of mitochondrial aconitase deficiency. Biochem Biophys Res Commun. PubMed ID: 23438437

    Tomioka, S., Aigaki, T. and Matsuo, T. (2012). Conserved cis-regulatory elements of two odorant-binding protein genes, Obp57d and Obp57e, in Drosophila. Genes Genet Syst 87: 323-329. PubMed ID: 23412634

    Nakagawa-Yagi, Y., Sato, Y., Matsumoto, E., Nakatsuka, S., Sakaki, T., Muramatsu, Y., Hara, T. and Aigaki, T. (2012). Pharmacological modulation of histone demethylase activity by a small molecule isolated from subcritical water extracts of Sasa senanensis leaves prolongs the lifespan of Drosophila melanogaster. BMC Complement Altern Med 12: 101. PubMed ID: 22809229

    Takeo, S., Swanson, S. K., Nandanan, K., Nakai, Y., Aigaki, T., Washburn, M. P., Florens, L. and Hawley, R. S. (2012). Shaggy/glycogen synthase kinase 3beta and phosphorylation of Sarah/regulator of calcineurin are essential for completion of Drosophila female meiosis. Proc Natl Acad Sci U S A 109: 6382-6389. PubMed ID: 22421435

  • Michael Akam Department of Zoology, University of Cambridge
    Tidswell, O. R. A., Benton, M. A. and Akam, M. (2021). The neuroblast timer gene nubbin exhibits functional redundancy with gap genes to regulate segment identity in Tribolium. Development 148(16). PubMed ID: 34351412

    Clark, E. and Akam, M. (2016). Odd-paired controls frequency doubling in Drosophila segmentation by altering the pair-rule gene regulatory network. Elife [Epub ahead of print]. PubMed ID: 27525481

    Janssens, H., Siggens, K., Cicin-Sain, D., Jimenez-Guri, E., Musy, M., Akam, M. and Jaeger, J. (2014). A quantitative atlas of Even-skipped and Hunchback expression in Clogmia albipunctata (Diptera: Psychodidae) blastoderm embryos. Evodevo 5: 1. PubMed ID: 24393251

    Pavlopoulos, A. and Akam, M. (2011). Hox gene Ultrabithorax regulates distinct sets of target genes at successive stages of Drosophila haltere morphogenesis. Proc. Natl. Acad. Sci. 108(7): 2855-60. PubMed ID: 21282633

    Steinmetz, P. R., Urbach, R., Posnien, N., Eriksson, J., Kostyuchenko, R. P., Brena, C., Guy, K., Akam, M., Bucher, G. and Arendt, D. (2010). Six3 demarcates the anterior-most developing brain region in bilaterian animals. Evodevo 1: 14. PubMed ID: 21190549

    de Navas, L. F., Reed, H., Akam, M., Barrio, R., Alonso, C. R. and Sanchez-Herrero, E. (2011). Integration of RNA processing and expression level control modulates the function of the Drosophila Hox gene Ultrabithorax during adult development. Development 138: 107-116. PubMed ID: 21115609

  • Anna Akhmanova Cell Biology, Utrecht University
    Doodhi, H., Katrukha, E. A., Kapitein, L. C. and Akhmanova, A. (2014). Mechanical and Geometrical Constraints Control Kinesin-Based Microtubule Guidance. Curr Biol. PubMed ID: 24462000

  • Asifa Akhtar Chromatin Regulation, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
    Iyer, S. S., Sun, Y., Seyfferth, J., Manjunath, V., Samata, M., Alexiadis, A., Kulkarni, T., Gutierrez, N., Georgiev, P., Shvedunova, M. and Akhtar, A. (2023). The NSL complex is required for piRNA production from telomeric clusters. Life Sci Alliance 6(9). PubMed ID: 37399316

    Keller Valsecchi, C. I., Marois, E., Basilicata, M. F., Georgiev, P. and Akhtar, A. (2021). Distinct mechanisms mediate X chromosome dosage compensation in Anopheles and Drosophila. Life Sci Alliance 4(9). PubMed ID: 34266874

    Valsecchi, C. I. K., Basilicata, M. F., Georgiev, P., Gaub, A., Seyfferth, J., Kulkarni, T., Panhale, A., Semplicio, G., Manjunath, V., Holz, H., Dasmeh, P. and Akhtar, A. (2020). RNA nucleation by MSL2 induces selective X chromosome compartmentalization. Nature. PubMed ID: 33208948

    Samata, M., Alexiadis, A., Richard, G., Georgiev, P., Nuebler, J., Kulkarni, T., Renschler, G., Basilicata, M. F., Zenk, F. L., Shvedunova, M., Semplicio, G., Mirny, L., Iovino, N. and Akhtar, A. (2020). Intergenerationally Maintained Histone H4 Lysine 16 Acetylation Is Instructive for Future Gene Activation. Cell. PubMed ID: 32502394

    Gaub, A., Sheikh, B. N., Basilicata, M. F., Vincent, M., Nizon, M., Colson, C., Bird, M. J., Bradner, J. E., Thevenon, J., Boutros, M. and Akhtar, A. (2020). Evolutionary conserved NSL complex/BRD4 axis controls transcription activation via histone acetylation. Nat Commun 11(1): 2243. PubMed ID: 32382029

    Renschler, G., Richard, G., Valsecchi, C. I. K., Toscano, S., Arrigoni, L., Ramirez, F. and Akhtar, A. (2019). Hi-C guided assemblies reveal conserved regulatory topologies on X and autosomes despite extensive genome shuffling. Genes Dev. PubMed ID: 31601616

    Bhardwaj, V., Semplicio, G., Erdogdu, N. U., Manke, T. and Akhtar, A. (2019). MAPCap allows high-resolution detection and differential expression analysis of transcription start sites. Nat Commun 10(1): 3219. PubMed ID: 31363093

    Sheikh, B. N., Guhathakurta, S. and Akhtar, A. (2019). The non-specific lethal (NSL) complex at the crossroads of transcriptional control and cellular homeostasis. EMBO Rep. PubMed ID: 31160312

    Panhale, A., Richter, F. M., Ramirez, F., Shvedunova, M., Manke, T., Mittler, G. and Akhtar, A. (2019). CAPRI enables comparison of evolutionarily conserved RNA interacting regions. Nat Commun 10(1): 2682. PubMed ID: 31213602

    Lam, K. C., Chung, H. R., Semplicio, G., Iyer, S. S., Gaub, A., Bhardwaj, V., Holz, H., Georgiev, P. and Akhtar, A. (2019). The NSL complex-mediated nucleosome landscape is required to maintain transcription fidelity and suppression of transcription noise. Genes Dev. PubMed ID: 30819819

    Ramirez, F., Bhardwaj, V., Arrigoni, L., Lam, K. C., Gruning, B. A., Villaveces, J., Habermann, B., Akhtar, A. and Manke, T. (2018). High-resolution TADs reveal DNA sequences underlying genome organization in flies. Nat Commun 9(1): 189. PubMed ID: 29335486

  • Joerg Albert School of Life and Medical Sciences, University College London
    Keder, A., Tardieu, C., Malong, L., Filia, A., Kashkenbayeva, A., Newton, F., Georgiades, M., Gale, J. E., Lovett, M., Jarman, A. P. and Albert, J. T. (2020). Homeostatic maintenance and age-related functional decline in the Drosophila ear. Sci Rep 10(1): 7431. PubMed ID: 32366993

    Simoni, A., Wolfgang, W., Topping, M. P., Kavlie, R. G., Stanewsky, R. and Albert, J. T. (2014). A mechanosensory pathway to the Drosophila circadian clock. Science 343: 525-528. PubMed ID: 24482478

    Effertz, T., Nadrowski, B., Piepenbrock, D., Albert, J. T. and Gopfert, M. C. (2012). Direct gating and mechanical integrity of Drosophila auditory transducers require TRPN1. Nat Neurosci 15: 1198-1200. PubMed ID: 22842145

    Riabinina, O., Dai, M., Duke, T. and Albert, J. T. (2011). Active process mediates species-specific tuning of Drosophila ears. Curr Biol 21: 658-664. PubMed ID: 21458268

  • Esther Alcorta Laboratory of Neurogenetics, Department of Functional Biology, University of Oviedo, Spain
    Calvin-Cejudo, L., Martin, F., Mendez, L. R., Coya, R., Castaneda-Sampedro, A., Gomez-Diaz, C. and Alcorta, E. (2023). Neuron-glia interaction at the receptor level affects olfactory perception in adult Drosophila. iScience 26(1): 105837. PubMed ID: 36624835

    Castaneda-Sampedro, A., Calvin-Cejudo, L., Martin, F., Gomez-Diaz, C. and Alcorta, E. (2022). The Ntan1 gene is expressed in perineural glia and neurons of adult Drosophila. Sci Rep 12(1): 14749. PubMed ID: 36042338

    Martin, F. and Alcorta, E. (2016). Measuring activity in olfactory receptor neurons in Drosophila: Focus on spike amplitude. J Insect Physiol 95: 23-41. PubMed ID: 27614176

    Martin, F., Boto, T., Gomez-Diaz, C. and Alcorta, E. (2013). Elements of Olfactory Reception in Adult Drosophila melanogaster. Anat Rec (Hoboken). PubMed ID: 23904114

    Riveron, J., Boto, T., Alcorta, E. (2013) Transcriptional basis of the acclimation to high environmental temperature at the olfactory receptor organs of Drosophila melanogaster. BMC Genomics 14: 259. PubMed ID: 23590196

    Martin, F., Riveron, J. and Alcorta, E. (2011). Environmental temperature modulates olfactory reception in Drosophila melanogaster. J Insect Physiol 57: 1631-1642. PubMed ID: 21924272

    Boto, T., Gomez-Diaz, C. and Alcorta, E. (2010). Expression analysis of the 3 G-protein subunits, Galpha, Gbeta, and Ggamma, in the olfactory receptor organs of adult Drosophila melanogaster. Chem Senses 35: 183-193. PubMed ID: 20047983

  • Mattias Alenius Linköping University, Sweden
    Zhao, Y., Khallaf, M. A., Johansson, E., Dzaki, N., Bhat, S., Alfredsson, J., Duan, J., Hansson, B. S., Knaden, M. and Alenius, M. (2022). Hedgehog-mediated gut-taste neuron axis controls sweet perception in Drosophila. Nat Commun 13(1): 7810. PubMed ID: 36535958

    Jafari, S., Henriksson, J., Yan, H. and Alenius, M. (2021). Stress and odorant receptor feedback during a critical period after hatching regulates olfactory sensory neuron differentiation in Drosophila. PLoS Biol 19(4): e3001101. PubMed ID: 33793547

    Jafari, S. and Alenius, M. (2021). Odor response adaptation in Drosophila-a continuous individualization process. Cell Tissue Res 383(1): 143-148. PubMed ID: 33492517

    Sanchez, G. M., Alkhori, L., Hatano, E., Schultz, S. W., Kuzhandaivel, A., Jafari, S., Granseth, B. and Alenius, M. (2016). Hedgehog signaling regulates the ciliary transport of odorant receptors in Drosophila. Cell Rep 14: 464-470. PubMed ID: 26774485

    Jafari, S. and Alenius, M. (2015). Cis-regulatory mechanisms for robust olfactory sensory neuron class-restricted odorant receptor gene expression in Drosophila. PLoS Genet 11: e1005051. PubMed ID: 25760344

    Kuzhandaivel, A., Schultz, S. W., Alkhori, L. and Alenius, M. (2014). Cilia-Mediated Hedgehog Signaling in Drosophila. Cell Rep. PubMed ID: 24768000

    Alkhori, L., Ost, A. and Alenius, M. (2013). The corepressor Atrophin specifies odorant receptor expression in Drosophila. FASEB J. PubMed ID: 24334704 <><>Jafari, S., et al. (2012). Combinatorial activation and repression by seven transcription factors specify Drosophila odorant receptor expression. PLoS Biol. 10(3): e1001280. PubMed Citation: 22427741

  • Nazif Alic Institute of Healthy Ageing, Department of Genetics, Evolution & Environment, University College London
    Corrales, G. M., Li, M., Svermova, T., Goncalves, A., Voicu, D., Dobson, A. J., Southall, T. D. and Alic, N. (2022). Transcriptional memory of dFOXO activation in youth curtails later-life mortality through chromatin remodelling and Xbp1. Nat Aging 2: 1176-1190. PubMed ID: 37064330

    Martinez Corrales, G., Filer, D., Wenz, K. C., Rogan, A., Phillips, G., Li, M., Feseha, Y., Broughton, S. J. and Alic, N. (2020). Partial Inhibition of RNA Polymerase I Promotes Animal Health and Longevity. Cell Rep 30(6): 1661-1669. PubMed ID: 32049000

    Dobson, A. J., Boulton-McDonald, R., Houchou, L., Svermova, T., Ren, Z., Subrini, J., Vazquez-Prada, M., Hoti, M., Rodriguez-Lopez, M., Ibrahim, R., Gregoriou, A., Gkantiragas, A., Bahler, J., Ezcurra, M. and Alic, N. (2019). Longevity is determined by ETS transcription factors in multiple tissues and diverse species. PLoS Genet 15(7): e1008212. PubMed ID: 31356597

    Dobson, A. J., Ezcurra, M., Flanagan, C. E., Summerfield, A. C., Piper, M. D., Gems, D. and Alic, N. (2017). Nutritional programming of lifespan by FOXO inhibition on sugar-rich diets. Cell Rep 18(2): 299-306. PubMed ID: 28076775

    Burnouf, S., Gronke, S., Augustin, H., Dols, J., Gorsky, M. K., Werner, J., Kerr, F., Alic, N., Martinez, P. and Partridge, L. (2016). Deletion of endogenous Tau proteins is not detrimental in Drosophila. Sci Rep 6: 23102. PubMed ID: 26976084

    Afschar, S., Toivonen, J. M., Hoffmann, J. M., Tain, L. S., Wieser, D., Finlayson, A. J., Driege, Y., Alic, N., Emran, S., Stinn, J., Froehlich, J., Piper, M. D. and Partridge, L. (2016). Nuclear hormone receptor DHR96 mediates the resistance to xenobiotics but not the increased lifespan of insulin-mutant Drosophila. Proc Natl Acad Sci U S A 113(5): 1321-1326. PubMed ID: 26787908

    Al Saud, S. N., Summerfield, A. C. and Alic, N. (2015). Ablation of insulin-producing cells prevents obesity but not premature mortality caused by a high-sugar diet in Drosophila. Proc Biol Sci 282(1800): 20141720. PubMed ID: 25520354

  • Ravi Allada Department of Neurobiology, Northwestern University, Evanston, Il
    Hwangbo, D. S., Kwon, Y. J., Iwanaszko, M., Jiang, P., Abbasi, L., Wright, N., Alli, S., Hutchison, A. L., Dinner, A. R., Braun, R. I. and Allada, R. (2023). Dietary Restriction Impacts Peripheral Circadian Clock Output Important for Longevity in Drosophila. bioRxiv. PubMed ID: 36711760

    van Alphen, B., Stewart, S., Iwanaszko, M., Xu, F., Li, K., Rozenfeld, S., Ramakrishnan, A., Itoh, T. Q., Sisobhan, S., Qin, Z., Lear, B. C. and Allada, R. (2022). Glial immune-related pathways mediate effects of closed head traumatic brain injury on behavior and lethality in Drosophila. PLoS Biol 20(1): e3001456. PubMed ID: 35081110

    Zhang, M. Y., Lear, B. C. and Allada, R. (2021). The microtubule associated protein tau suppresses the axonal distribution of PDF neuropeptide and mitochondria in circadian clock neurons. Hum Mol Genet. PubMed ID: 34750631

    Lee, J., Lim, C., Han, T. H., Andreani, T., Moye, M., Curran, J., Johnson, E., Kath, W. L., Diekman, C. O., Lear, B. C. and Allada, R. (2021). The E3 ubiquitin ligase adaptor Tango10 links the core circadian clock to neuropeptide and behavioral rhythms. Proc Natl Acad Sci U S A 118(47). PubMed ID: 34799448

    van Alphen, B., Semenza, E. R., Yap, M., van Swinderen, B. and Allada, R. (2021). A deep sleep stage in Drosophila with a functional role in waste clearance. Sci Adv 7(4). PubMed ID: 33523916

    Kula-Eversole, E., Lee, D. H., Samba, I., Yildirim, E., Levine, D. C., Hong, H. K., Lear, B. C., Bass, J., Rosbash, M. and Allada, R. (2020). Phosphatase of Regenerating Liver-1 Selectively Times Circadian Behavior in Darkness via Function in PDF Neurons and Dephosphorylation of TIMELESS. Curr Biol. PubMed ID: 33157022

    Xu, F., Kula-Eversole, E., Iwanaszko, M., Lim, C. and Allada, R. (2019). Ataxin2 functions via CrebA to mediate Huntingtin toxicity in circadian clock neurons. PLoS Genet 15(10): e1008356. PubMed ID: 31593562

    Xu, F., Kula-Eversole, E., Iwanaszko, M., Hutchison, A. L., Dinner, A. and Allada, R. (2019). Circadian clocks function in concert with heat shock organizing protein to modulate mutant Huntingtin aggregation and toxicity. Cell Rep 27(1): 59-70.e54. PubMed ID: 30943415

    Flourakis, M., Kula-Eversole, E., Hutchison, A. L., Han, T. H., Aranda, K., Moose, D. L., White, K. P., Dinner, A. R., Lear, B. C., Ren, D., Diekman, C. O., Raman, I. M. and Allada, R. (2015). A Conserved Bicycle Model for Circadian Clock Control of Membrane Excitability. Cell 162: 836-848. PubMed ID: 26276633

    Hutchison, A. L., Maienschein-Cline, M., Chiang, A. H., Tabei, S. M., Gudjonson, H., Bahroos, N., Allada, R. and Dinner, A. R. (2015). Improved statistical methods enable greater sensitivity in rhythm detection for genome-wide data. PLoS Comput Biol 11: e1004094. PubMed ID: 25793520

  • Doug Allan Life Sciences Centre, Vancouver
    Vaikakkara Chithran, A., Allan, D. W. and O'Connor, T. P. (2023). Adult expression of Semaphorins and Plexins is essential for motor neuron survival. Sci Rep 13(1): 5894. PubMed ID: 37041188

    Vuilleumier, R., Miao, M., Medina-Giro, S., Ell, C. M., Flibotte, S., Lian, T., Kauwe, G., Collins, A., Ly, S., Pyrowolakis, G., Haghighi, A. P. and Allan, D. W. (2022). Dichotomous cis-regulatory motifs mediate the maturation of the neuromuscular junction by retrograde BMP signaling. Nucleic Acids Res 50(17): 9748-9764. PubMed ID: 36029115

    Ganguly, P., Madonsela, L., Chao, J. T., Loewen, C. J. R., O'Connor, T. P., Verheyen, E. M. and Allan, D. W. (2021). A scalable Drosophila assay for clinical interpretation of human PTEN variants in suppression of PI3K/AKT induced cellular proliferation. PLoS Genet 17(9): e1009774. PubMed ID: 34492006

    Berndt, A. J., Othonos, K. M., Lian, T., Flibotte, S., Miao, M., Bhuiyan, S. A., Cho, R. Y., Fong, J. S., Hur, S. A., Pavlidis, P. and Allan, D. W. (2020). A low affinity cis-regulatory BMP response element restricts target gene activation to subsets of Drosophila neurons. Elife 9. PubMed ID: 33124981

    Vuilleumier, R., Lian, T., Flibotte, S., Khan, Z. N., Fuchs, A., Pyrowolakis, G. and Allan, D. W. (2018). Retrograde BMP signaling activates neuronal gene expression through widespread deployment of a conserved BMP-responsive cis-regulatory activation element. Nucleic Acids Res. PubMed ID: 30476189

    Garner, S. R. C., Castellanos, M. C., Baillie, K. E., Lian, T. and Allan, D. W. (2018). Drosophila female-specific Ilp7 motoneurons are generated by Fruitless-dependent cell death in males and by a double-assurance survival role for Transformer in females. Development 145(1). PubMed ID: 29229771

    Hegle, A. P., Frank, C. A., Berndt, A., Klose, M., Allan, D. W. and Accili, E. A. (2017). The Ih channel gene promotes synaptic transmission and coordinated movement in Drosophila melanogaster. Front Mol Neurosci 10: 41. PubMed ID: 28286469

    Berndt, A. J., Tang, J. C., Ridyard, M. S., Lian, T., Keatings, K. and Allan, D. W. (2015). Gene regulatory mechanisms underlying the spatial and temporal regulation of target-dependent gene expression in Drosophila neurons. PLoS Genet 11: e1005754. PubMed ID: 26713626

    Allan, D. W. and Thor, S. (2015). Transcriptional selectors, masters, and combinatorial codes: regulatory principles of neural subtype specification. Wiley Interdiscip Rev Dev Biol. PubMed ID: 25855098

    Veverytsa, L., Allan, D. W. (2013) Subtype-specific neuronal remodeling during Drosophila metamorphosis. Fly (Austin) 7. PubMed ID: 23579264

    Eade, K. T., Fancher, H. A., Ridyard, M. S. and Allan, D. W. (2012). Developmental transcriptional networks are required to maintain neuronal subtype identity in the mature nervous system. PLoS Genet 8: e1002501. PubMed ID: 22383890

  • Debasmita Alone National Institute of Science Education and Research, Bhubaneswar, Odisha
    Das, S. S., Nanda, G. G. and Alone, D. P. (2014). Artemisinin and Curcumin inhibit Drosophila brain tumor, prolong life span, and restore locomotor activity. IUBMB Life. PubMed ID: 24975030

    Alone, D. P., Rodriguez, J. C., Noland, C. L. and Nash, H. A. (2009). Impact of gene copy number variation on anesthesia in Drosophila melanogaster. Anesthesiology 111: 15-24. PubMed ID: 19546691

  • Claudio Alonso University of Sussex
    Raouf Issa, A., J, A. C. M., Padmanabhan, A. and Alonso, C. R. (2022). A novel post-developmental role of the Hox genes underlies normal adult behavior. Proc Natl Acad Sci U S A 119(49): e2209531119. PubMed ID: 36454751

    Klann, M., Issa, A. R., Pinho, S. and Alonso, C. R. (2021). microRNA-dependent control of sensory neuron function regulates posture behaviour in Drosophila. J Neurosci. PubMed ID: 34417328

    Issa, A. R., Picao-Osorio, J., Rito, N., Chiappe, M. E. and Alonso, C. R. (2019). A single microRNA-Hox gene module controls equivalent movements in biomechanically distinct forms of Drosophila. Curr Biol. PubMed ID: 31327720

    Kaschula, R., Pinho, S. and Alonso, C. R. (2018). microRNA-dependent regulation of Hox gene expression sculpts fine-grain morphological patterns in a Drosophila appendage. Development. PubMed ID: 30143542

    Picao-Osorio, J., Lago-Baldaia, I., Patraquim, P. and Alonso, C. R. (2017). Pervasive Behavioural Effects of microRNA Regulation in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 28468905

    Vallejos Baier, R., Picao-Osorio, J. and Alonso, C. R. (2017). Molecular Regulation of Alternative Polyadenylation (APA) within the Drosophila Nervous System. J Mol Biol. PubMed ID: 28366829

    Warnefors, M., Hartmann, B., Thomsen, S. and Alonso, C. R. (2016). Combinatorial gene regulatory functions underlie ultraconserved elements in Drosophila. Mol Biol Evol [Epub ahead of print]. PubMed ID: 27247329

    Picao-Osorio, J., Johnston, J., Landgraf, M., Berni, J. and Alonso, C.R. (2015).MicroRNA-encoded behavior in Drosophila. Science [Epub ahead of print]. PubMed ID: 26494171

    Casadio, A., Longman, D., Hug, N., Delavaine, L., Vallejos Baier, R., Alonso, C. R. and Caceres, J. F. (2014). Identification and characterization of novel factors that act in the nonsense-mediated mRNA decay pathway in nematodes, flies and mammals. EMBO Rep. PubMed ID: 25452588

    Rogulja-Ortmann, A., Picao-Osorio, J., Villava, C., Patraquim, P., Lafuente, E., Aspden, J., Thomsen, S., Technau, G. M. and Alonso, C. R. (2014). The RNA-binding protein ELAV regulates Hox RNA processing, expression and function within the Drosophila nervous system. Development 141: 2046-2056. PubMed ID: 24803653

    Mallo, M. and Alonso, C. R. (2013). The regulation of Hox gene expression during animal development. Development 140: 3951-3963. PubMed ID: 24046316

    Patraquim, P., Warnefors, M. and Alonso, C. R. (2011). Evolution of Hox post-transcriptional regulation by alternative polyadenylation and microRNA modulation within 12 Drosophila genomes. Mol Biol Evol 28: 2453-2460. PubMed ID: 21436120

  • Benjamin Altenhein Institut für Genetic, Gutenberg Universitat, Mainz
    Zulbahar, S., Sieglitz, F., Kottmeier, R., Altenhein, B., Rumpf, S. and Klambt, C. (2018). Differential expression of the Drosophila Ntan/Obek controls ploidy in the blood-brain barrier. Development. PubMed ID: 30002129

    Ryglewski, S., Duch, C. and Altenhein, B. (2017). Tyramine actions on Drosophila flight behavior are affected by a glial dehydrogenase/reductase. Front Syst Neurosci 11: 68. PubMed ID: 29021745

    Altenhein, B., Cattenoz, P. B. and Giangrande, A. (2015). The early life of a fly glial cell. Wiley Interdiscip Rev Dev Biol. PubMed ID: 26224590

    Altenhein, B. (2015). Glial cell progenitors in the Drosophila embryo. Glia [Epub ahead of print]. PubMed ID: 25779863

    von Hilchen, C. and Altenhein, B. (2014). Tracing cells throughout development: insights into single glial cell differentiation. Fly (Austin) 8: 7-11. PubMed ID: 25483246

    von Hilchen, C. and Altenhein, B. (2014). Tracing cells throughout development: insights into single glial cell differentiation. Fly (Austin) 8. PubMed ID: 24769855

    Sieglitz, F., Matzat, T., Yuva-Adyemir, Y., Neuert, H., Altenhein, B. and Klambt, C. (2013). Antagonistic feedback loops involving rau and sprouty in the Drosophila eye control neuronal and glial differentiation. Sci Signal 6: ra96. PubMed ID: 24194583

    von Hilchen, C. M., Bustos, A. E., Giangrande, A., Technau, G. M. and Altenhein, B. (2013). Predetermined embryonic glial cells form the distinct glial sheaths of the Drosophila peripheral nervous system. Development. PubMed ID: 23903191

    Wang, S., Meyer, H., Ochoa-Espinosa, A., Buchwald, U., Onel, S., Altenhein, B., Heinisch, J. J., Affolter, M. and Paululat, A. (2012). GBF1 (Gartenzwerg)-dependent secretion is required for Drosophila tubulogenesis. J Cell Sci 125: 461-472. PubMed ID: 22302994

    Albrecht, S., Altenhein, B. and Paululat, A. (2011). The transmembrane receptor Uncoordinated5 (Unc5) is essential for heart lumen formation in Drosophila melanogaster. Dev Biol 350: 89-100. PubMed ID: 21094637

  • Stefan Ameres Institute of Molecular Biotechnology, Vienna
    Xie, W., Sowemimo, I., Hayashi, R., Wang, J., Burkard, T. R., Brennecke, J., Ameres, S. L. and Patel, D. J. (2020). Structure-function analysis of microRNA 3'-end trimming by Nibbler. Proc Natl Acad Sci U S A. PubMed ID: 33199607

    Reichholf, B., Herzog, V. A., Fasching, N., Manzenreither, R. A., Sowemimo, I. and Ameres, S. L. (2019). Time-resolved small RNA sequencing unravels the molecular principles of microRNA homeostasis. Mol Cell. PubMed ID: 31350118

    Reimao-Pinto, M. M., Manzenreither, R. A., Burkard, T. R., Sledz, P., Jinek, M., Mechtler, K. and Ameres, S. L. (2016). Molecular basis for cytoplasmic RNA surveillance by uridylation-triggered decay in Drosophila. EMBO J [Epub ahead of print]. PubMed ID: 27729457

    Reimao-Pinto, M. M., Ignatova, V., Burkard, T. R., Hung, J. H., Manzenreither, R. A., Sowemimo, I., Herzog, V. A., Reichholf, B., Farina-Lopez, S. and Ameres, S. L. (2015). Uridylation of RNA Hairpins by Tailor Confines the Emergence of MicroRNAs in Drosophila. Mol Cell. PubMed ID: 26145176

    Bortolamiol-Becet, D., Hu, F., Jee, D., Wen, J., Okamura, K., Lin, C. J., Ameres, S. L. and Lai, E. C. (2015). Selective Suppression of the Splicing-Mediated MicroRNA Pathway by the Terminal Uridyltransferase Tailor. Mol Cell. PubMed ID: 26145174

    Han, B. W., Hung, J. H., Weng, Z., Zamore, P. D. and Ameres, S. L. (2011). The 3'-to-5' exoribonuclease Nibbler shapes the 3' ends of microRNAs bound to Drosophila Argonaute1. Curr Biol 21: 1878-1887. PubMed ID: 22055293

    Ameres, S. L., Hung, J. H., Xu, J., Weng, Z. and Zamore, P. D. (2011). Target RNA-directed tailing and trimming purifies the sorting of endo-siRNAs between the two Drosophila Argonaute proteins. RNA 17: 54-63. PubMed ID: 21106652

  • Marc Amoyel Cell & Developmental Biology, Div of Biosciences, University College London
    Yuen, A. C., Prasad, A. R., Fernandes, V. M. and Amoyel, M. (2022). A kinase translocation reporter reveals real-time dynamics of ERK activity in Drosophila. Biol Open 11(5). PubMed ID: 35608229

    Sainz de la Maza, D., Hof-Michel, S., Phillimore, L., Bokel, C. and Amoyel, M. (2022). Cell-cycle exit and stem cell differentiation are coupled through regulation of mitochondrial activity in the Drosophila testis. Cell Rep 39(6): 110774. PubMed ID: 35545055

    Yuen, A. C., Hillion, K. H., Wang, R. and Amoyel, M. (2021). Germ cells commit somatic stem cells to differentiation following priming by PI3K/Tor activity in the Drosophila testis. PLoS Genet 17(12): e1009609. PubMed ID: 34898607

    Herrera, S. C., Sainz de la Maza, D., Grmai, L., Margolis, S., Plessel, R., Burel, M., O'Connor, M., Amoyel, M. and Bach, E. A. (2021). Proliferative stem cells maintain quiescence of their niche by secreting the Activin inhibitor Follistatin. Dev Cell 56(16): 2284-2294 e2286. PubMed ID: 34363758

  • Hubert Amrein Texas A&M Health Science Center, College Station
    Fujii, S., Ahn, J. E., Jagge, C., Shetty, V., Janes, C., Mohanty, A., Slotman, M., Adelman, Z. N. and Amrein, H. (2023). RNA Taste Is Conserved in Dipteran Insects. J Nutr. PubMed ID: 36907444
    Miyamoto, T. and Amrein, H. (2019). Neuronal gluconeogenesis regulates systemic glucose homeostasis in Drosophila melanogaster. Curr Biol 29(8): 1263-1272.e1265. PubMed ID: 30930040

    Mishra, D., Thorne, N., Miyamoto, C., Jagge, C. and Amrein, H. (2018). The taste of ribonucleosides: Novel macronutrients essential for larval growth are sensed by Drosophila gustatory receptor proteins. PLoS Biol 16(8): e2005570. PubMed ID: 30086130

    Ahn, J. E., Chen, Y. and Amrein, H. (2017). Molecular basis of fatty acid taste in Drosophila. Elife 6. PubMed ID: 29231818

    Chen, Y. and Amrein, H. (2017). Ionotropic receptors mediate Drosophila oviposition preference through sour gustatory receptor neurons. Curr Biol 27(18): 2741-2750.e2744. PubMed ID: 28889974

    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

    Fujii, S., Yavuz, A., Slone, J., Jagge, C., Song, X. and Amrein, H. (2015). Drosophila sugar receptors in sweet taste perception, olfaction, and internal nutrient sensing. Curr Biol 25: 621-627. PubMed ID: 25702577

    Chen, Y. and Amrein, H. (2014). Enhancing Perception of Contaminated Food through Acid-Mediated Modulation of Taste Neuron Responses. Curr Biol. PubMed ID: 25131671

    Miyamoto, T. and Amrein, H. (2013). Diverse roles for the fructose sensor Gr43a. Fly (Austin) 8. PubMed ID: 24406333

    Mishra, D., Miyamoto, T., Rezenom, Y. H., Broussard, A., Yavuz, A., Slone, J., Russell, D. H. and Amrein, H. (2013). The Molecular Basis of Sugar Sensing in Drosophila Larvae. Curr Biol. PubMed ID: 23850280

    Miyamoto, T., Wright, G., Amrein, H. (2013) Nutrient sensors. Curr Biol 23: R369-373. PubMed ID: 23660359

  • David Anderson Division of Biology, California Institute of Technology
    Chiu, H., Hoopfer, E. D., Coughlan, M. L. and Anderson, D. J. (2020). A circuit logic for sexually shared and dimorphic aggressive behaviors in Drosophila. Cell. PubMed ID: 33382967

    Jung, Y., Kennedy, A., Chiu, H., Mohammad, F., Claridge-Chang, A. and Anderson, D. J. (2019). Neurons that function within an integrator to promote a persistent behavioral state in Drosophila. Neuron. PubMed ID: 31810837

    Ding, K., Han, Y., Seid, T. W., Buser, C., Karigo, T., Zhang, S., Dickman, D. K. and Anderson, D. J. (2019). Imaging neuropeptide release at synapses with a genetically engineered reporter. Elife 8. PubMed ID: 31241464

    Duistermars, B. J., Pfeiffer, B. D., Hoopfer, E. D. and Anderson, D. J. (2018). A brain module for scalable control of complex, multi-motor threat displays. Neuron. PubMed ID: 30415997

    Hoopfer, E. D., Jung, Y., Inagaki, H. K., Rubin, G. M. and Anderson, D. J. (2015). P1 interneurons promote a persistent internal state that enhances inter-male aggression in Drosophila. Elife 4. PubMed ID: 26714106

    Inagaki, H. K., Panse, K. M. and Anderson, D. J. (2014). Independent, reciprocal neuromodulatory control of sweet and bitter taste sensitivity during starvation in Drosophila. Neuron 84: 806-820. PubMed ID: 25451195

    Lim, R. S., Eyjolfsdottir, E., Shin, E., Perona, P. and Anderson, D. J. (2014). How food controls aggression in Drosophila. PLoS One 9: e105626. PubMed ID: 25162609

    Anderson, D. J. and Adolphs, R. (2014). A Framework for Studying Emotions across Species. Cell 157: 187-200. PubMed ID: 24679535

    Asahina, K., Watanabe, K., Duistermars, B. J., Hoopfer, E., Gonzalez, C. R., Eyjolfsdottir, E. A., Perona, P. and Anderson, D. J. (2014). Tachykinin-expressing neurons control male-specific aggressive arousal in Drosophila. Cell 156: 221-235. PubMed ID: 24439378

    Inagaki, H. K., Jung, Y., Hoopfer, E. D., Wong, A. M., Mishra, N., Lin, J. Y., Tsien, R. Y. and Anderson, D. J. (2013). Optogenetic control of Drosophila using a red-shifted channelrhodopsin reveals experience-dependent influences on courtship. Nat Methods. PubMed ID: 24363022

  • István Andó Biological Research Centre, Hungarian Academy of Sciences, Szeged
    Hultmark, D. and Ando, I. (2022). Hematopoietic plasticity mapped in Drosophila and other insects. Elife 11. PubMed ID: 35920811

    Cinege, G., Magyar, L. B., Kovacs, A. L., Lerner, Z., Juhasz, G., Lukacsovich, D., Winterer, J., Lukacsovich, T., Hegedus, Z., Kurucz, E., Hultmark, D., Foldy, C. and Ando, I. (2021). Broad Ultrastructural and Transcriptomic Changes Underlie the Multinucleated Giant Hemocyte Mediated Innate Immune Response against Parasitoids. J Innate Immun: 1-20. PubMed ID: 34864742

    Saito, T., Chiku, T., Oka, M., Wada-Kakuda, S., Nobuhara, M., Oba, T., Shinno, K., Abe, S., Asada, A., Sumioka, A., Takashima, A., Miyasaka, T. and Ando, K. (2021). Disulfide bond formation in microtubule-associated tau protein promotes tau accumulation and toxicity in vivo. Hum Mol Genet. PubMed ID: 34137825

    Balog, J., Honti, V., Kurucz, E., Kari, B., Pusas, L. G., Ando, I. and Szebeni, G. J. (2021). Immunoprofiling of Drosophila Hemocytes by Single-cell Mass Cytometry. Genomics Proteomics Bioinformatics. PubMed ID: 33713850

    Saito, T., Oba, T., Shimizu, S., Asada, A., Iijima, K. M. and Ando, K. (2019). Cdk5 increases MARK4 activity and augments pathological tau accumulation and toxicity through tau phosphorylation at Ser262. Hum Mol Genet. PubMed ID: 31174206

    Markus, R., Lerner, Z., Honti, V., Csordas, G., Zsamboki, J., Cinege, G., Parducz, A., Lukacsovich, T., Kurucz, E. and Ando, I. (2015). Multinucleated giant hemocytes are effector cells in cell-mediated immune responses of Drosophila. J Innate Immun [Epub ahead of print]. PubMed ID: 25659341

    Csordas, G., Varga, G. I., Honti, V., Jankovics, F., Kurucz, E. and Ando, I. (2014). In vivo immunostaining of hemocyte compartments in Drosophila for live imaging. PLoS One 9: e98191. PubMed ID: 24892745

    Honti, V., Cinege, G., Csordas, G., Kurucz, E., Zsamboki, J., Evans, C. J., Banerjee, U. and Ando, I. (2013). Variation of NimC1 expression in Drosophila stocks and transgenic strains. Fly (Austin) 7. PubMed ID: 23899817

    Honti, V., Csordas, G., Kurucz, E., Markus, R. and Ando, I. (2013). The cell-mediated immunity of Drosophila melanogaster; hemocyte lineages, immune compartments, microanatomy and regulation. Dev Comp Immunol. PubMed ID: 23800719

    Miyauchi, C., Kitazawa, D., Ando, I., Hayashi, D. and Inoue, Y. H. (2013). Orbit/CLASP is required for germline cyst formation through its developmental control of fusomes and ring canals in Drosophila males. PLoS One 8: e58220. PubMed ID: 23520495

  • Kanae Ando Department of Biological Sciences,Tokyo Metropolitan University
    Oka, M., Suzuki, E., Asada, A., Saito, T., Iijima, K. M. and Ando, K. (2021). Increasing neuronal glucose uptake attenuates brain aging and promotes life span under dietary restriction in Drosophila. iScience 24(1): 101979. PubMed ID: 33490892

    Chiku, T., Hayashishita, M., Saito, T., Oka, M., Shinno, K., Ohtake, Y., Shimizu, S., Asada, A., Hisanaga, S. I., Iijima, K. M. and Ando, K. (2018). S6K/p70S6K1 protects against tau-mediated neurodegeneration by decreasing the level of tau phosphorylated at Ser262 in a Drosophila model of tauopathy. Neurobiol Aging 71: 255-264. PubMed ID: 30172839

    Sekiya, M., Wang, M., Fujisaki, N., Sakakibara, Y., Quan, X., Ehrlich, M. E., De Jager, P. L., Bennett, D. A., Schadt, E. E., Gandy, S., Ando, K., Zhang, B. and Iijima, K. M. (2018). Integrated biology approach reveals molecular and pathological interactions among Alzheimer's Abeta42, Tau, TREM2, and TYROBP in Drosophila models. Genome Med 10(1): 26. PubMed ID: 29598827

    Oka, M., Fujisaki, N., Maruko-Otake, A., Ohtake, Y., Shimizu, S., Saito, T., Hisanaga, S. I., Iijima, K. M. and Ando, K. (2017). Ca2+/calmodulin-dependent protein kinase II promotes neurodegeneration caused by tau phosphorylated at Ser262/356 in a transgenic Drosophila model of tauopathy. J Biochem 162(5): 335-342. PubMed ID: 28992057

  • Deborah Andrew Department of Cell Biology, Johns Hopkins University School of Medicine
    Loganathan, R., Levings, D. C., Kim, J. H., Wells, M. B., Chiu, H., Wu, Y., Slattery, M. and Andrew, D. J. (2022). Ribbon boosts ribosomal protein gene expression to coordinate organ form and function. J Cell Biol 221(4). PubMed ID: 35195669
    Chung, S., Le, T. P., Vishwakarma, V., Cheng, Y. L. and Andrew, D. J. (2021). Isoform-specific roles of the Drosophila filamin-type protein Jitterbug (Jbug) during development. Genetics. PubMed ID: 34173831

    Kim, J. H., Hanlon, C. D., Vohra, S., Devreotes, P. N. and Andrew, D. J. (2021). Hedgehog signaling and Tre1 regulate actin dynamics through PI(4,5)P(2) to direct migration of Drosophila embryonic germ cells. Cell Rep 34(9): 108799. PubMed ID: 33657369

    Johnson, D. M., Wells, M. B., Fox, R., Lee, J. S., Loganathan, R., Levings, D., Bastien, A., Slattery, M. and Andrew, D. J. (2020). CrebA increases secretory capacity through direct transcriptional regulation of the secretory machinery, a subset of secretory cargo, and other key regulators. Traffic. PubMed ID: 32613751

    Johnson, D. M. and Andrew, D. J. (2019). Role of tbc1 in Drosophila embryonic salivary glands. BMC Mol Cell Biol 20(1): 19. PubMed ID: 31242864

    Hanlon, C.D. and Andrew, D.J. (2016). Drosophila FoxL1 non-autonomously coordinates organ placement during embryonic development. Dev Biol [Epub ahead of print]. PubMed ID: 27618755

    Loganathan, R., Lee, J. S., Wells, M. B., Grevengoed, E., Slattery, M. and Andrew, D. J. (2015). Ribbon regulates morphogenesis of the Drosophila embryonic salivary gland through transcriptional activation and repression. Dev Biol [Epub ahead of print]. PubMed ID: 26477561

    Hanlon, C. D. and Andrew, D. J. (2015). Outside-in signaling - a brief review of GPCR signaling with a focus on the Drosophila GPCR family. J Cell Sci [Epub ahead of print]. PubMed ID: 26345366

    Fox, R. M. and Andrew, D. J. (2015). Changes in organelle position and epithelial architecture associated with loss of CrebA. Biol Open [Epub ahead of print]. PubMed ID: 25681391

    Chung, S., Hanlon, C. D. and Andrew, D. J. (2014). 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 Interdiscip Rev Dev Biol 3: 281-300. PubMed ID: 25208491

    Chung, S. and Andrew, D. J. (2014). Cadherin 99C regulates apical expansion and cell rearrangement during epithelial tube elongation. Development. PubMed ID: 24718992

  • Luiz Andrioli School of Arts, Sciences and Humanities, University of Sao Paulo, Brazil
    Baltruk, L. J., Lavezzo, G. M., Machado-Lima, A., Digiampietri, L. A. and Andrioli, L. P. (2022). An additive repression mechanism sets the anterior limits of anterior pair-rule stripes 1. Cells Dev 171: 203802. PubMed ID: 35934285

    Uliana, J. V. C., Brancini, G. T. P., Hombria, J. C., Digiampietri, L. A., Andrioli, L. P. and Monesi, N. (2018). Characterizing the embryonic development of B. hygida (Diptera: Sciaridae) following enzymatic treatment to permeabilize the serosal cuticle. Mech Dev 154: 270-276. PubMed ID: 30081091

    Andrioli, L. P., Dos Santos, W. S., Aguiar, F. D. S. and Digiampietri, L. A. (2017). Repression activity of Tailless on h 1 and eve 1 pair-rule stripes. Mech Dev 144(Pt B): 156-162. PubMed ID: 27773632

  • Robert Anholt North Carolina State University: Environmental Health
    MacPherson, R. A., Shankar, V., Anholt, R. R. H. and Mackay, T. F. C. (2023). Genetic and Genomic Analyses of Drosophila melanogaster Models of Chromatin Modification Disorders. Genetics. PubMed ID: 37036413

    Mokashi, S. S., Shankar, V., Johnstun, J. A., Mackay, T. F. C. and Anholt, R. R. H. (2022). Pleiotropic Fitness Effects of a Drosophila Odorant-binding Protein. G3 (Bethesda). PubMed ID: 36454098

    Morozova, T. V., Shankar, V., MacPherson, R. A., Mackay, T. F. C. and Anholt, R. R. H. (2022). Modulation of the Drosophila transcriptome by developmental exposure to alcohol. BMC Genomics 23(1): 347. PubMed ID: 35524193

    Huggett, S. B., Hatfield, J. S., Walters, J. D., McGeary, J. E., Welsh, J. W., Mackay, T. F. C., Anholt, R. R. H. and Palmer, R. H. C. (2021). Ibrutinib as a potential therapeutic for cocaine use disorder. Transl Psychiatry 11(1): 623. PubMed ID: 34880215

    Ozsoy, E. D., Yilmaz, M., Patlar, B., Emecen, G., Durmaz, E., Magwire, M. M., Zhou, S., Huang, W., Anholt, R. R. H. and Mackay, T. F. C. (2021). Epistasis for head morphology in Drosophila melanogaster. G3 (Bethesda) 11(10). PubMed ID: 34568933

    Mokashi, S. S., Shankar, V., MacPherson, R. A., Hannah, R. C., Mackay, T. F. C. and Anholt, R. R. H. (2021). Developmental Alcohol Exposure in Drosophila: Effects on Adult Phenotypes and Gene Expression in the Brain. Front Psychiatry 12: 699033. PubMed ID: 34366927

    Walters, J. D., Hatfield, J. S., Baker, B. B., Mackay, T. F. C. and Anholt, R. R. H. (2021). A High Throughput Microplate Feeder Assay for Quantification of Consumption in Drosophila. J Vis Exp(172). PubMed ID: 34180893

    Johnstun, J. A., Shankar, V., Mokashi, S. S., Sunkara, L. T., Ihearahu, U. E., Lyman, R. L., Mackay, T. F. C. and Anholt, R. R. H. (2021). Functional Diversification, Redundancy and Epistasis among Paralogs of the Drosophila melanogaster Obp50a-d Gene Cluster. Mol Biol Evol. PubMed ID: 33560417

    Huang, W., Carbone, M. A., Lyman, R. F., Anholt, R. R. H. and Mackay, T. F. C. (2020). Genotype by environment interaction for gene expression in Drosophila melanogaster. Nat Commun 11(1): 5451. PubMed ID: 33116142

    Highfill, C. A., Baker, B. M., Stevens, S. D., Anholt, R. R. H. and Mackay, T. F. C. (2019). Genetics of cocaine and methamphetamine consumption and preference in Drosophila melanogaster. PLoS Genet 15(5): e1007834. PubMed ID: 31107875

    Zhou, S., Luoma, S. E., St Armour, G. E., Thakkar, E., Mackay, T. F. C. and Anholt, R. R. H. (2017). A Drosophila model for toxicogenomics: Genetic variation in susceptibility to heavy metal exposure. PLoS Genet 13(7): e1006907. PubMed ID: 28732062

    Garcia, J. F., Carbone, M. A., Mackay, T. F. C. and Anholt, R. R. H. (2017). Regulation of Drosophila lifespan by bellwether promoter alleles. Sci Rep 7(1): 4109. PubMed ID: 28646164

    Shorter, J. R., Dembeck, L. M., Everett, L. J., Morozova, T. V., Arya, G. H., Turlapati, L., St Armour, G. E., Schal, C., Mackay, T. F. and Anholt, R. R. (2016). Obp56h Modulates Mating Behavior in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 27558663

  • Christophe Antoniewski Drosophila Genetics and Epigenetics, Centre national de la recherche scientifique, Paris
    van den Beek, M., da Silva, B., Pouch, J., Ali Chaouche, M. E. A., Carre, C. and Antoniewski, C. (2018). Dual-layer transposon repression in heads of Drosophila melanogaster. RNA. PubMed ID: 30217866

    Besnard-Guerin, C., Jacquier, C., Pidoux, J., Deddouche, S. and Antoniewski, C. (2015). The cricket paralysis virus suppressor inhibits microRNA silencing mediated by the Drosophila Argonaute-2 protein. PLoS One 10: e0120205. PubMed ID: 25793377

    Molla-Herman, A., Vallés, A.M., Ganem-Elbaz, C., Antoniewski, C. and Huynh, J.R. (2015). tRNA processing defects induce replication stress and Chk2-dependent disruption of piRNA transcription. EMBO J [Epub ahead of print]. PubMed ID: 26471728

    Besnard-Guerin, C., Jacquier, C., Pidoux, J., Deddouche, S. and Antoniewsk, C. (2015). The cricket paralysis virus suppressor inhibits microRNA silencing mediated by the Drosophila Argonaute-2 protein. PLoS One 10: e0120205. PubMed ID: 25793377

    Carre, C., Jacquier, C., Bouge, A. L., de Chaumont, F., Besnard-Guerin, C., Thomassin, H., Pidoux, J., Da Silva, B., Chalatsi, E., Zahra, S., Olivo-Marin, J. C., Munier-Lehmann, H. and Antoniewski, C. (2013). AutomiG, a Biosensor to Detect Alterations in miRNA Biogenesis and in Small RNA Silencing Guided by Perfect Target Complementarity. PLoS One 8: e74296. PubMed ID: 24019960

    Reinhardt, A., Feuillette, S., Cassar, M., Callens, C., Thomassin, H., Birman, S., Lecourtois, M., Antoniewski, C. and Tricoire, H. (2012). Lack of miRNA Misregulation at Early Pathological Stages in Drosophila Neurodegenerative Disease Models. Front Genet 3: 226. PubMed ID: 23115562

    de Vanssay, A., Bouge, A. L., Boivin, A., Hermant, C., Teysset, L., Delmarre, V., Antoniewski, C. and Ronsseray, S. (2012). Paramutation in Drosophila linked to emergence of a piRNA-producing locus. Nature 490: 112-115. PubMed ID: 22922650

  • Yiorgos Apidianakis Department of Biological Sciences, University of Cyprus,
    Shianiou, G., Teloni, S. and Apidianakis, Y. (2023). Intestinal Immune Deficiency and Juvenile Hormone Signaling Mediate a Metabolic Trade-off in Adult Drosophila Females. Metabolites 13(3). PubMed ID: 36984780

    Kotronarou, K., Charalambous, A., Evangelou, A., Georgiou, O., Demetriou, A. and Apidianakis, Y. (2023). Dietary Stimuli, Intestinal Bacteria and Peptide Hormones Regulate Female Drosophila Defecation Rate. Metabolites 13(2). PubMed ID: 36837883 Kapsetaki, S. E., Tzelepis, I., Avgousti, K., Livadaras, I., Garantonakis, N., Varikou, K. and Apidianakis, Y. (2014). The bacterial metabolite 2-aminoacetophenone promotes association of pathogenic bacteria with flies. Nat Commun 5: 4401. PubMed ID: 25043228

    Righi, V., Apidianakis, Y., Psychogios, N., Rahme, L. G., Tompkins, R. G. and Tzika, A. A. (2014). In vivo high-resolution magic angle spinning proton NMR spectroscopy of Drosophila melanogaster flies as a model system to investigate mitochondrial dysfunction in Drosophila GST2 mutants. Int J Mol Med 34: 327-333. PubMed ID: 24789699

    Panayidou, S., Ioannidou, E. and Apidianakis, Y. (2014). Human pathogenic bacteria, fungi, and viruses in Drosophila: disease modeling, lessons, and shortcomings. Virulence 5: 253-269. PubMed ID: 24398387

    Christofi, T. and Apidianakis, Y. (2013). Drosophila immune priming against Pseudomonas aeruginosa is short-lasting and depends on cellular and humoral immunity. F1000Res 2: 76. PubMed ID: 24358857

  • Derek Applewhite Department of Biology, Reed College, Portland, OR
    Zhao, A. J., Montes-Laing, J., Perry, W. M. G., Shiratori, M., Merfeld, E., Rogers, S. L. and Applewhite, D. A. (2022). The Drosophila spectraplakin Short stop regulates focal adhesion dynamics by cross-linking microtubules and actin. Mol Biol Cell 33(5): ar19. PubMed ID: 35235367

    Platenkamp, A., Detmar, E., Sepulveda, L., Ritz, A., Rogers, S. L. and Applewhite, D. A. (2020). The Drosophila melanogaster Rab GAP RN-tre cross-talks with the Rho1 signaling pathway to regulate nonmuscle myosin II localization and function. Mol Biol Cell 31(21): 2379-2397. PubMed ID: 32816624

    O'Connell, M. E., Sridharan, D., Driscoll, T., Krishnamurthy, I., Perry, W. G. and Applewhite, D. A. (2019). The Drosophila protein, Nausicaa, regulates lamellipodial actin dynamics in a Cortactin-dependent manner. Biol Open 8(6). PubMed ID: 31164339

    Harker, A. J., Katkar, H. H., Bidone, T. C., Aydin, F., Voth, G. A., Applewhite, D. A. and Kovar, D. R. (2019). Ena/VASP processive elongation is modulated by avidity on actin filaments bundled by the filopodia cross-linker fascin. Mol Biol Cell 30(7): 851-862. PubMed ID: 30601697

  • Charles Aquadro Department of Molecular Biology and Genetics, Cornell University
    Bubnell, J. E., Ulbing, C. K. S., Fernandez Begne, P. and Aquadro, C. F. (2022). Functional divergence of the bag of marbles gene in the Drosophila melanogaster species group. Mol Biol Evol. PubMed ID: 35714266

    Bauer DuMont, V. L., White, S. L., Zinshteyn, D. and Aquadro, C. F. (2021). Molecular population genetics of Sex-lethal (Sxl) in the Drosophila melanogaster species group: a locus that genetically interacts with Wolbachia pipientis in Drosophila melanogaster. G3 (Bethesda) 11(8). PubMed ID: 34849818

    Bubnell, J. E., Fernandez-Begne, P., Ulbing, C. K. S. and Aquadro, C. F. (2021). Diverse wMel variants of Wolbachia pipientis differentially rescue fertility and cytological defects of the bag of marbles partial loss of function mutation in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 34580706

    Choi, J. Y. and Aquadro, C. F. (2015). Molecular Evolution of Drosophila Germline Stem Cell and Neural Stem Cell Regulating Genes. Genome Biol Evol. PubMed ID: 26507797

    Choi, J. Y., Bubnell, J. E. and Aquadro, C. F. (2015). Population genomics of infectious and integrated Wolbachia pipientis genomes in Drosophila ananassae. Genome Biol Evol 7: 2362-2382. PubMed ID: 26254486

    Rebeiz, M., Pool, J. E., Kassner, V. A., Aquadro, C. F. and Carroll, S. B. (2009). Stepwise modification of a modular enhancer underlies adaptation in a Drosophila population. Science 326(5960): 1663-7. PubMed ID: 20019281

  • Eli Arama Department of Molecular Genetics, Weizmann Institute
    Tarayrah-Ibraheim, L., Maurice, E. C., Hadary, G., Ben-Hur, S., Kolpakova, A., Braun, T., Peleg, Y., Yacobi-Sharon, K. and Arama, E. (2021). DNase II mediates a parthanatos-like developmental cell death pathway in Drosophila primordial germ cells. Nat Commun 12(1): 2285. PubMed ID: 33863891

    Gorelick-Ashkenazi, A., Weiss, R., Sapozhnikov, L., Florentin, A., Tarayrah-Ibraheim, L., Dweik, D., Yacobi-Sharon, K. and Arama, E. (2018). Caspases maintain tissue integrity by an apoptosis-independent inhibition of cell migration and invasion. Nat Commun 9(1): 2806. PubMed ID: 30022065

    Aram, L., Braun, T., Braverman, C., Kaplan, Y., Ravid, L., Levin-Zaidman, S. and Arama, E. (2016). A Krebs cycle component limits caspase activation rate through mitochondrial surface restriction of CRL activation. Dev Cell 37: 15-33. PubMed ID: 27052834

    Politi, Y., Gal, L., Kalifa, Y., Ravid, L., Elazar, Z. and Arama, E. (2014). Paternal mitochondrial destruction after fertilization is mediated by a common endocytic and autophagic pathway in Drosophila. Dev Cell 29: 305-320. PubMed ID: 24823375

    Yacobi-Sharon, K., Namdar, Y., Arama, E. (2013) Alternative Germ Cell Death Pathway in Drosophila Involves HtrA2/Omi, Lysosomes, and a Caspase-9 Counterpart. Dev Cell. PubMed ID: 23523076

    Bader, M., Arama, E. and Steller, H. (2010). A novel F-box protein is required for caspase activation during cellular remodeling in Drosophila. Development 137: 1679-1688. PubMed ID: 20392747

    Olswang-Kutz, Y., Gertel, Y., Benjamin, S., Sela, O., Pekar, O., Arama, E., Steller, H., Horowitz, M. and Segal, D. (2009). Drosophila Past1 is involved in endocytosis and is required for germline development and survival of the adult fly. J Cell Sci 122: 471-480. PubMed ID: 19174465

  • Sofia Araújo IRB Barcelona Institute for Research in Biomedicine
    Ricolo, D. and Araujo, S. J. (2020). Coordinated crosstalk between microtubules and actin by a spectraplakin regulates lumen formation and branching. Elife 9. PubMed ID: 33112231

    Negreiros, E., Herszterg, S., Hwa, K., Camara, A., Dias, W. B., Carneiro, K., Bier, E., Todeschini, A. and Araujo, H. (2018). N-linked glycosylation restricts the function of short gastrulation to bind and shuttle BMPs. Development. PubMed ID: 30355725

    Ozturk-Colak, A., Stephan-Otto Attolini, C., Casanova, J. and Araujo, S. J. (2018). Blimp-1 mediates tracheal lumen maturation in Drosophila melanogaster. Genetics. PubMed ID: 30082278

    Cardoso, M. A., Fontenele, M., Lim, B., Bisch, P. M., Shvartsman, S. Y. and Araujo, H. M. (2017). A novel function for the IkappaB inhibitor Cactus in promoting Dorsal nuclear localization and activity in the Drosophila embryo. Development 144(16): 2907-2913. PubMed ID: 28705899

    Barrecheguren, P. J., Ros, O., Cotrufo, T., Kunz, B., Soriano, E., Ulloa, F., Stoeckli, E. T. and Araujo, S. J. (2016). SNARE proteins play a role in motor axon guidance in vertebrates and invertebrates. Dev Neurobiol [Epub ahead of print]. PubMed ID: 28033683

    Ricolo, D., Deligiannaki, M., Casanova, J. and Araújo, S.J. (2016). Centrosome amplification increases single-cell branching in post-mitotic cells. Curr Biol [Epub ahead of print]. PubMed ID: 27693136

    Ozturk-Colak, A., Moussian, B., Araujo, S. J. and Casanova, J. (2016). A feedback mechanism converts individual cell features into a supracellular ECM structure in trachea. Elife 5. PubMed ID: 26836303

    Ozturk-Colak, A., Moussian, B. and Araujo, S. J. (2015). Drosophila chitinous aECM and its cellular interactions during tracheal development. Dev Dyn [Epub ahead of print]. PubMed ID: 26442625

    Ricolo, D., Buti, E. and Araujo, S. J. (2015). Drosophila melanogaster Hedgehog cooperates with Frazzled to guide axons through a non-canonical signalling pathway. Mech Dev. PubMed ID: 25936631

    Buti, E., Mesquita, D. and Araujo, S. J. (2014). Hedgehog Is a Positive Regulator of FGF Signalling during Embryonic Tracheal Cell Migration. PLoS One 9: e92682. PubMed ID: 24651658

  • Alexei Aravin Division of Biology and Biological Engineering, Cal Tech
    Chen, P. and Aravin, A. A. (2023). Genetic control of a sex-specific piRNA program. Curr Biol. PubMed ID: 37059098

    Chen, P., Luo, Y. and Aravin, A. A. (2021). RDC complex executes a dynamic piRNA program during Drosophila spermatogenesis to safeguard male fertility. PLoS Genet 17(9): e1009591. PubMed ID: 34473737

    Huang, X., Hu, H., Webster, A., Zou, F., Du, J., Patel, D. J., Sachidanandam, R., Toth, K. F., Aravin, A. A. and Li, S. (2021). Binding of guide piRNA triggers methylation of the unstructured N-terminal region of Aub leading to assembly of the piRNA amplification complex. Nat Commun 12(1): 4061. PubMed ID: 34210982

    Chen, P., Kotov, A. A., Godneeva, B. K., Bazylev, S. S., Olenina, L. V. and Aravin, A. A. (2021). piRNA-mediated gene regulation and adaptation to sex-specific transposon expression in D. melanogaster male germline. Genes Dev. PubMed ID: 33985970

    Luo, Y., Fefelova, E., Ninova, M., Chen, Y. A. and Aravin, A. A. (2020). Repression of interrupted and intact rDNA by the SUMO pathway in Drosophila melanogaster. Elife 9. PubMed ID: 33164748

    Ninova, M., Godneeva, B., Chen, Y. A., Luo, Y., Prakash, S. J., Jankovics, F., Erdelyi, M., Aravin, A. A. and Fejes Toth, K. (2020). The SUMO Ligase Su(var)2-10 Controls Hetero- and Euchromatic Gene Expression via Establishing H3K9 Trimethylation and Negative Feedback Regulation. Mol Cell 77(3): 571-585. PubMed ID: 31901448

    Ninova, M., Chen, Y. A., Godneeva, B., Rogers, A. K., Luo, Y., Fejes Toth, K. and Aravin, A. A. (2020). Su(var)2-10 and the SUMO Pathway Link piRNA-Guided Target Recognition to Chromatin Silencing. Mol Cell 77(3): 556-570.e556. PubMed ID: 31901446

    Kotov, A. A., Adashev, V. E., Godneeva, B. K., Ninova, M., Shatskikh, A. S., Bazylev, S. S., Aravin, A. A. and Olenina, L. V. (2019). piRNA silencing contributes to interspecies hybrid sterility and reproductive isolation in Drosophila melanogaster. Nucleic Acids Res. PubMed ID: 30788506

    Ciabrelli, F., Comoglio, F., Fellous, S., Bonev, B., Ninova, M., Szabo, Q., Xuereb, A., Klopp, C., Aravin, A., Paro, R., Bantignies, F. and Cavalli, G. (2017). Stable Polycomb-dependent transgenerational inheritance of chromatin states in Drosophila. Nat Genet 49(6): 876-886. PubMed ID: 28436983

    Chen, Y. A., Stuwe, E., Luo, Y., Ninova, M., Le Thomas, A., Rozhavskaya, E., Li, S., Vempati, S., Laver, J. D., Patel, D. J., Smibert, C. A., Lipshitz, H. D., Fejes Toth, K. and Aravin, A. A. (2016). Cutoff Suppresses RNA Polymerase II Termination to Ensure Expression of piRNA Precursors. Mol Cell. PubMed ID: 27292797

    Hur, J.K., Luo, Y., Moon, S., Ninova, M., Marinov, G.K., Chung, Y.D. and Aravin, A.A. (2016). Splicing-independent loading of TREX on nascent RNA is required for efficient expression of dual-strand piRNA clusters in Drosophila. Genes Dev 30: 840-855. PubMed ID: 27036967

    Webster, A., Li, S., Hur, J.K., Wachsmuth, M., Bois, J.S., Perkins, E.M., Patel, D.J. and Aravin, A.A. (2015). Aub and Ago3 are recruited to nuage through two mechanisms to form a ping-pong complex assembled by Krimper. Mol Cell 59: 564-575. PubMed ID: 26295961

  • Michelle Arbeitman College of Medicine, Florida State University, Tallahassee, FL
    Palmateer, C. M., Artikis, C., Brovero, S. G., Friedman, B., Gresham, A. and Arbeitman, M. N. (2023). Single-cell transcriptome profiles of Drosophila fruitless-expressing neurons from both sexes. Elife 12. PubMed ID: 36724009

    Palmateer, C. M., Moseley, S. C., Ray, S., Brovero, S. G. and Arbeitman, M. N. (2021). Analysis of cell-type-specific chromatin modifications and gene expression in Drosophila neurons that direct reproductive behavior. PLoS Genet 17(4): e1009240. PubMed ID: 33901168

    Brovero, S. G., Fortier, J. C., Hu, H., Lovejoy, P. C., Newell, N. R., Palmateer, C. M., Tzeng, R. Y., Lee, P. T., Zinn, K. and Arbeitman, M. N. (2021). Investigation of Drosophila fruitless neurons that express Dpr/DIP cell adhesion molecules. Elife 10. PubMed ID: 33616528

    Newell, N. R., Ray, S., Dalton, J. E., Fortier, J. C., Kao, J. Y., Chang, P. L., Nuzhdin, S. V. and Arbeitman, M. N. (2020). The Drosophila post-mating response: Gene expression and behavioral changes reveal perdurance and variation in cross-tissue interactions. G3 (Bethesda). PubMed ID: 31907222

    Rice, G. R., Barmina, O., Luecke, D., Hu, K., Arbeitman, M. and Kopp, A. (2019). Modular tissue-specific regulation of doublesex underpins sexually dimorphic development in Drosophila. Development 146(14). PubMed ID: 31285355

    Graze, R. M., Tzeng, R. Y., Howard, T. S. and Arbeitman, M. N. (2018). Perturbation of IIS/TOR signaling alters the landscape of sex-differential gene expression in Drosophila. BMC Genomics 19(1): 893. PubMed ID: 30526477

    Newell, N.R., New, F.N., Dalton, J.E., McIntyre, L.M. and Arbeitman, M.N. (2016). Neurons that underlie Drosophila melanogaster reproductive behaviors: detection of a large male-bias in gene expression in fruitless-expressing neurons. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27247289

    Arbeitman, M. N., New, F., Fear, J. M., Howard, T. S., Dalton, J. E. and Graze, R. M. (2016). Sex differences in Drosophila somatic gene expression: Variation and regulation by Doublesex. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27172187

    Dalton, J. E., Fear, J. M., Knott, S., Baker, B. S., McIntyre, L. M. and Arbeitman, M. N. (2013). Male-specific fruitless isoforms have different regulatory roles conferred by distinct zinc finger DNA binding domains. BMC Genomics 14: 659. PubMed ID: 24074028

    Winbush, A., Reed, D., Chang, P. L., Nuzhdin, S. V., Lyons, L. C. and Arbeitman, M. N. (2012). Identification of gene expression changes associated with long-term memory of courtship rejection in Drosophila males. G3 (Bethesda) 2: 1437-1445. PubMed ID: 23173095

  • Vincent Archambault Institute for Research in Immunology and Cancer, University of Montreal
    Emond-Fraser, V., Larouche, M., Kubiniok, P., Bonneil, É., Li, J., Bourouh, M., Frizzi, L., Thibault, P. and Archambault, V. (2023). Identification of PP2A-B55 targets uncovers regulation of emerin during nuclear envelope reassembly in Drosophila. Open Biol 13(7): 230104. PubMed ID: 37463656

    Garrido, D., Bourouh, M., Bonneil, É., Thibault, P., Swan, A. and Archambault, V. (2020). Cyclin B3 activates the Anaphase-Promoting Complex/Cyclosome in meiosis and mitosis. PLoS Genet 16(11): e1009184. PubMed ID: 33137813

    Mehsen, H., Boudreau, V., Garrido, D., Bourouh, M., Larouche, M., Maddox, P. S., Swan, A. and Archambault, V. (2018). PP2A-B55 promotes nuclear envelope reformation after mitosis in Drosophila. J Cell Biol. PubMed ID: 30309980

    Kachaner, D., Garrido, D., Mehsen, H., Normandin, K., Lavoie, H. and Archambault, V. (2017). Coupling of Polo kinase activation to nuclear localization by a bifunctional NLS is required during mitotic entry. Nat Commun 8(1): 1701. PubMed ID: 29167465

    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

    Kachaner, D., Pinson, X., El Kadhi, K. B., Normandin, K., Talje, L., Lavoie, H., Lepine, G., Carreno, S., Kwok, B. H., Hickson, G. R. and Archambault, V. (2014). Interdomain allosteric regulation of Polo kinase by Aurora B and Map205 is required for cytokinesis. J Cell Biol 207: 201-211. PubMed ID: 25332165

    Lipinszki, Z., Wang, P., Grant, R., Lindon, C., Dzhindzhev, N. S., D'Avino, P. P., Przewloka, M. R., Glover, D. M. and Archambault, V. (2014). Affinity purification of protein complexes from Drosophila embryos in cell cycle studies. Methods Mol Biol 1170: 571-588. PubMed ID: 24906338

    Wang, P., Pinson, X. and Archambault, V. (2011). PP2A-twins is antagonized by greatwall and collaborates with polo for cell cycle progression and centrosome attachment to nuclei in drosophila embryos. PLoS Genet 7: e1002227. PubMed ID: 21852958

    Montembault, E., Zhang, W., Przewloka, M. R., Archambault, V., Sevin, E. W., Laue, E. D., Glover, D. M. and D'Avino, P. P. (2010). Nessun Dorma, a novel centralspindlin partner, is required for cytokinesis in Drosophila spermatocytes. J Cell Biol 191: 1351-1365. PubMed ID: 21187330

    Blagden, S. P., Gatt, M. K., Archambault, V., Lada, K., Ichihara, K., Lilley, K. S., Inoue, Y. H. and Glover, D. M. (2009). Drosophila Larp associates with poly(A)-binding protein and is required for male fertility and syncytial embryo development. Dev Biol 334: 186-197. PubMed ID: 19631203

  • Roman Arguello Department of Ecology and Evolution, Universite de Lausanne
    Auer, T. O., Alvarez-Ocana, R., Cruchet, S., Benton, R. and Arguello, J. R. (2022). Copy number changes in co-expressed odorant receptor genes enable selection for sensory differences in drosophilid species. Nat Ecol Evol 6(9): 1343-1353. PubMed ID: 35864227

    Arguello, J. R., Laurent, S. and Clark, A. G. (2021). Corrigendum to: Demographic history of the human commensal Drosophila melanogaster. Genome Biol Evol 13(3). PubMed ID: 33763690

    Arguello, J. R., Abuin, L., Armida, J., Mika, K., Chai, P. C. and Benton, R. (2021). Targeted molecular profiling of rare olfactory sensory neurons identifies fate, wiring, and functional determinants. Elife 10. PubMed ID: 33666172

    Auer, T. O., Khallaf, M. A., Silbering, A. F., Zappia, G., Ellis, K., Alvarez-Ocana, R., Arguello, J. R., Hansson, B. S., Jefferis, G., Caron, S. J. C., Knaden, M. and Benton, R. (2020). Olfactory receptor and circuit evolution promote host specialization. Nature 579(7799): 402-408. PubMed ID: 32132713

  • Alexey Arkov Murray State University, Murray, Kentucky
    Tindell, S. J., Rouchka, E. C. and Arkov, A. L. (2020). Glial granules contain germline proteins in the Drosophila brain, which regulate brain transcriptome. Commun Biol 3(1): 699. PubMed ID: 33219296

    Vo, H. D. L., Wahiduzzaman, Tindell, S. J., Zheng, J., Gao, M. and Arkov, A. L. (2019). Protein components of ribonucleoprotein granules from Drosophila germ cells oligomerize and show distinct spatial organization during germline development. Sci Rep 9(1): 19190. PubMed ID: 31844131

    Gao, M., Thomson, T. C., Creed, T. M., Tu, S., Loganathan, S. N., Jackson, C. A., McCluskey, P., Lin, Y., Collier, S. E., Weng, Z., Lasko, P., Ohi, M. D. and Arkov, A. L. (2015). Glycolytic enzymes localize to ribonucleoprotein granules in Drosophila germ cells, bind Tudor and protect from transposable elements. EMBO Rep. PubMed ID: 25600116

    Gao, M. and Arkov, A. L. (2012). Next generation organelles: Structure and role of germ granules in the germline. Mol Reprod Dev 80(8):610-23. PubMed ID: 23011946

    Creed, T. M., Loganathan, S. N., Varonin, D., Jackson, C. A. and Arkov, A. L. (2010). Novel role of specific Tudor domains in Tudor-Aubergine protein complex assembly and distribution during Drosophila oogenesis. Biochem Biophys Res Commun 402: 384-389. PubMed ID: 20946872

  • Douglas Armstrong Institute for Adaptive and Neural Computation, Edinburgh
    Court, R., Costa, M., Pilgrim, C., Millburn, G., Holmes, A., McLachlan, A., Larkin, A., Matentzoglu, N., Kir, H., Parkinson, H., Brown, N. H., O'Kane, C. J., Armstrong, J. D., Jefferis, G. and Osumi-Sutherland, D. (2023). Virtual Fly Brain-An interactive atlas of the Drosophila nervous system. Front Physiol 14: 1076533. PubMed ID: 36776967

    Marescotti, M., Lagogiannis, K., Webb, B., Davies, R. W. and Armstrong, J. D. (2018). Monitoring brain activity and behaviour in freely moving Drosophila larvae using bioluminescence. Sci Rep 8(1): 9246. PubMed ID: 29915372

    Suslak, T. J., Watson, S., Thompson, K. J., Shenton, F. C., Bewick, G. S., Armstrong, J. D. and Jarman, A. P. (2015). Piezo is essential for amiloride-sensitive stretch-activated mechanotransduction in larval Drosophila dorsal bipolar dendritic sensory neurons. PLoS One 10: e0130969. PubMed ID: 26186008

    Green, E. W., O'Callaghan, E. K., Hansen, C. N., Bastianello, S., Bhutani, S., Vanin, S., Armstrong, J. D., Costa, R. and Kyriacou, C. P. (2015). Drosophila circadian rhythms in seminatural environments: Summer afternoon component is not an artifact and requires TrpA1 channels. Proc Natl Acad Sci U S A 112: 8702-8707. PubMed ID: 26124142

    Wessnitzer, J., Young, J. M., Armstrong, J. D. and Webb, B. (2012). A model of non-elemental olfactory learning in Drosophila. J Comput Neurosci 32: 197-212. PubMed ID: 21698405

    Young, J. M., Wessnitzer, J., Armstrong, J. D. and Webb, B. (2011). Elemental and non-elemental olfactory learning in Drosophila. Neurobiol Learn Mem 96: 339-352. PubMed ID: 21742045

    Russell, C., Wessnitzer, J., Young, J. M., Armstrong, J. D. and Webb, B. (2011). Dietary salt levels affect salt preference and learning in larval Drosophila. PLoS One 6: e20100. PubMed ID: 21687789

    Cachero, S., Simpson, T. I., Zur Lage, P. I., Ma, L., Newton, F. G., Holohan, E. E., Armstrong, J. D. and Jarman, A. P. (2011). The gene regulatory cascade linking proneural specification with differentiation in Drosophila sensory neurons. PLoS Biol 9: e1000568. PubMed ID: 21283833

  • Jennifer Armstrong Keck Science Department, Scripps College, Claremont McKenna College, Claremont, CA
    Kim, S., Bugga, L., Hong, E. S., Zabinsky, R., Edwards, R. G., Deodhar, P. A. and Armstrong, J. A. (2015). An RNAi-based candidate screen for modifiers of the CHD1 chromatin remodeler and assembly factor in Drosophila melanogaster. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26596648

    Bugga, L., McDaniel, I. E., Engie, L., Armstrong, J. A. (2013) The Drosophila melanogaster CHD1 Chromatin Remodeling Factor Modulates Global Chromosome Structure and Counteracts HP1a and H3K9me2. PLoS One 8: e59496. PubMed ID: 23533627

    McDaniel, I. E., Lee, J. M., Berger, M. S., Hanagami, C. K. and Armstrong, J. A. (2008). Investigations of CHD1 function in transcription and development of Drosophila melanogaster. Genetics 178: 583-587. PubMed ID: 18202396

  • David Arnosti Biochemistry & Molecular Biology, Michigan State University
    Raicu, A. M., Kadiyala, D., Niblock, M., Jain, A., Yang, Y., Bird, K. M., Bertholf, K., Seenivasan, A., Siddiq, M. and Arnosti, D. N. (2023). The cynosure of CtBP: evolution of a bilaterian transcriptional corepressor. Mol Biol Evol. PubMed ID: 36625090

    Payankaulam, S., Hickey, S. L. and Arnosti, D. N. (2021). Cell cycle expression of polarity genes features Rb targeting of Vang. Cells Dev 169: 203747. PubMed ID: 34583062

    Mouawad, R., Himadewi, P., Kadiyala, D. and Arnosti, D. N. (2020). Selective repression of the Drosophila cyclin B promoter by retinoblastoma and E2F proteins. Biochim Biophys Acta Gene Regul Mech 1863(7): 194549. PubMed ID: 32275964

    Mouawad, R., Prasad, J., Thorley, D., Himadewi, P., Kadiyala, D., Wilson, N., Kapranov, P. and Arnosti, D. N. (2019). Diversification of retinoblastoma protein function associated with cis and trans adaptations. Mol Biol Evol. PubMed ID: 31418797

    Wei, Y., Gokhale, R. H., Sonnenschein, A., Montgomery, K. M., Ingersoll, A. and Arnosti, D. N. (2016). Complex cis-regulatory landscape of the insulin receptor gene underlies the broad expression of a central signaling regulator. Development 143: 3591-3603. PubMed ID: 27702787

    Sayal, R., Dresch, J.M., Pushel, I., Taylor, B.R. and Arnosti, D.N. (2016). Quantitative perturbation-based analysis of gene expression predicts enhancer activity in early Drosophila embryo. Elife 5. PubMed ID: 27152947

    Payankaulam, S., Yeung, K., McNeill, H., Henry, R.W. and Arnosti, D.N. (2016). Regulation of cell polarity determinants by the Retinoblastoma tumor suppressor protein. Sci Rep 6: 22879. PubMed ID: 26971715

    Kok, K., Ay, A., Li, L. M. and Arnosti, D. N. (2015). Genome-wide errant targeting by Hairy. Elife 4. PubMed ID: 26305409

    Elenbaas, J. S., Mouawad, R., Henry, W. R., Arnosti, D. N. and Payankaulam, S. (2014). Role of Drosophila retinoblastoma protein instability element in cell growth and proliferation. Cell Cycle: [Epub ahead of print]. PubMed ID: 25496208

    Zhang, L., Wei, Y., Pushel, I., Heinze, K., Elenbaas, J., Henry, R. W. and Arnosti, D. N. (2014). Integrated Stability and Activity Control of the Drosophila Rbf1 Retinoblastoma Protein. J Biol Chem. PubMed ID: 25049232

    Suleimenov, Y., Ay, A., Abul Hassan Samee, M., Dresch, J. M., Sinha, S. and Arnosti, D. N. (2013). Global Parameter Estimation for Thermodynamic Models of Transcriptional Regulation. Methods. PubMed ID: 23726942

  • Kavita Arora Developmental & Cell Biology, School of Biological Sciences, University of California, Irvine
    Lujan, E., Bornemann, D. J., Rottig, C., Bayless, B. A., Stocker, H., Hafen, E., Arora, K. and Warrior, R. (2016). Analysis of novel alleles of brother of tout-velu, the Drosophila ortholog of human EXTL3 using a newly developed FRT42D ovoD chromosome. Genesis [Epub ahead of print]. PubMed ID: 27636555

    Kunnapuu, J., Tauscher, P. M., Tiusanen, N., Nguyen, M., Loytynoja, A., Arora, K. and Shimmi, O. (2014). Cleavage of the Drosophila screw prodomain is critical for a dynamic BMP morphogen gradient in embryogenesis. Dev Biol 389: 149-159. PubMed ID: 24560644

    Ellis, J. E., Parker, L., Cho, J. and Arora, K. (2010). Activin signaling functions upstream of Gbb to regulate synaptic growth at the Drosophila neuromuscular junction. Dev Biol 342: 121-133. PubMed ID: 20346940

  • Spyros Artavanis-Tsakonas Harvard Medical School
    Kankel, M. W., Sen, A., Lu, L., Theodorou, M., Dimlich, D. N., McCampbell, A., Henderson, C. E., Shneider, N. A. and Artavanis-Tsakonas, S. (2020). Amyotrophic Lateral Sclerosis Modifiers in Drosophila Reveal the Phospholipase D Pathway as a Potential Therapeutic Target. Genetics. PubMed ID: 32345615

    Stoiber, M. H., Olson, S., May, G. E., Duff, M. O., Manent, J., Obar, R., Guruharsha, K., Artavanis-Tsakonas, S., Brown, J. B., Graveley, B. R. and Celniker, S. E. (2015). Extensive cross-regulation of post-transcriptional regulatory networks in Drosophila. Genome Res [Epub ahead of print]. PubMed ID: 26294687

    Ho, D. M., Pallavi, S. K. and Artavanis-Tsakonas, S. (2015). The Notch-mediated hyperplasia circuitry in Drosophila reveals a Src-JNK signaling axis. Elife 4. PubMed ID: 26222204

    Guruharsha, K. G., Hori, K., Obar, R. A. and Artavanis-Tsakonas, S. (2014). Proteomic analysis of the Notch interactome. Methods Mol Biol 1187: 181-192. PubMed ID: 25053490

    Rhee, D. Y., Cho, D. Y., Zhai, B., Slattery, M., Ma, L., Mintseris, J., Wong, C. Y., White, K. P., Celniker, S. E., Przytycka, T. M., Gygi, S. P., Obar, R. A. and Artavanis-Tsakonas, S. (2014). Transcription factor networks in Drosophila melanogaster. Cell Rep 8: 2031-2043. PubMed ID: 25242320

    Hori, K., Sen, A. and Artavanis-Tsakonas, S. (2014). Genetic circuitry modulating notch signals through endosomal trafficking. Methods Enzymol 534: 283-299. PubMed ID: 24359960

    Sen, A., Dimlich, D. N., Guruharsha, K. G., Kankel, M. W., Hori, K., Yokokura, T., Brachat, S., Richardson, D., Loureiro, J., Sivasankaran, R., Curtis, D., Davidow, L. S., Rubin, L. L., Hart, A. C., Van Vactor, D. and Artavanis-Tsakonas, S. (2013). Genetic circuitry of Survival motor neuron, the gene underlying spinal muscular atrophy. Proc Natl Acad Sci U S A. PubMed ID: 23757500

    Fleming, R. J., Hori, K., Sen, A., Filloramo, G. V., Langer, J. M., Obar, R. A., Artavanis-Tsakonas, S., Maharaj-Best, A. C. (2013) An extracellular region of Serrate is essential for ligand-induced cis-inhibition of Notch signaling. Development 140: 2039-2049. PubMed ID: 23571220

    Pallavi, S. K., Ho, D. M., Hicks, C., Miele, L. and Artavanis-Tsakonas, S. (2012). Notch and Mef2 synergize to promote proliferation and metastasis through JNK signal activation in Drosophila. EMBO J. 31(13): 2895-907. PubMed ID: 22580825

  • Ruben Artero Department of Genetics, University of Valencia
    Blazquez-Bernal, Á., Fernandez-Costa, J. M., Bargiela, A. and Artero, R. (2021). Inhibition of autophagy rescues muscle atrophy in a LGMDD2 Drosophila model. Faseb j 35(10): e21914. PubMed ID: 34547132

    Selma-Soriano, E., Llamusi, B., Fernandez-Costa, J. M., Ozimski, L. L., Artero, R. and Redon, J. (2020). Rabphilin involvement in filtration and molecular uptake in Drosophila nephrocytes suggests a similar role in human podocytes. Dis Model Mech. PubMed ID: 32680845

    Bargiela, A., Sabater-Arcis, M., Espinosa-Espinosa, J., Zulaica, M., Lopez de Munain, A. and Artero, R. (2019). Increased Muscleblind levels by chloroquine treatment improve myotonic dystrophy type 1 phenotypes in in vitro and in vivo models. Proc Natl Acad Sci U S A 116(50): 25203-25213. PubMed ID: 31754023

    Sabater-Arcis, M., Bargiela, A., Furling, D. and Artero, R. (2019). miR-7 Restores Phenotypes in Myotonic Dystrophy Muscle Cells by Repressing Hyperactivated Autophagy. Mol Ther Nucleic Acids 19: 278-292. PubMed ID: 31855836

    Bargiela, A., Sabater-Arcis, M., Espinosa-Espinosa, J., Zulaica, M., Lopez de Munain, A. and Artero, R. (2019). Increased Muscleblind levels by chloroquine treatment improve myotonic dystrophy type 1 phenotypes in in vitro and in vivo models. Proc Natl Acad Sci U S A. PubMed ID: 31754023

    Chakraborty, M., Llamusi, B. and Artero, R. (2018). Modeling of Myotonic Dystrophy cardiac phenotypes in Drosophila. Front Neurol 9: 473. PubMed ID: 30061855

    Chakraborty, M., Sellier, C., Ney, M., Pascal, V., Charlet-Berguerand, N., Artero, R. and Llamusi, B. (2018). Daunorubicin reduces MBNL1 sequestration caused by CUG-repeat expansion and rescues cardiac dysfunctions in a Drosophila model of myotonic dystrophy. Dis Model Mech 11(4). PubMed ID: 29592894

    Cerro-Herreros, E., Chakraborty, M., Perez-Alonso, M., Artero, R. and Llamusi, B. (2017). Expanded CCUG repeat RNA expression in Drosophila heart and muscle trigger Myotonic Dystrophy type 1-like phenotypes and activate autophagocytosis genes. Sci Rep 7(1): 2843. PubMed ID: 28588248

    Cerro-Herreros, E., Fernandez-Costa, J. M., Sabater-Arcis, M., Llamusi, B. and Artero, R. (2016). Derepressing muscleblind expression by miRNA sponges ameliorates myotonic dystrophy-like phenotypes in Drosophila. Sci Rep 6: 36230. PubMed ID: 27805016

    Bargiela, A., Cerro-Herreros, E., Fernandez-Costa, J. M., Vilchez, J. J., Llamusi, B. and Artero, R. (2015). Increased autophagy and apoptosis contribute to muscle atrophy in a myotonic dystrophy type 1 Drosophila model. Dis Model Mech 8: 679-690. PubMed ID: 26092529

    Llamusi, B., Munoz-Soriano, V., Paricio, N. and Artero, R. (2014). The use of whole-mount in situ hybridization to illustrate gene expression regulation. Biochem Mol Biol Educ. PubMed ID: 24979316

    Bargiela, A., Llamusi, B., Cerro-Herreros, E. and Artero, R. (2014). Two Enhancers Control Transcription of Drosophila muscleblind in the Embryonic Somatic Musculature and in the Central Nervous System. PLoS One 9: e93125. PubMed ID: 24667536

  • Kenta Asahina Molecular Neurobiology Laboratory, Salk Institute, La Jolla, CA
    Wohl, M. P., Liu, J. and Asahina, K. (2023). Drosophila tachykininergic neurons modulate the activity of two groups of receptor-expressing neurons to regulate aggressive tone. J Neurosci. PubMed ID: 36977580

    Ishii, K., Cortese, M., Leng, X., Shokhirev, M. N. and Asahina, K. (2022). A neurogenetic mechanism of experience-dependent suppression of aggression. Sci Adv 8(36): eabg3203. PubMed ID: 36070378

    Wohl, M., Ishii, K. and Asahina, K. (2020). Layered roles of fruitless isoforms in specification and function of male aggression-promoting neurons in Drosophila. Elife 9. PubMed ID: 32314957

    Ishii, K., Wohl, M., DeSouza, A. and Asahina, K. (2020). Sex-determining genes distinctly regulate courtship capability and target preference via sexually dimorphic neurons. Elife 9. PubMed ID: 32314964

    Wohl, M., Ishii, K. and Asahina, K. (2020). Layered roles of fruitless isoforms in specification and function of male aggression-promoting neurons in Drosophila. Elife 9. PubMed ID: 32314957

    Asahina, K., Watanabe, K., Duistermars, B. J., Hoopfer, E., Gonzalez, C. R., Eyjolfsdottir, E. A., Perona, P. and Anderson, D. J. (2014). Tachykinin-expressing neurons control male-specific aggressive arousal in Drosophila. Cell 156(1-2): 221-235. PubMed ID: 24439378

  • Michael Ashburner Department of Cambridge, University of Cambridge
    von Grotthuss, M., Ashburner, M. and Ranz, J. M. (2010). Fragile regions and not functional constraints predominate in shaping gene organization in the genus Drosophila. Genome Res 20: 1084-1096. PubMed ID: 20601587

    Teixeira, L., Ferreira, A. and Ashburner, M. (2008). The bacterial symbiont Wolbachia induces resistance to RNA viral infections in Drosophila melanogaster. PLoS Biol 6(12): e2. PubMed ID: 19222304

  • Hillary Ashe University of Manchester
    Forbes Beadle, L., Zhou, H., Rattray, M. and Ashe, H. L. (2023). Modulation of transcription burst amplitude underpins dosage compensation in the Drosophila embryo. Cell Rep 42(4): 112382. PubMed ID: 37060568'

    Forbes Beadle, L., Love, J. C., Shapovalova, Y., Artemev, A., Rattray, M. and Ashe, H. L. (2023). Combined modelling of mRNA decay dynamics and single-molecule imaging in the Drosophila embryo uncovers a role for P-bodies in 5' to 3' degradation. PLoS Biol 21(1): e3001956. PubMed ID: 36649329

    Frampton, S. L., Sutcliffe, C., Baldock, C. and Ashe, H. L. (2022). Modelling the structure of Short Gastrulation and generation of a toolkit for studying its function in Drosophila. Biol Open 11(6). PubMed ID: 35603711

    Vinter, D. J., Hoppe, C., Minchington, T. G., Sutcliffe, C. and Ashe, H. L. (2021). Dynamics of hunchback translation in real time and at single mRNA resolution in the Drosophila embryo. Development 148(18). PubMed ID: 33722899

    Hoppe, C. and Ashe, H. L. (2021). CRISPR-Cas9 strategies to insert MS2 stem-loops into endogenous loci in Drosophila embryos. STAR Protoc 2(1): 100380. PubMed ID: 33786461

    Hoppe, C., Bowles, J. R., Minchington, T. G., Sutcliffe, C., Upadhyai, P., Rattray, M. and Ashe, H. L. (2020). Modulation of the Promoter Activation Rate Dictates the Transcriptional Response to Graded BMP Signaling Levels in the Drosophila Embryo. Dev Cell 54(6): 727-741. PubMed ID: 32758422

    Wilcockson, S. G. and Ashe, H. L. (2019). Drosophila Ovarian Germline Stem Cell Cytocensor Projections Dynamically Receive and Attenuate BMP Signaling. Dev Cell. PubMed ID: 31178401

    Deignan, L., Pinheiro, M.T., Sutcliffe, C., Saunders, A., Wilcockson, S.G., Zeef, L.A., Donaldson, I.J. and Ashe, H.L. (2016). Regulation of the BMP signaling-responsive transcriptional network in the Drosophila embryo. PLoS Genet 12: e1006164. PubMed ID: 27379389

    Sawala, A., Scarcia, M., Sutcliffe, C., Wilcockson, S. G. and Ashe, H. L. (2015). Peak BMP responses in the Drosophila embryo are dependent on the activation of integrin signaling. Cell Rep 12: 1584-1593. PubMed ID: 26321638

    Newton, F. G., Harris, R. E., Sutcliffe, C. and Ashe, H. L. (2015). Coordinate post-transcriptional repression of Dpp-dependent transcription factors attenuates signal range during development. Development. PubMed ID: 26293305

  • Yoshi Aso Janelia Research Campus, Ashburn, Virginia
    Yamada, D., Bushey, D., Li, F., Hibbard, K. L., Sammons, M., Funke, J., Litwin-Kumar, A., Hige, T. and Aso, Y. (2023). Hierarchical architecture of dopaminergic circuits enables second-order conditioning in Drosophila. Elife 12. PubMed ID: 36692262

    Scaplen, K. M., Talay, M., Fisher, J. D., Cohn, R., Sorkac, A., Aso, Y., Barnea, G. and Kaun, K. R. (2021). Transsynaptic mapping of Drosophila mushroom body output neurons. Elife 10. PubMed ID: 33570489

    Li, F., Lindsey, J. W., Marin, E. C., Otto, N., Dreher, M., Dempsey, G., Stark, I., Bates, A. S., Pleijzier, M. W., Schlegel, P., Nern, A., Takemura, S. Y., Eckstein, N., Yang, T., Francis, A., Braun, A., Parekh, R., Costa, M., Scheffer, L. K., Aso, Y., Jefferis, G. S., Abbott, L. F., Litwin-Kumar, A., Waddell, S. and Rubin, G. M. (2020). The connectome of the adult Drosophila mushroom body provides insights into function. Elife 9. PubMed ID: 33315010

  • Nigel Atkinson University of Texas, Austin
    Wijesekera, T. P., Wu, Z., Stephens, N. P., Godula, R., Lew, L. K. and Atkinson, N. S. (2022). A Non-Nuclear NF-κB Modulates Alcohol Sensitivity But Not Immunity. J Neurosci 42(16): 3329-3343. PubMed ID: 35273084

    Park, A., Tran, T., Scheuermann, E. A., Smith, D. P. and Atkinson, N. S. (2020). Alcohol potentiates a pheromone signal in flies. Elife 9. PubMed ID: 33141025

    Troutwine, B., Park, A., Velez-Hernandez, M. E., Lew, L., Mihic, S. J. and Atkinson, N. S. (2019). F654A and K558Q Mutations in NMDA Receptor 1 affect ethanol-induced behaviors in Drosophila. Alcohol Clin Exp Res. PubMed ID: 31593608

    Park, A., Tran, T. and Atkinson, N. S. (2018). Monitoring food preference in Drosophila by oligonucleotide tagging. Proc Natl Acad Sci U S A 115(36): 9020-9025. PubMed ID: 30127010

    Ghezzi, A., Li, X., Lew, L. K., Wijesekera, T. P. and Atkinson, N. S. (2017). Alcohol-induced neuroadaptation is orchestrated by the histone acetyltransferase CBP. Front Mol Neurosci 10: 103. PubMed ID: 28442993

    Ghezzi, A., Zomeno, M., Pietrzykowski, A. Z. and Atkinson, N. S. (2016). Immediate-early alcohol-responsive miRNA expression in Drosophila. J Neurogenet: 1-10. PubMed ID: 27845601

    Krishnan, H. R., Li, X., Ghezzi, A. and Atkinson, N. S. (2016). A DNA element in the slo gene modulates ethanol tolerance. Alcohol 51: 37-42. PubMed ID: 26992698

    Troutwine, B.R., Ghezzi, A., Pietrzykowski, A.Z. and Atkinson, N.S. (2016). Alcohol resistance in Drosophila is modulated by the Toll innate immune pathway. Genes Brain Behav [Epub ahead of print]. PubMed ID: 26916032

    Li, X., Ghezzi, A., Krishnan, H. R., Pohl, J. B., Bohm, A. Y. and Atkinson, N. S. (2015). A histone modification identifies a DNA element controlling slo BK channel gene expression in muscle. J Neurogenet: 1-32. PubMed ID: 25967280

    Ghezzi, A., Liebeskind, B. J., Thompson, A., Atkinson, N. S. and Zakon, H. H. (2014). Ancient association between cation leak channels and Mid1 proteins is conserved in fungi and animals. Front Mol Neurosci 7: 15. PubMed ID: 24639627

  • Agnes Audibert Sorbonne University, Developmental Biology Laboratory
    Darnat, P., Burg, A., Salle, J., Lacoste, J., Louvet-Vallee, S., Gho, M. and Audibert, A. (2022). Cortical Cyclin A controls spindle orientation during asymmetric cell divisions in Drosophila. Nat Commun 13(1): 2723. PubMed ID: 35581185

    Simon, F., Ramat, A., Louvet-Vallee, S., Lacoste, J., Burg, A., Audibert, A. and Gho, M. (2019). Shaping of Drosophila Neural Cell Lineages Through Coordination of Cell Proliferation and Cell Fate by the BTB-ZF Transcription Factor Tramtrack-69. Genetics 212(3): 773-788. PubMed ID: 31073020

    Ramat, A., Audibert, A., Louvet-Vallee, S., Simon, F., Fichelson, P. and Gho, M. (2016). Escargot and Scratch regulate neural commitment by antagonizing Notch activity in Drosophila sensory organs. Development 143(16): 3024-3034. PubMed ID: 27471258

    Fernando, C., Audibert, A., Simon, F., Tazi, J. and Juge, F. (2015). A role for the serine/arginine-rich (SR) protein B52/SRSF6 in cell growth and myc expression in Drosophila. Genetics 199(4): 1201-1211. PubMed ID: 25680814

  • Vanessa Auld Department of Biology, University of British Columbia
    Das, M., Cheng, D., Matzat, T. and Auld, V. J. (2023). Innexin-Mediated Adhesion between Glia Is Required for Axon Ensheathment in the Peripheral Nervous System. J Neurosci 43(13): 2260-2276. PubMed ID: 36801823

    Sharifkhodaei, Z. and Auld, V. J. (2020). Overexpressed Gliotactin activates BMP signaling through interfering with the Tkv-Dad association. Genome: 1-12. PubMed ID: 33064024

    Hunter, A. C., Petley-Ragan, L. M., Das, M. and Auld, V. J. (2020). Basigin Associates with Integrin in Order to Regulate Perineurial Glia and Drosophila Nervous System Morphology. J Neurosci 40(17): 3360-3373. PubMed ID: 32265259

    Sharifkhodaei, Z., Gilbert, M. M. and Auld, V. J. (2019). Scribble and Discs-large mediate tricellular junction formation. Development. PubMed ID: 31444218

    Daniel, E., Daude, M., Kolotuev, I., Charish, K., Auld, V. and Le Borgne, R. (2018). Coordination of septate junctions assembly and completion of cytokinesis in proliferative epithelial tissues. Curr Biol [Epub ahead of print]. PubMed ID: 29706514

    Samarasekera, G. and Auld, V. J. (2017). C-terminal Src kinase (Csk) regulates the tricellular junction protein Gliotactin independent of Src. Mol Biol Cell. PubMed ID: 29167383

    Padash Barmchi, M., Gilbert, M., Thomas, M., Banks, L., Zhang, B. and Auld, V. J. (2016). A Drosophila model of HPV E6-induced malignancy reveals essential roles for Magi and the Insulin Receptor. PLoS Pathog 12: e1005789. PubMed ID: 27537218

    Padash Barmchi, M., Samarasekera, G., Gilbert, M., Auld, V.J. and Zhang, B. (2016). Magi is associated with the Par complex and functions antagonistically with Bazooka to regulate the apical polarity complex. PLoS One 11: e0153259. PubMed ID: 27074039

    Khodaei, Z. S., Barmchi, M. P., Gilbert, M. M., Samarasekera, G., Fulga, T. A., Van Vactor, D. and Auld, V. J. (2016). The tricellular junction protein Gliotactin auto-regulates mRNA levels via BMP signaling induction of miR-184. J Cell Sci [Epub ahead of print]. PubMed ID: 26906422

    Petley-Ragan, L. M., Ardiel, E. L., Rankin, C. H. and Auld, V. J. (2016). Accumulation of Laminin Monomers in Drosophila Glia Leads to Glial Endoplasmic Reticulum Stress and Disrupted Larval Locomotion. J Neurosci 36: 1151-1164. PubMed ID: 26818504

    Xie, X., Gilbert, M., Petley-Ragan, L., Auld, V. J. (2014) Loss of focal adhesions in glia disrupts both glial and photoreceptor axon migration in the Drosophila visual system. Development 141: 3072-3083. PubMed ID: 25053436

  • Michalis Averof Institut de Génomique Fonctionnelle de Lyon
    Grillo, M., Casanova, J. and Averof, M. (2014). Development: a deep breath for endocrine organ evolution. Curr Biol 24: R38-40. PubMed ID: 24405679

    Rodel, C. J., Gilles, A. F. and Averof, M. (2013). MicroRNAs Act as Cofactors in Bicoid-Mediated Translational Repression. Curr Biol. PubMed ID: 23910663

    El-Sherif, E., Averof, M. and Brown, S. J. (2012). A segmentation clock operating in blastoderm and germband stages of Tribolium development. Development 139: 4341-4346. PubMed ID: 23095886

    Kontarakis, Z., Konstantinides, N., Pavlopoulos, A. and Averof, M. (2011). Reconfiguring gene traps for new tasks using iTRAC. Fly (Austin) 5: 352-355. PubMed ID: 22004889

  • Tomer Avidor-Reiss Department of Biological Sciences, University of Toledo
    Jo, K. H., Jaiswal, A., Khanal, S., Fishman, E. L., Curry, A. N. and Avidor-Reiss, T. (2019). Poc1B and Sas-6 function together during the atypical centriole formation in Drosophila melanogaster. Cells 8(8). PubMed ID: 31387336

    Khire, A., Jo, K.H., Kong, D., Akhshi, T., Blachon, S., Cekic, A.R., Hynek, S., Ha, A., Loncarek, J., Mennella, V. and Avidor-Reiss, T. (2017). Centriole remodeling during spermiogenesis in Drosophila. Curr Biol 26: 3183-3189. PubMed ID: 28094036

    Khire, A., Vizuet, A. A., Davila, E. and Avidor-Reiss, T. (2015). Asterless reduction during spermiogenesis is regulated by Plk4 and is essential for zygote development in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26480844

    Ha, A., Polyanovsky, A. and Avidor-Reiss, T. (2015). Drosophila Hook-Related Protein (Girdin) s Essential for Sensory Dendrite Formation. Genetics [Epub ahead of print]. PubMed ID: 26058848

    Basiri, M. L., Ha, A., Chadha, A., Clark, N. M., Polyanovsky, A., Cook, B. and Avidor-Reiss, T. (2014). A migrating ciliary gate compartmentalizes the site of axoneme assembly in Drosophila spermatids. Curr Biol 24: 2622-2631. PubMed ID: 25447994

    Blachon, S., Khire, A. and Avidor-Reiss, T. (2014). The origin of the second centriole in the zygote of Drosophila melanogaster. Genetics 197: 199-205. PubMed ID: 24532732

  • Richard Axel College of Physicians and Surgeons, Columbia University
    Cury, K. M. and Axel, R. (2023). Flexible neural control of transition points within the egg-laying behavioral sequence in Drosophila. Nat Neurosci 26(6): 1054-1067. PubMed ID: 37217726

    Bonheur, M., Swartz, K. J., Metcalf, M. G., Wen, X., Zhukovskaya, A., Mehta, A., Connors, K. E., Barasch, J. G., Jamieson, A. R., Martin, K. C., Axel, R. and Hattori, D. (2023). A rapid and bidirectional reporter of neural activity reveals neural correlates of social behaviors in Drosophila. Nat Neurosci 26(7): 1295-1307. PubMed ID: 37308660

    Devineni, A. V., Deere, J. U., Sun, B. and Axel, R. (2021). Individual bitter-sensing neurons in Drosophila exhibit both ON and OFF responses that influence synaptic plasticity. Curr Biol. PubMed ID: 34731675

    Devineni, A. V., Sun, B., Zhukovskaya, A. and Axel, R. (2019). Acetic acid activates distinct taste pathways in Drosophila to elicit opposing, state-dependent feeding responses. Elife 8. PubMed ID: 31205005

    Hattori, D., Aso, Y., Swartz, K. J., Rubin, G. M., Abbott, L. F. and Axel, R. (2017). Representations of novelty and familiarity in a mushroom body compartment. Cell 169(5): 956-969. PubMed ID: 28502772

    Litwin-Kumar, A., Harris, K. D., Axel, R., Sompolinsky, H. and Abbott, L. F. (2017). Optimal Degrees of Synaptic Connectivity. Neuron 93(5): 1153-1164.e1157. PubMed ID: 28215558

    Jagadish, S., Barnea, G., Clandinin, T. R., Axel, R. (2014) Identifying Functional Connections of the Inner Photoreceptors in Drosophila using Tango-Trace. Neuron. PubMed ID: 25043419

    Caron, S. J., Ruta, V., Abbott, L. F. and Axel, R. (2013). Random convergence of olfactory inputs in the Drosophila mushroom body. Nature. PubMed ID: 23615618

    Ruta, V., Datta, S. R., Vasconcelos, M. L., Freeland, J., Looger, L. L. and Axel, R. (2010). A dimorphic pheromone circuit in Drosophila from sensory input to descending output. Nature 468: 686-690. PubMed ID: 21124455

  • Jeff Axelrod Department of Pathology, Stanford University School of Medicine
    Cho, B., Song, S., Wan, J. Y. and Axelrod, J. D. (2022). Prickle isoform participation in distinct polarization events in the Drosophila eye. PLoS One 17(2): e0262328. PubMed ID: 35148314

    Cho, B., Song, S. and Axelrod, J. D. (2020). Prickle isoforms determine handedness of helical morphogenesis. Elife 9. PubMed ID: 31934858

    Cho, B., Pierre-Louis, G., Sagner, A., Eaton, S. and Axelrod, J. D. (2015). Clustering and negative feedback by endocytosis in planar cell polarity signaling is modulated by ubiquitinylation of prickle. PLoS Genet 11: e1005259. PubMed ID: 25996914

    Sharp, K.A. and Axelrod, J.D. (2016). Prickle isoforms control the direction of tissue polarity by microtubule independent and dependent mechanisms. Biol Open [Epub ahead of print]. PubMed ID: 26863941

    Matis, M., Russler-Germain, D. A., Hu, Q., Tomlin, C. J. and Axelrod, J. D. (2014). Microtubules provide directional information for core PCP function. Elife 3: e02893. PubMed ID: 25124458

    Olofsson, J., Sharp, K. A., Matis, M., Cho, B. and Axelrod, J. D. (2014). Prickle/spiny-legs isoforms control the polarity of the apical microtubule network in planar cell polarity. Development 141: 2866-2874. PubMed ID: 25005476

    Olofsson, J. and Axelrod, J. D. (2014). Methods for studying planar cell polarity. Methods. PubMed ID: 24680701

    Matis, M. and Axelrod, J. D. (2013). Regulation of PCP by the Fat signaling pathway. Genes Dev 27: 2207-2220. PubMed ID: 24142873

    Ducuing, A., Mollereau, B., Axelrod, J. D. and Vincent, S. (2013). Absolute requirement of cholesterol binding for Hedgehog gradient formation in Drosophila. Biol Open 2: 596-604. PubMed ID: 23789110

    Vladar, E. K., Bayly, R. D., Sangoram, A. M., Scott, M. P. and Axelrod, J. D. (2012). Microtubules enable the planar cell polarity of airway cilia. Curr Biol 22: 2203-2212. PubMed ID: 23122850

  • Julien Ayroles Department of Ecology and Evolutionary Biology, Princeton University, Princeton, N.J.
    Pallares, L. F., Lea, A. J., Han, C., Filippova, E. V., Andolfatto, P. and Ayroles, J. F. (2023). Dietary stress remodels the genetic architecture of lifespan variation in outbred Drosophila. Nat Genet 55(1): 123-129. PubMed ID: 36550361

    Henry, L. P. and Ayroles, J. F. (2022). Drosophila melanogaster microbiome is shaped by strict filtering and neutrality along a latitudinal cline. Mol Ecol. PubMed ID: 36094780

    Akhund-Zade, J., Lall, S., Gajda, E., Yoon, D., Ayroles, J. F. and de Bivort, B. L. (2021). Genetic basis of offspring number-body weight tradeoff in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 33871609

    Henry, L. P. and Ayroles, J. F. (2021). Meta-analysis suggests the microbiome responds to Evolve and Resequence experiments in Drosophila melanogaster. BMC Microbiol 21(1): 108. PubMed ID: 33836662

    Pallares, L. F., Picard, S. and Ayroles, J. F. (2020). TM3'seq: A Tagmentation-Mediated 3' Sequencing Approach for Improving Scalability of RNAseq Experiments. G3 (Bethesda) 10(1): 143-150. PubMed ID: 31676507

  • Ferran Azorín Institute for Research in Biomedicine (IRB), Barcelona
    Bernues, J., Izquierdo-Boulstridge, A., Reina, O., Castejon, L., Fernandez-Castanner, E., Leal, N., Guerrero-Pepinosa, N., Bonet-Costa, C., Vujatovic, O., Climent-Canto, P. and Azorin, F. (2022). Lysine 27 dimethylation of Drosophila linker histone dH1 contributes to heterochromatin organization independently of H3K9 methylation. Nucleic Acids Res 50(16): 9212-9225. PubMed ID: 36039761

    Climent-Canto, P., Carbonell, A., Tamirisa, S., Henn, L., Perez-Montero, S., Boros, I. M. and Azorin, F. (2021). The tumour suppressor brain tumour (Brat) regulates linker histone dBigH1 expression in the Drosophila female germline and the early embryo. Open Biol 11(5): 200408. PubMed ID: 33947246

    Torras-Llort, M., Medina-Giro, S., Escudero-Ferruz, P., Lipinszki, Z., Moreno-Moreno, O., Karman, Z., Przewloka, M. R. and Azorin, F. (2020). A fraction of barrier-to-autointegration factor (BAF) associates with centromeres and controls mitosis progression. Commun Biol 3(1): 454. PubMed ID: 32814801

    Climent-Canto, P., Carbonell, A., Tatarski, M., Reina, O., Bujosa, P., Font-Mateu, J., Bernues, J., Beato, M. and Azorin, F. (2020). The embryonic linker histone dBigH1 alters the functional state of active chromatin. Nucleic Acids Res. PubMed ID: 32103264

    Di Mauro, G., Carbonell, A., Escudero-Ferruz, P. and Azorin, F. (2020). The zinc-finger proteins WOC and ROW play distinct functions within the HP1c transcription complex. Biochim Biophys Acta Gene Regul Mech: 194492. PubMed ID: 32006714

    Moreno-Moreno, O., Torras-Llort, M. and Azorin, F. (2019). The E3-ligases SCFPpa and APC/CCdh1 co-operate to regulate CENP-ACID expression across the cell cycle. Nucleic Acids Res. PubMed ID: 30753559

    Satovic, E., Font-Mateu, J., Carbonell, A., Beato, M. and Azorin, F. (2018). Chromatin remodeling in Drosophila preblastodermic embryo extract. Sci Rep 8(1): 10927. PubMed ID: 30026552

    Carbonell, A., Perez-Montero, S., Climent-Canto, P., Reina, O. and Azorin, F. (2017). The germline linker histone dBigH1 and the translational regulator Bam form a repressor loop essential for male germ stem cell differentiation. Cell Rep 21(11): 3178-3189. PubMed ID: 29241545

    Kessler, R., Tisserand, J., Font-Burgada, J., Reina, O., Coch, L., Attolini, C. S., Garcia-Bassets, I. and Azorin, F. (2015). dDsk2 regulates H2Bub1 and RNA polymerase II pausing at dHP1c complex target genes. Nat Commun 6: 7049. PubMed ID: 25916810

    Morán, T., Bernués, J. and Azorín, F. (2015) The Drosophila histone demethylase dKDM5/LID regulates hematopoietic development. Dev Biol [Epub ahead of print]. PubMed ID: 26183107

    Perez-Montero, S., Carbonell, A., Moran, T., Vaquero, A. and Azorin, F. (2013). The Embryonic Linker Histone H1 Variant of Drosophila, dBigH1, Regulates Zygotic Genome Activation. Dev Cell. PubMed ID: 24055651

    Lloret-Llinares, M., Perez-Lluch, S., Rossell, D., Moran, T., Ponsa-Cobas, J., Auer, H., Corominas, M. and Azorin, F. (2012). dKDM5/LID regulates H3K4me3 dynamics at the transcription-start site (TSS) of actively transcribed developmental genes. Nucleic Acids Res 40: 9493-9505. PubMed ID: 22904080

  • Natalia Azpiazu Centro de Biología Molecular Severo Ochoa, Madrid
    Blom-Dahl, D. and Azpiazu, N. (2017). The Pax protein Eyegone (Eyg) interacts with the pi-RNA component Aubergine (Aub) and controls egg chamber development in Drosophila. Dev Biol [Epub ahead of print]. PubMed ID: 29278721

    Zaballos, M. A., Cantero, W. and Azpiazu, N. (2015). The TALE Transcription Factor Homothorax Functions to Assemble Heterochromatin during Drosophila Embryogenesis. PLoS One 10: e0120662. PubMed ID: 25794008

    Corsetti, E. and Azpiazu, N. (2013). Functional dissection of the splice variants of the Drosophila gene homothorax (hth). Dev Biol 384: 72-82. PubMed ID: 24075905

    Salvany, L., Aldaz, S., Corsetti, E. and Azpiazu, N. (2009). A new role for hth in the early pre-blastodermic divisions in drosophila. Cell Cycle 8: 2748-2755. PubMed ID: 19652544


  • Return to The Interactive Fly