Fly Labs and References


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


  • Gabriel Haddad Biomedical Sciences Graduate Program, University of California, San Diego
    Iranmehr, A., Stobdan, T., Zhou, D., Zhao, H., Kryazhimskiy, S., Bafna, V. and Haddad, G. G. (2021). Multiple mechanisms drive genomic adaptation to extreme O(2) levels in Drosophila melanogaster. Nat Commun 12(1): 997. PubMed ID: 33579965

    Stobdan, T., Sahoo, D., Azad, P., Hartley, I., Heinrichsen, E., Zhou, D. and Haddad, G. G. (2019). High fat diet induces sex-specific differential gene expression in Drosophila melanogaster. PLoS One 14(3): e0213474. PubMed ID: 30861021

    Jha, A. R., Zhou, D., Brown, C. D., Kreitman, M., Haddad, G. G. and White, K. P. (2015). Shared Genetic Signals of Hypoxia Adaptation in Drosophila and in High-Altitude Human Populations. Mol Biol Evol [Epub ahead of print]. PubMed ID: 26576852

    Huang, H., Lu-Bo, Y. and Haddad, G. G. (2014). A Drosophila ABC Transporter Regulates Lifespan. PLoS Genet 10: e1004844. PubMed ID: 25474322

    Heinrichsen, E. T., Zhang, H., Robinson, J. E., Ngo, J., Diop, S., Bodmer, R., Joiner, W. J., Metallo, C. M. and Haddad, G. G. (2014). Metabolic and transcriptional response to a high-fat diet in Drosophila melanogaster. Mol Metab 3: 42-54. PubMed ID: 24567903

    Udpa, N., et al. (2014). Whole genome sequencing of Ethiopian highlanders reveals conserved hypoxia tolerance genes. Genome Biol 15: R36. PubMed ID: 24555826

    Yin, S., Xue, J., Sun, H., Wen, B., Wang, Q., Perkins, G., Zhao, H. W., Ellisman, M. H., Hsiao, Y. H., Yin, L., Xie, Y., Hou, G., Zi, J., Lin, L., Haddad, G. G., Zhou, D. and Liu, S. (2013). Quantitative evaluation of the mitochondrial proteomes of Drosophila melanogaster adapted to extreme oxygen conditions. PLoS One 8: e74011. PubMed ID: 24069262

    Xie, L., Ng, C., Ali, T., Valencia, R., Ferreira, B. L., Xue, V., Tanweer, M., Zhou, D., Haddad, G. G., Bourne, P. E. and Xie, L. (2013). Multiscale modeling of the causal functional roles of nsSNPs in a genome-wide association study: application to hypoxia. BMC Genomics 14 Suppl 3: S9. PubMed ID: 23819581

    Zhou, D. and Haddad, G. G. (2013). Genetic analysis of hypoxia tolerance and susceptibility in Drosophila and humans. Annu Rev Genomics Hum Genet 14: 25-43. PubMed ID: 23808366

  • Penelope Haddrill Institute of Evolutionary Biology, University of Edinburgh
    Keightley, P. D., Ness, R. W., Halligan, D. L. and Haddrill, P. R. (2013). Estimation of the Spontaneous Mutation Rate per Nucleotide Site in a Drosophila melanogaster Full-Sib Family. Genetics. PubMed ID: 24214343

    Campos, J. L., Zeng, K., Parker, D. J., Charlesworth, B. and Haddrill, P. R. (2013). Codon usage bias and effective population sizes on the X chromosome versus the autosomes in Drosophila melanogaster. Mol Biol Evol 30: 811-823. PubMed ID: 23204387

    Campos, J. L., Charlesworth, B. and Haddrill, P. R. (2012). Molecular evolution in nonrecombining regions of the Drosophila melanogaster genome. Genome Biol Evol 4: 278-288. PubMed ID: 22275518

  • Marc Haenlin Centre de Biologie du Développement, Université Toulouse
    Moussalem, D., Augé, B., Di Stefano, L., Osman, D., Gobert, V. and Haenlin, M. (2021). Two Isoforms of serpent Containing Either One or Two GATA Zinc Fingers Provide Functional Diversity During Drosophila Development. Front Cell Dev Biol 9: 795680. PubMed ID: 35178397

    Miller, M., Chen, A., Gobert, V., Auge, B., Beau, M., Burlet-Schiltz, O., Haenlin, M. and Waltzer, L. (2017). Control of RUNX-induced repression of Notch signaling by MLF and its partner DnaJ-1 during Drosophila hematopoiesis. PLoS Genet 13(7): e1006932. PubMed ID: 28742844

    Benmimoun, B., Polesello, C., Haenlin, M. and Waltzer L. (2015). The EBF transcription factor Collier directly promotes Drosophila blood cell progenitor maintenance independently of the niche. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26150488

    Breig, O., Bras, S., Soria, N. M., Osman, D., Heidenreich, O., Haenlin, M. and Waltzer, L. (2013). Pontin is a critical regulator for AML1-ETO-induced leukemia. Leukemia. PubMed ID: 24342949

    Benmimoun, B., Polesello, C., Waltzer, L. and Haenlin, M. (2012). Dual role for Insulin/TOR signaling in the control of hematopoietic progenitor maintenance in Drosophila. Development 139: 1713-1717. PubMed ID: 22510984

    Hijazi, A., Haenlin, M., Waltzer, L. and Roch, F. (2011). The Ly6 protein coiled is required for septate junction and blood brain barrier organisation in Drosophila. PLoS One 6: e17763. PubMed ID: 21423573

    Polesello, C., Roch, F., Gobert, V., Haenlin, M. and Waltzer, L. (2011). Modeling cancers in Drosophila. Prog Mol Biol Transl Sci 100: 51-82. PubMed ID: 21377624

  • Ernst Hafen Institute of Molecular Systems Biology, Zurich
    Hodge, S. H., Watts, A., Marley, R., Baines, R. A., Hafen, E. and MacDougall, L. K. (2021). Twitchy, the Drosophila orthologue of the ciliary gating protein FBF1/dyf-19, is required for coordinated locomotion and male fertility. Biol Open 10(8). PubMed ID: 34357392

    Hodge, S. H., Watts, A., Marley, R., Baines, R. A., Hafen, E. and MacDougall, L. K. (2021). Twitchy, the Drosophila orthologue of the ciliary gating protein FBF1/dyf-19, is required for coordinated locomotion and male fertility. Biol Open. PubMed ID: 34232985

    Okada, H., Yagi, R., Gardeux, V., Deplancke, B. and Hafen, E. (2019). Sex-dependent and sex-independent regulatory systems of size variation in natural populations. Mol Syst Biol 15(11): e9012. PubMed ID: 31777173

    Okada, H., Ebhardt, H. A., Vonesch, S. C., Aebersold, R. and Hafen, E. (2016). Proteome-wide association studies identify biochemical modules associated with a wing-size phenotype in Drosophila melanogaster. Nat Commun 7: 12649. PubMed ID: 27582081

    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

    Vonesch, S.C., Lamparter, D., Mackay, T.F., Bergmann, S. and Hafen, E. (2016). Genome-wide analysis reveals novel regulators of growth in Drosophila melanogaster. PLoS Genet 12: e1005616. PubMed ID: 26751788

    Jevtov, I., Zacharogianni, M., van Oorschot, M. M., van Zadelhoff, G., Aguilera-Gomez, A., Vuillez, I., Braakman, I., Hafen, E., Stocker, H. and Rabouille, C. (2015). TORC2 mediates the heat stress response in Drosophila by promoting the formation of stress granules. J Cell Sci. PubMed ID: 26054799

    Okada, H., Schittenhelm, R. B., Straessle, A. and Hafen, E. (2015). Multi-Functional Regulation of 4E-BP Gene Expression by the Ccr4-Not Complex. PLoS One 10: e0113902. PubMed ID: 25793896

    Medici, V., Vonesch, S. C., Fry, S. N. and Hafen, E. (2015). The FlyCatwalk: A high throughput feature-based sorting system for artificial selection in Drosophila. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 25556112

    Handke, B., Szabad, J., Lidsky, P. V., Hafen, E. and Lehner, C. F. (2014). Towards long term cultivation of Drosophila wing imaginal discs in vitro. PLoS One 9: e107333. PubMed ID: 25203426

  • Pejmun Haghighi Buck Institute for Research on Aging, Novato, CA
    Kauwe, G., Tsurudome, K., Penney, J., Mori, M., Gray, L., Calderon, M. R., Elazouzzi, F., Chicoine, N., Sonenberg, N. and Haghighi, A. P. (2016). Acute fasting regulates retrograde synaptic enhancement through a 4E-BP-dependent mechanism. Neuron [Epub ahead of print]. PubMed ID: 27916456

    Penney, J., Tsurudome, K., Liao, E. H., Kauwe, G., Gray, L., Yanagiya, A., M, R. C., Sonenberg, N. and Haghighi, A. P. (2016). LRRK2 regulates retrograde synaptic compensation at the Drosophila neuromuscular junction. Nat Commun 7: 12188. PubMed ID: 27432119

    Penney, J., Tsurudome, K., Liao, E. H., Elazzouzi, F., Livingstone, M., Gonzalez, M., Sonenberg, N. and Haghighi, A. P. (2012). TOR is required for the retrograde regulation of synaptic homeostasis at the Drosophila neuromuscular junction. Neuron 74(1): 166-178. PubMed ID: 22500638

    Sun, K., Westholm, J. O., Tsurudome, K., Hagen, J. W., Lu, Y., Kohwi, M., Betel, D., Gao, F. B., Haghighi, A. P., Doe, C. Q. and Lai, E. C. (2012). Neurophysiological defects and neuronal gene deregulation in Drosophila mir-124 mutants. PLoS Genet 8(2): e1002515. PubMed ID: 22347817

  • Kaisa Haglund Oslo University Hospital
    Lie-Jensen, A., Ivanauskiene, K., Malerod, L., Jain, A., Tan, K. W., Laerdahl, J. K., Liestol, K., Stenmark, H. and Haglund, K. (2019). Centralspindlin Recruits ALIX to the Midbody during Cytokinetic Abscission in Drosophila via a Mechanism Analogous to Virus Budding. Curr Biol. PubMed ID: 31607533

    Malerod, L., Le Borgne, R., Lie-Jensen, A., Eikenes, A. H., Brech, A., Liestol, K., Stenmark, H. and Haglund, K. (2018). Centrosomal ALIX regulates mitotic spindle orientation by modulating astral microtubule dynamics. Embo j. PubMed ID: 29858227

    Eikenes, A. H., Malerod, L., Christensen, A. L., Steen, C. B., Mathieu, J., Nezis, I. P., Liestol, K., Huynh, J. R., Stenmark, H. and Haglund, K. (2015). ALIX and ESCRT-III coordinately control cytokinetic abscission during germline stem cell division in vivo. PLoS Genet 11: e1004904. PubMed ID: 25635693

    Eikenes, A. H., Brech, A., Stenmark, H., Haglund, K. (2013) Spatiotemporal control of Cindr at ring canals during incomplete cytokinesis in the Drosophila male germline. Dev Biol. PubMed ID: 23499247

    Oppelt, A., Lobert, V. H., Haglund, K., Mackey, A. M., Rameh, L. E., Liestol, K., Schink, K. O., Pedersen, N. M., Wenzel, E. M., Haugsten, E. M., Brech, A., Rusten, T. E., Stenmark, H. and Wesche, J. (2013). Production of phosphatidylinositol 5-phosphate via PIKfyve and MTMR3 regulates cell migration. EMBO Rep 14: 57-64. PubMed ID: 23154468

    Haglund, K., Nezis, I. P., Lemus, D., Grabbe, C., Wesche, J., Liestol, K., Dikic, I., Palmer, R. and Stenmark, H. (2010). Cindr interacts with anillin to control cytokinesis in Drosophila melanogaster. Curr Biol 20: 944-950. PubMed ID: 20451383

  • Daniel Hahn Entomology and Nematology Department, University of Florida, Gainesville
    Williams, C. M., Rocca, J. R., Edison, A. S., Allison, D. B., Morgan, T. J. and Hahn, D. A. (2018). Cold adaptation does not alter ATP homeostasis during cold exposure in Drosophila melanogaster. Integr Zool. PubMed ID: 29722155

    Williams, C. M., McCue, M. D., Sunny, N. E., Szejner-Sigal, A., Morgan, T. J., Allison, D. B. and Hahn, D. A. (2016). Cold adaptation increases rates of nutrient flow and metabolic plasticity during cold exposure in Drosophila melanogaster. Proc Biol Sci 283. PubMed ID: 27605506

    Williams, C. M., Watanabe, M., Guarracino, M. R., Ferraro, M. B., Edison, A. S., Morgan, T. J., Boroujerdi, A. F. and Hahn, D. A. (2014). Cold adaptation shapes the robustness of metabolic networks in Drosophila melanogaster. Evolution. PubMed ID: 25308124

  • Matthew Hahn Department of Biology and School of Informatics and Computing, Indiana University, Bloomington
    Adrion, J. R., Begun, D. J. and Hahn, M. W. (2018). Patterns of transposable element variation and clinality in Drosophila. Mol Ecol. PubMed ID: 30484926

    Adrion, J. R., Song, M. J., Schrider, D. R., Hahn, M. W. and Schaack, S. (2017). Genome-Wide Estimates of Transposable Element Insertion and Deletion Rates in Drosophila Melanogaster. Genome Biol Evol 9(5): 1329-1340. PubMed ID: 28338986

    Schrider, D. R., Hahn, M. W. and Begun, D. J. (2016). Parallel Evolution of Copy-Number Variation across Continents in Drosophila melanogaster. Mol Biol Evol 33(5): 1308-1316. PubMed ID: 26809315

    Pease, J. B. and Hahn, M. W. (2013). More accurate phylogenies inferred from low-recombination regions in the presence of incomplete lineage sorting. Evolution 67: 2376-2384. PubMed ID: 23888858

    Schrider, D. R., Houle, D., Lynch, M. and Hahn, M. W. (2013). Rates and Genomic Consequences of Spontaneous Mutational Events in Drosophila melanogaster. Genetics. PubMed ID: 23733788

    Langley, C. H., Stevens, K., Cardeno, C., Lee, Y. C., Schrider, D. R., Pool, J. E., Langley, S. A., Suarez, C., Corbett-Detig, R. B., Kolaczkowski, B., Fang, S., Nista, P. M., Holloway, A. K., Kern, A. D., Dewey, C. N., Song, Y. S., Hahn, M. W. and Begun, D. J. (2012). Genomic variation in natural populations of Drosophila melanogaster. Genetics 192: 533-598. PubMed ID: 22673804

    Han, M. V. and Hahn, M. W. (2012). Inferring the history of interchromosomal gene transposition in Drosophila using n-dimensional parsimony. Genetics 190: 813-825. PubMed ID: 22095076

    Schrider, D. R., Stevens, K., Cardeno, C. M., Langley, C. H. and Hahn, M. W. (2011). Genome-wide analysis of retrogene polymorphisms in Drosophila melanogaster. Genome Res 21: 2087-2095. PubMed ID: 22135405

  • Georg Halder VIB, Flanders, Belgium
    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

    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

    Davie, K., Jacobs, J., Atkins, M., Potier, D., Christiaens, V., Halder, G. and Aerts, S. (2015). Discovery of transcription factors and regulatory regions driving in vivo tumor development by ATAC-seq and FAIRE-seq open chromatin profiling. PLoS Genet 11: e1004994. PubMed ID: 25679813

    Yang, C.C., Graves, H.K., Moya, I.M., Tao, C., Hamaratoglu, F., Gladden, A.B. and Halder, G. (2015). Differential regulation of the Hippo pathway by adherens junctions and apical-basal cell polarity modules. Proc Natl Acad Sci USA [Epub ahead of print]. PubMed ID: 25624491Atkins, M., Jiang, Y., Sansores-Garcia, L.,

    Jusiak, B., Halder, G. and Mardon, G. (2013). Dynamic rewiring of the Drosophila retinal determination network switches its function from selector to differentiation. PLoS Genet 9: e1003731. PubMed ID: 24009524

    Bossuyt, W., Chen, C. L., Chen, Q., Sudol, M., McNeill, H., Pan, D., Kopp, A., Halder, G. (2013) An evolutionary shift in the regulation of the Hippo pathway between mice and flies. Oncogene. PubMed ID: 23563179

    Sansores-Garcia, L., Atkins, M., Moya, I. M., Shahmoradgoli, M., Tao, C., Mills, G. B. and Halder, G. (2013). Mask Is Required for the Activity of the Hippo Pathway Effector Yki/YAP. Curr Biol. PubMed ID: 23333314

    Schroeder, M. C., Chen, C. L., Gajewski, K. and Halder, G. (2012). A non-cell-autonomous tumor suppressor role for Stat in eliminating oncogenic scribble cells. Oncogene. PubMed ID: 23108407

    Graves, H. K., Woodfield, S. E., Yang, C. C., Halder, G. and Bergmann, A. (2012). Notch signaling activates Yorkie non-cell autonomously in Drosophila. PLoS One 7: e37615. PubMed ID: 22679484

  • Mark Halfon Department of Biochemistry, State University of New York at Buffalo
    Schember, I. and Halfon, M. S. (2021). Identification of new Anopheles gambiae transcriptional enhancers using a cross-species prediction approach. Insect Mol Biol. PubMed ID: 33866636

    Zhou, Y., Popadowski, S. E., Deustchman, E. and Halfon, M. S. (2019). Distinct roles and requirements for Ras pathway signaling in visceral versus somatic muscle founder specification. Development 146(2). PubMed ID: 30630823

    Suryamohan, K., Hanson, C., Andrews, E., Sinha, S., Scheel, M. D. and Halfon, M. S. (2016). Redeployment of a conserved gene regulatory network during Aedes aegypti development. Dev Biol [Epub ahead of print]. PubMed ID: 27341759

    Halfon, M. S., Zhu, Q., Brennan, E. R. and Zhou, Y. (2011). Erroneous attribution of relevant transcription factor binding sites despite successful prediction of cis-regulatory modules. BMC Genomics 12: 578. PubMed ID: 22115527

    Kazemian, M., Zhu, Q., Halfon, M. S. and Sinha, S. (2011). Improved accuracy of supervised CRM discovery with interpolated Markov models and cross-species comparison. Nucleic Acids Res 39: 9463-9472. PubMed ID: 21821659

    Gallo, S. M., Gerrard, D. T., Miner, D., Simich, M., Des Soye, B., Bergman, C. M. and Halfon, M. S. (2011). REDfly v3.0: toward a comprehensive database of transcriptional regulatory elements in Drosophila. Nucleic Acids Res 39: D118-123. PubMed ID: 20965965

  • Fumika Hamada University of Cincinnati Department of Pediatrics
    Chen, S. C., Tang, X., Goda, T., Umezaki, Y., Riley, A. C., Sekiguchi, M., Yoshii, T. and Hamada, F. N. (2022). Dorsal clock networks drive temperature preference rhythms in Drosophila. Cell Rep 39(2): 110668. PubMed ID: 35417715

    Goda, T. and Hamada, F. N. (2019). Drosophila temperature preference rhythms: An innovative model to understand body temperature rhythms. Int J Mol Sci 20(8). PubMed ID: 31018551

    Goda, T., Umezaki, Y., Alwattari, F., Seo, H. W. and Hamada, F. N. (2019). Neuropeptides PDF and DH31 hierarchically regulate free-running rhythmicity in Drosophila circadian locomotor activity. Sci Rep 9(1): 838. PubMed ID: 30696873

    Goda, T., Doi, M., Umezaki, Y., Murai, I., Shimatani, H., Chu, M. L., Nguyen, V. H., Okamura, H. and Hamada, F. N. (2018). Calcitonin receptors are ancient modulators for rhythms of preferential temperature in insects and body temperature in mammals. Genes Dev 32(2): 140-155. PubMed ID: 29440246

    Tang, X., Roessingh, S., Hayley, S. E., Chu, M. L., Tanaka, N. K., Wolfgang, W., Song, S., Stanewsky, R. and Hamada, F. N. (2017). The role of PDF neurons in setting preferred temperature before dawn in Drosophila. Elife 6. PubMed ID: 28463109

    Goda, T., Tang, X., Umezaki, Y., Chu, M. L. and Hamada, F. N. (2016). Drosophila DH31 neuropeptide and PDF receptor regulate night-onset temperature preference. J Neurosci 36: 11739-11754. PubMed ID: 27852781

    Head, L.M., Tang, X., Hayley, S.E., Goda, T., Umezaki, Y., Chang, E.C., Leslie, J.R., Fujiwara, M., Garrity, P.A. and Hamada, F.N. (2015). The influence of light on temperature preference in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 25866391

    Goda, T., Leslie, J. R. and Hamada, F. N. (2014). Design and analysis of temperature preference behavior and its circadian rhythm in Drosophila. J Vis Exp. PubMed ID: 24457268

    Tang, X., Platt, M. D., Lagnese, C. M., Leslie, J. R., Hamada, F. N. (2013) Temperature Integration at the AC Thermosensory Neurons in Drosophila. J Neurosci 33: 894-901. PubMed ID: 23325228

    Kaneko, H., Head, L. M., Ling, J., Tang, X., Liu, Y., Hardin, P. E., Emery, P. and Hamada, F. N. (2012). Circadian rhythm of temperature preference and its neural control in Drosophila. Curr Biol 22: 1851-1857. PubMed ID: 22981774

  • Chun Han Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY
    Ji, H., Wang, B., Shen, Y., Labib, D., Lei, J., Chen, X., Sapar, M., Boulanger, A., Dura, J. M. and Han, C. (2023). The Drosophila chemokine-like Orion bridges phosphatidylserine and Draper in phagocytosis of neurons. Proc Natl Acad Sci U S A 120(24): e2303392120. PubMed ID: 37276397

    Ji, H., Sapar, M. L., Sarkar, A., Wang, B. and Han, C. (2022). Phagocytosis and self-destruction break down dendrites of Drosophila sensory neurons at distinct steps of Wallerian degeneration. Proc Natl Acad Sci U S A 119(4). PubMed ID: 35058357

    Allen, S. E., Koreman, G. T., Sarkar, A., Wang, B., Wolfner, M. F. and Han, C. (2021). Versatile CRISPR/Cas9-mediated mosaic analysis by gRNA-induced crossing-over for unmodified genomes. PLoS Biol 19(1): e3001061. PubMed ID: 33444322

    Poe, A. R., Xu, Y., Zhang, C., Lei, J., Li, K., Labib, D. and Han, C. (2020). Low FoxO expression in Drosophila somatosensory neurons protects dendrite growth under nutrient restriction. Elife 9. PubMed ID: 32427101

    Sapar, M. L. and Han, C. (2019). Die in pieces: How Drosophila sheds light on neurite degeneration and clearance. J Genet Genomics 46(4): 187-199. PubMed ID: 31080046

    Sapar, M. L., Ji, H., Wang, B., Poe, A. R., Dubey, K., Ren, X., Ni, J. Q. and Han, C. (2018). Phosphatidylserine Externalization Results from and Causes Neurite Degeneration in Drosophila. Cell Rep 24(9): 2273-2286. PubMed ID: 30157423

  • Junhai Han Institute of Life Sciences, Southeast University, Nanjing, China
    Wu, J., Tian, Y., Dong, W. and Han, J. (2022). Protocol for electroretinogram recording of the Drosophila compound eye. STAR Protoc 3(2): 101286. PubMed ID: 35463470

    Jin, X., Gu, P. and Han, J. (2021). Protocol for Drosophila sleep deprivation using single-chip board. STAR Protoc 2(4): 100827. PubMed ID: 34585161

    Jin, X., Tian, Y., Zhang, Z. C., Gu, P., Liu, C. and Han, J. (2021). A subset of DN1p neurons integrates thermosensory inputs to promote wakefulness via CNMa signaling. Curr Biol. PubMed ID: 33740429

    Wu, J., Ji, X., Gu, Q., Liao, B., Dong, W. and Han, J. (2021). Parallel Synaptic Acetylcholine Signals Facilitate Large Monopolar Cell Repolarization and Modulate Visual Behavior in Drosophila. J Neurosci 41(10): 2164-2176. PubMed ID: 33468565

    Gu, Q., Wu, J., Tian, Y., Cheng, S., Zhang, Z. C. and Han, J. (2020). Galphaq splice variants mediate phototransduction, rhodopsin synthesis, and retinal integrity in Drosophila. J Biol Chem. PubMed ID: 32198182

    Elvira, R., Cha, S. J., Noh, G. M., Kim, K. and Han, J. (2020). PERK-mediated eIF2alpha phosphorylation contributes to the protection of dopaminergic neurons from chronic heat stress in Drosophila. Int J Mol Sci 21(3). PubMed ID: 32013014

    Mu, Y., Tian, Y., Zhang, Z. C. and Han, J. (2019). Metallophosphoesterase regulates light-induced rhodopsin endocytosis by promoting an association between arrestin and the adaptor protein AP2. J Biol Chem. PubMed ID: 31324721

    Li, Q., Li, Y., Wang, X., Qi, J., Jin, X., Tong, H., Zhou, Z., Zhang, Z. C. and Han, J. (2017). Fbxl4 serves as a clock output molecule that regulates sleep through promotion of rhythmic degradation of the GABAA receptor. Curr Biol [Epub ahead of print]. PubMed ID: 29174887

    Liu, L., Tian, Y., Zhang, X. Y., Zhang, X., Li, T., Xie, W. and Han, J. (2017). Neurexin restricts axonal branching in columns by promoting Ephrin clustering. Dev Cell 41(1): 94-106.e104. PubMed ID: 28366281

    Tong, H., Li, Q., Zhang, Z. C., Li, Y. and Han, J. (2016). Neurexin regulates nighttime sleep by modulating synaptic transmission. Sci Rep 6: 38246. PubMed ID: 27905548
    Wan, D., Zhang, Z. C., Zhang, X., Li, Q. and Han, J. (2015). X chromosome-linked intellectual disability protein PQBP1 associates with and regulates the translation of specific mRNAs. Hum Mol Genet. PubMed ID: 26002102

  • Kyung-An Han The University of Texas at El Paso
    Sabandal, P. R., Saldes, E. B. and Han, K. A. (2022). Acetylcholine deficit causes dysfunctional inhibitory control in an aging-dependent manner. Sci Rep 12(1): 20903. PubMed ID: 36463374

    Sabandal, J. M., Sabandal, P. R., Kim, Y. C. and Han, K. A. (2020). Concerted Actions of Octopamine and Dopamine Receptors Drive Olfactory Learning. J Neurosci. PubMed ID: 32277043

    Lim, J., Fernandez, A. I., Hinojos, S. J., Aranda, G. P., James, J., Seong, C. S. and Han, K. A. (2017). The mushroom body D1 dopamine receptor controls innate courtship drive. Genes Brain Behav [Epub ahead of print]. PubMed ID: 28902472

    Kim, Y. C., Lee, H. G., Lim, J., Han, K. A. (2013) Appetitive Learning Requires the Alpha1-Like Octopamine Receptor OAMB in the Drosophila Mushroom Body Neurons. J Neurosci 33: 1672-1677. PubMed ID: 23345239

    Zhou, C., Huang, H., Kim, S. M., Lin, H., Meng, X., Han, K. A., Chiang, A. S., Wang, J. W., Jiao, R. and Rao, Y. (2012). Molecular genetic analysis of sexual rejection: roles of octopamine and its receptor OAMB in Drosophila courtship conditioning. J Neurosci 32: 14281-14287. PubMed ID: 23055498

    Makos, M. A., Han, K. A., Heien, M. L. and Ewing, A. G. (2010). Using In Vivo Electrochemistry to Study the Physiological Effects of Cocaine and Other Stimulants on the Drosophila melanogaster Dopamine Transporter. ACS Chem Neurosci 1: 74-83. PubMed ID: 20352129

  • Zhe Han Division of Molecular Medicine & Genetics, University of Michigan Medical School
    Huang, X., Fu, Y., Lee, H., Zhao, Y., Yang, W., van de Leemput, J. and Han, Z. (2023). Single-cell profiling of the developing embryonic heart in Drosophila. Development 150(16). PubMed ID: 37526610
    Zhu, J. Y., Liu, C., Huang, X., van de Leemput, J., Lee, H. and Han, Z. (2023). H3K36 Di-Methylation Marks, Mediated by Ash1 in Complex with Caf1-55 and MRG15, Are Required during Drosophila Heart Development. J Cardiovasc Dev Dis 10(7). PubMed ID:
    37504562

    Huang, W., Zhu, J. Y., Fu, Y., van de Leemput, J. and Han, Z. (2022). Lpt, trr, and Hcf regulate histone mono- and dimethylation that are essential for Drosophila heart development. Dev Biol 490: 53-65. PubMed ID: 35853502

    Wang, L., Wen, P., van de Leemput, J., Zhao, Z. and Han, Z. (2021). Slit diaphragm maintenance requires dynamic clathrin-mediated endocytosis facilitated by AP-2, Lap, Aux and Hsc70-4 in nephrocytes. Cell Biosci 11(1): 83. PubMed ID: 33975644

    Fu, Y., Huang, X., Zhang, P., van de Leemput, J. and Han, Z. (2020). Single-cell RNA sequencing identifies novel cell types in Drosophila blood. J Genet Genomics. PubMed ID: 32487456

    Wen, P., Zhang, F., Fu, Y., Zhu, J. Y. and Han, Z. (2020). Exocyst Genes Are Essential for Recycling Membrane Proteins and Maintaining Slit Diaphragm in Drosophila Nephrocytes. J Am Soc Nephrol 31(5): 1024-1034. PubMed ID: 32238475

    Fu, Y., Zhu, J. Y., Zhang, F., Richman, A., Zhao, Z. and Han, Z. (2017). Comprehensive functional analysis of Rab GTPases in Drosophila nephrocytes. Cell Tissue Res [Epub ahead of print]. PubMed ID: 28180992

    Zhu, J.Y., Fu, Y., Nettleton, M., Richman, A. and Han, Z. (2017). High throughput in vivo functional validation of candidate congenital heart disease genes in Drosophila. Elife [Epub ahead of print]. PubMed ID: 28084990

    Patel, M.V., Zhu, J.Y., Jiang, Z., Richman, A., VanBerkum, M.F. and Han, Z. (2016). Gia/Mthl5 is an aorta specific GPCR required for Drosophila heart tube morphology and normal pericardial cell positioning. Dev Biol [Epub ahead of print]. PubMed ID: 26994946

    Zhang, F., Zhao, Y. and Han, Z. (2013). An In Vivo Functional Analysis System for Renal Gene Discovery in Drosophila Pericardial Nephrocytes. J Am Soc Nephrol. PubMed ID: 23291470

  • Alfred Handler Agricultural Research Service, USDA, Gainesville, Florida
    Schetelig, M. F. and Handler, A. M. (2013). Germline transformation of the spotted wing drosophilid, Drosophila suzukii, with a piggyBac transposon vector. Genetica. PubMed ID: 23564446

    Schetelig, M. F. and Handler, A. M. (2013). A Functional Comparison of the 3xP3 Promoter by Recombinase-Mediated Cassette Exchange in Drosophila and a Tephritid Fly, Anastrepha suspensa. G3 (Bethesda). PubMed ID: 23550127

  • Greg Hannon Cold Spring Harbor Labs
    Bornelov, S., Czech, B. and Hannon, G. J. (2022). An evolutionarily conserved stop codon enrichment at the 5' ends of mammalian piRNAs. Nat Commun 13(1): 2118. PubMed ID: 35440552

    Fabry, M. H., Falconio, F. A., Joud, F., Lythgoe, E. K., Czech, B. and Hannon, G. J. (2021). Maternally inherited piRNAs direct transient heterochromatin formation at active transposons during early Drosophila embryogenesis. Elife 10. PubMed ID: 34236313

    Eastwood, E. L., Jara, K. A., Bornelov, S., Munafo, M., Frantzis, V., Kneuss, E., Barbar, E. J., Czech, B. and Hannon, G. J. (2021). Dimerisation of the PICTS complex via LC8/Cut-up drives co-transcriptional transposon silencing in Drosophila. Elife 10. PubMed ID: 33538693

    Kneuss, E., Munafo, M., Eastwood, E. L., Deumer, U. S., Preall, J. B., Hannon, G. J. and Czech, B. (2019). Specialization of the Drosophila nuclear export family protein Nxf3 for piRNA precursor export. Genes Dev 33(17-18): 1208-1220. PubMed ID: 31416967

    Munafo, M., Manelli, V., Falconio, F. A., Sawle, A., Kneuss, E., Eastwood, E. L., Seah, J. W. E., Czech, B. and Hannon, G. J. (2019). Daedalus and Gasz recruit Armitage to mitochondria, bringing piRNA precursors to the biogenesis machinery. Genes Dev. PubMed ID: 31123065

    Fagegaltier, D., Falciatori, I., Czech, B., Castel, S., Perrimon, N., Simcox, A. and Hannon, G. J. (2016). Oncogenic transformation of Drosophila somatic cells induces a functional piRNA pathway. Genes Dev 30: 1623-1635. PubMed ID: 27474441

    Yu, Y., Gu, J., Jin, Y., Luo, Y., Preall, J. B., Ma, J., Czech, B. and Hannon, G. J. (2015). Panoramix enforces piRNA-dependent cotranscriptional silencing. Science 350: 339-342. PubMed ID: 26472911

    Dequéant, M.L., Fagegaltier, D., Hu, Y., Spirohn, K., Simcox, A., Hannon, G.J. and Perrimon, N. (2015). Discovery of progenitor cell signatures by time-series synexpression analysis during Drosophila embryonic cell immortalization. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26438832

    Fagegaltier, D., Konig, A., Gordon, A., Lai, E. C., Gingeras, T. R., Hannon, G. J. and Shcherbata, H. R. (2014). A Genome-Wide Survey of Sexually Dimorphic Expression of Drosophila miRNAs Identifies the Steroid Hormone-Induced miRNA let-7 as a Regulator of Sexual Identity. Genetics. PubMed ID: 25081570

    Zhou, R., Mohr, S., Hannon, G. J. and Perrimon, N. (2014). Inducing RNAi in Drosophila Cells by Soaking with dsRNA. Cold Spring Harb Protoc 2014. PubMed ID: 24786505
    Chang, K., Marran, K., Valentine, A. and Hannon, G. J. (2014). Generation of Transgenic Drosophila Expressing shRNAs in the miR-1 Backbone. Cold Spring Harb Protoc 2014. PubMed ID: 24786506

  • Bill Hansson Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena
    Das Chakraborty, S., Chang, H., Hansson, B. S. and Sachse, S. (2022). Higher-order olfactory neurons in the lateral horn support odor valence and odor identity coding in Drosophila. Elife 11. PubMed ID: 35621267

    Prelic, S., Pal Mahadevan, V., Venkateswaran, V., Lavista-Llanos, S., Hansson, B. S. and Wicher, D. (2021). Functional Interaction Between Drosophila Olfactory Sensory Neurons and Their Support Cells. Front Cell Neurosci 15: 789086. PubMed ID: 35069116

    Keesey, I. W., Grabe, V., Knaden, M. and Hansson, B. S. (2020). Divergent sensory investment mirrors potential speciation via niche partitioning across Drosophila. Elife 9. PubMed ID: 32602834

    Grabe, V., Schubert, M., Strube-Bloss, M., Reinert, A., Trautheim, S., Lavista-Llanos, S., Fiala, A., Hansson, B. S. and Sachse, S. (2019). Odor-induced multi-level inhibitory maps in Drosophila. eNeuro. PubMed ID: 31888962

    Keesey, I. W., Grabe, V., Gruber, L., Koerte, S., Obiero, G. F., Bolton, G., Khallaf, M. A., Kunert, G., Lavista-Llanos, S., Valenzano, D. R., Rybak, J., Barrett, B. A., Knaden, M. and Hansson, B. S. (2019). Inverse resource allocation between vision and olfaction across the genus Drosophila. Nat Commun 10(1): 1162. PubMed ID: 30858374

    Gruber, L., Rybak, J., Hansson, B. S. and Cantera, R. (2018). Synaptic spinules in the olfactory circuit of Drosophila melanogaster. Front Cell Neurosci 12: 86. PubMed ID: 29636666

    Strube-Bloss, M. F., Grabe, V., Hansson, B. S. and Sachse, S. (2017). Calcium imaging revealed no modulatory effect on odor-evoked responses of the Drosophila antennal lobe by two populations of inhibitory local interneurons. Sci Rep 7(1): 7854. PubMed ID: 28798324

    Keesey, I. W., Koerte, S., Khallaf, M. A., Retzke, T., Guillou, A., Grosse-Wilde, E., Buchon, N., Knaden, M. and Hansson, B. S. (2017). Pathogenic bacteria enhance dispersal through alteration of Drosophila social communication. Nat Commun 8(1): 265. PubMed ID: 28814724

    Seki, Y., Dweck, H. K. M., Rybak, J., Wicher, D., Sachse, S. and Hansson, B. S. (2017). Olfactory coding from the periphery to higher brain centers in the Drosophila brain. BMC Biol 15(1): 56. PubMed ID: 28666437

    Getahun, M. N., Thoma, M., Lavista-Llanos, S., Keesey, I., Fandino, R. A., Knaden, M., Wicher, D., Olsson, S. B. and Hansson, B. S. (2016). Intracellular regulation of the insect chemoreceptor complex impacts odor localization in flying insects. J Exp Biol [Epub ahead of print]. PubMed ID: 27591307

    Grabe, V., Baschwitz, A., Dweck, H. K., Lavista-Llanos, S., Hansson, B. S. and Sachse, S. (2016). Elucidating the neuronal architecture of olfactory glomeruli in the Drosophila antennal lobe. Cell Rep 16: 3401-3413. PubMed ID: 27653699

  • Susan Harbison Laboratory of Systems Genetics, NHLBI, NIH
    Souto-Maior, C., Serrano Negron, Y. L. and Harbison, S. T. (2023). Nonlinear expression patterns and multiple shifts in gene network interactions underlie robust phenotypic change in Drosophila melanogaster selected for night sleep duration. PLoS Comput Biol 19(8): e1011389. PubMed ID: 37561813

    Souto-Maior, C., Serrano Negron, Y. L. and Harbison, S. T. (2020). Natural selection on sleep duration in Drosophila melanogaster. Sci Rep 10(1): 20652. PubMed ID: 33244154

    Kumar, S., Tunc, I., Tansey, T. R., Pirooznia, M. and Harbison, S. T. (2020). Identification of Genes Contributing to a Long Circadian Period in Drosophila Melanogaster. J Biol Rhythms: 748730420975946. PubMed ID: 33274675

    Kumar, S., Smith, K. R., Serrano Negron, Y. L. and Harbison, S. T. (2019). Short-Term Memory Deficits in the SLEEP Inbred Panel. Clocks Sleep 1(4): 471-488. PubMed ID: 32596662

    Harbison, S. T., Kumar, S., Huang, W., McCoy, L. J., Smith, K. R. and Mackay, T. F. C. (2018). Genome-Wide Association Study of Circadian Behavior in Drosophila melanogaster. Behav Genet. PubMed ID: 30341464

    Serrano Negron, Y. L., Hansen, N. F. and Harbison, S. T. (2018). The Sleep inbred panel, a collection of inbred Drosophila melanogaster with extreme long and short sleep duration. G3 (Bethesda). PubMed ID: 29991508

    Wu, K. J., Kumar, S., Serrano Negron, Y. L. and Harbison, S. T. (2017). Genotype influences day-to-day variability in sleep in Drosophila melanogaster. Sleep. PubMed ID: 29228366

    Harbison, S. T., Serrano Negron, Y. L., Hansen, N. F. and Lobell, A. S. (2017). Selection for long and short sleep duration in Drosophila melanogaster reveals the complex genetic network underlying natural variation in sleep. PLoS Genet 13(12): e1007098. PubMed ID: 29240764

    Lobell, A. S., Kaspari, R. R., Serrano Negron, Y. L. and Harbison, S. T. (2017). The Genetic architecture of ovariole number in Drosophila melanogaster: Genes with major, quantitative, and pleiotropic effects. G3 (Bethesda). PubMed ID: 28550012

    Huang, W., Lyman, R. F., Lyman, R. A., Carbone, M. A., Harbison, S. T., Magwire, M. M. and Mackay, T. F. (2016). Spontaneous mutations and the origin and maintenance of quantitative genetic variation. Elife 5. PubMed ID: 27213517

    Jumbo-Lucioni, P., Bu, S., Harbison, S. T., Slaughter, J. C., Mackay, T. F., Moellering, D. R. and De Luca, M. (2012). Nuclear genomic control of naturally occurring variation in mitochondrial function in Drosophila melanogaster. BMC Genomics 13: 659. PubMed ID: 23171078

  • Roger Hardie Department of Physiology, Development and Neuroscience, University of Cambridge
    Li, X., Abou Tayoun, A., Song, Z., Dau, A., Rien, D., Jaciuch, D., Dongre, S., Blanchard, F., Nikolaev, A., Zheng, L., Bollepalli, M. K., Chu, B., Hardie, R. C., Dolph, P. J. and Juusola, M. (2019). Ca(2+)-activated K(+) channels reduce network excitability, improving adaptability and energetics for transmitting and perceiving sensory information. J Neurosci. PubMed ID: 31350259

    Liu, C. H., Bollepalli, M. K., Long, S. V., Asteriti, S., Tan, J., Brill, J. A. and Hardie, R. C. (2018). Genetic dissection of the phosphoinositide cycle in Drosophila photoreceptors. J Cell Sci 131(8). PubMed ID: 29567856

    Cao, J., Bollepalli, M. K., Hu, Y., Zhang, J., Li, Q., Li, H., Chang, H., Xiao, F., Hardie, R. C., Rong, Y. S. and Hu, W. (2017). A single residue mutation in the Galphaq subunit of the G protein complex causes blindness in Drosophila. G3 (Bethesda). PubMed ID: 29158337

    Bollepalli, M. K., Kuipers, M. E., Liu, C. H., Asteriti, S. and Hardie, R. C. (2017). Phototransduction in Drosophila is compromised by Gal4 expression but not by InsP3 receptor knockdown or mutation. eNeuro 4(3). PubMed ID: 28660247

    Asteriti, S., Liu, C. H. and Hardie, R. C. (2017). Calcium signalling in Drosophila photoreceptors measured with GCaMP6f. Cell Calcium. PubMed ID: 28238353

    Friederich, U., Billings, S. A., Hardie, R. C., Juusola, M. and Coca, D. (2016). Fly photoreceptors encode phase congruency. PLoS One 11: e0157993. PubMed ID: 27336733

    Dau, A., Friederich, U., Dongre, S., Li, X., Bollepalli, M. K., Hardie, R. C. and Juusola, M. (2016). Evidence for Dynamic Network Regulation of Drosophila Photoreceptor Function from Mutants Lacking the Neurotransmitter Histamine. Front Neural Circuits 10: 19. PubMed ID: 27047343

    Hardie, R. C., Liu, C. H., Randall, A. S. and Sengupta, S. (2015). In vivo tracking of phosphoinositides in Drosophila photoreceptors. J Cell Sci [Epub ahead of print]. PubMed ID: 26483384

    Randall, A. S., Liu, C. H., Chu, B., Zhang, Q., Dongre, S. A., Juusola, M., Franze, K., Wakelam, M. J. and Hardie, R. C. (2015). Speed and sensitivity of phototransduction in Drosophila depend on degree of saturation of membrane phospholipids. J Neurosci 35: 2731-2746. PubMed ID: 25673862

    Hardie, R. C. and Juusola, M. (2015). Phototransduction in Drosophila. Curr Opin Neurobiol 34C: 37-45. PubMed ID: 25638280

  • Nicholas Harden Simon Fraser University, Vancouver
    Rivas, G. B. S., Zhou, J., Merlin, C. and Hardin, P. E. (2021). CLOCKWORK ORANGE promotes CLOCK-CYCLE activation via the putative Drosophila ortholog of CLOCK INTERACTING PROTEIN CIRCADIAN. Curr Biol. PubMed ID: 34331859

    Yoo, B., Kim, H. Y., Chen, X., Shen, W., Jang, J. S., Stein, S. N., Cormier, O., Pereira, L., Shih, C. R. Y., Krieger, C., Reed, B., Harden, N. and Wang, S. J. H. (2021). 20-hydroxyecdysone (20E) signaling regulates amnioserosa morphogenesis during Drosophila dorsal closure: EcR modulates gene expression in a complex with the AP-1 subunit, Jun. Biol Open. PubMed ID: 34296248

    Reed, B. and Harden, N. (2017). Studying nonproliferative roles for Egfr signaling in tissue morphogenesis using Dorsal closure of the Drosophila embryo. Methods Mol Biol 1652: 229-256. PubMed ID: 28791646

    Wang, S., Yoo, S., Kim, H. Y., Wang, M., Zheng, C., Parkhouse, W., Krieger, C. and Harden, N. (2015). Detection of in situ protein-protein complexes at the Drosophila larval neuromuscular junction using proximity ligation assay. J Vis Exp. PubMed ID: 25650626

    Vlachos, S., Jangam, S., Conder, R., Chou, M., Nystul, T. and Harden, N. (2015). A Pak-regulated cell intercalation event leading to a novel radial cell polarity is involved in positioning of the follicle stem cell niche in the Drosophila ovary. Development 142: 82-91. PubMed ID: 25516970

    Wang, S. J., Tsai, A., Wang, M., Yoo, S., Kim, H. Y., Yoo, B., Chui, V., Kisiel, M., Stewart, B., Parkhouse, W., Harden, N. and Krieger, C. (2014). Phospho-regulated Drosophila adducin is a determinant of synaptic plasticity in a complex with Dlg and PIP2 at the larval neuromuscular junction. Biol Open. PubMed ID: 25416060

    Shen, W., Chen, X., Cormier, O., Cheng, D. C., Reed, B., Harden, N. (2013) Modulation of Morphogenesis by Egfr during Dorsal Closure in Drosophila. PLoS One 8: e60180. PubMed ID: 23579691

    Wang, S., Yang, J., Tsai, A., Kuca, T., Sanny, J., Lee, J., Dong, K., Harden, N. and Krieger, C. (2011). Drosophila adducin regulates Dlg phosphorylation and targeting of Dlg to the synapse and epithelial membrane. Dev Biol 357: 392-403. PubMed ID: 21791202

    Vlachos, S. and Harden, N. (2011). Genetic evidence for antagonism between Pak protein kinase and Rho1 small GTPase signaling in regulation of the actin cytoskeleton during Drosophila oogenesis. Genetics 187: 501-512. PubMed ID: 21098722

  • Paul Hardin Department of Biology, Texas A&M, College Station
    Gunawardhana, K. L., Rivas, G. B. S., Caster, C. and Hardin, P. E. (2021). Crosstalk between vrille transcripts, proteins, and regulatory elements controlling circadian rhythms and development in Drosophila. iScience 24(1): 101893. PubMed ID: 33364582

    Mahesh, G., Rivas, G. B. S., Caster, C., Ost, E. B., Amunugama, R., Jones, R., Allen, D. L. and Hardin, P. E. (2020). Proteomic analysis of Drosophila CLOCK complexes identifies rhythmic interactions with SAGA and Tip60 complex component NIPPED-A. Sci Rep 10(1): 17951. PubMed ID: 33087840

    Liu, T., Mahesh, G., Yu, W. and Hardin, P. E. (2017). CLOCK stabilizes CYCLE to initiate clock function in Drosophila. Proc Natl Acad Sci U S A 114(41): 10972-10977. PubMed ID: 28973907

    Gunawardhana, K. L. and Hardin, P. E. (2017). VRILLE vontrols PDF neuropeptide accumulation and arborization rhythms in small ventrolateral neurons to drive rhythmic behavior in Drosophila. Curr Biol 27(22): 3442-3453.e3444. PubMed ID: 29103936

    Agrawal, P., Houl, J. H., Gunawardhana, K. L., Liu, T., Zhou, J., Zoran, M. J. and Hardin, P. E. (2017). Drosophila CRY entrains clocks in body tissues to light and maintains passive membrane properties in a non-clock body tissue independent of light. Curr Biol [Epub ahead of print]. PubMed ID: 28781048

    Zhou, J., Yu, W. and Hardin, P. E. (2016). CLOCKWORK ORANGE enhances PERIOD mediated rhythms in transcriptional repression by antagonizing E-box binding by CLOCK-CYCLE. PLoS Genet 12: e1006430. PubMed ID: 27814361

    Agrawal, P. and Hardin, P. E. (2016). The Drosophila receptor protein tyrosine phosphatase LAR is required for development of circadian pacemaker neuron processes that support rhythmic activity in constant darkness but not during light/dark cycles. J Neurosci 36: 3860-3870. PubMed ID: 27030770

    Liu, T., Mahesh, G., Houl, J. H. and Hardin, P. E. (2015). Circadian activators are expressed days before they initiate clock function in late pacemaker neurons from drosophila. J Neurosci 35: 8662-8671. PubMed ID: 26041931

    Zhou, J., Yu, W. and Hardin, P. E. (2015). ChIPping Away at the Drosophila Clock. Methods Enzymol 551: 323-347. PubMed ID: 25662463

    Glossop, N. R., Gummadova, J. O., Ghangrekar, I., Hardin, P. E. and Coutts, G. A. (2014). Effects of TWIN-OF-EYELESS on Clock Gene Expression and Central-Pacemaker Neuron Development in Drosophila. J Biol Rhythms 29: 151-166. PubMed ID: 24916389

  • Iswar Hariharan Molecular and Cell Biology, U. C. Berkeley
    Spitzer, D. C., Sun, W. Y., Rodríguez-Vargas, A. and Hariharan, I. K. (2023). The cell adhesion molecule Echinoid promotes tissue survival and separately restricts tissue overgrowth in Drosophila imaginal discs. bioRxiv. PubMed ID: 37577631

    Worley, M. I., Everetts, N. J., Yasutomi, R., Chang, R. J., Saretha, S., Yosef, N. and Hariharan, I. K. (2022). Ets21C sustains a pro-regenerative transcriptional program in blastema cells of Drosophila imaginal discs. Curr Biol 32(15): 3350-3364.e3356. PubMed ID: 35820420

    Everetts, N. J., Worley, M. I., Yasutomi, R., Yosef, N. and Hariharan, I. K. (2021). Single-cell transcriptomics of the Drosophila wing disc reveals instructive epithelium-to-myoblast interactions. Elife 10. PubMed ID: 33749594

    Emmons-Bell, M. and Hariharan, I. K. (2021). Membrane potential regulates Hedgehog signalling in the Drosophila wing imaginal disc. EMBO Rep: e51861. PubMed ID: 33629503

    Bairzin, J. C. D., Emmons-Bell, M. and Hariharan, I. K. (2020). The Hippo pathway coactivator Yorkie can reprogram cell fates and create compartment-boundary-like interactions at clone margins. Sci Adv 6(50). PubMed ID: 33298454

    Harris, R. E., Stinchfield, M. J., Nystrom, S. L., McKay, D. J. and Hariharan, I. K. (2020). Damage-responsive, maturity-silenced enhancers regulate multiple genes that direct regeneration in Drosophila. Elife 9. PubMed ID: 32490812

    Lahvic, J. L. and Hariharan, I. K. (2019). Harnessing epithelial homeostatic mechanisms to fight cancer. Mol Biol Cell 30(14): 1641-1644. PubMed ID: 31246542

    Setiawan, L., Pan, X., Woods, A. L., O'Connor, M. B. and Hariharan, I. K. (2018). The BMP2/4 ortholog Dpp can function as an inter-organ signal that regulates developmental timing. Life Sci Alliance 1(6): e201800216. PubMed ID: 30515478

    Worley, M. I., Alexander, L. A. and Hariharan, I. K. (2018). CtBP impedes JNK- and Upd/STAT-driven cell fate misspecifications in regenerating Drosophila imaginal discs. Elife 7. PubMed ID: 29372681

    Yoo, S. K., Pascoe, H. G., Pereira, T., Kondo, S., Jacinto, A., Zhang, X. and Hariharan, I. K. (2016). Plexins function in epithelial repair in both Drosophila and zebrafish. Nat Commun 7: 12282. PubMed ID: 27452696

    Bosch, J.A., Sumabat, T.M. and Hariharan, I.K. (2016). Persistence of RNAi-mediated knockdown in Drosophila complicates mosaic analysis yet enables highly sensitive lineage tracing. Genetics [Epub ahead of print]. PubMed ID: 26984059

    Harris, R.E., Setiawan, L., Saul, J. and Hariharan, I.K. (2016). Localized epigenetic silencing of a damage-activated WNT enhancer limits regeneration in mature Drosophila imaginal discs. Elife [Epub ahead of print]. PubMed ID: 26840050

  • Greg Harris San Diego State University
    Ratliff, E. P., Mauntz, R. E., Kotzebue, R. W., Gonzalez, A., Achal, M., Barekat, A., Finley, K. A., Sparhawk, J. M., Robinson, J. E., Herr, D. R., Harris, G. L., Joiner, W. J. and Finley, K. D. (2015). Aging and Autophagic Function Influences the Progressive Decline of Adult Drosophila Behaviors. PLoS One 10: e0132768. PubMed ID: 26182057

    Walls, S. M., Attle, S. J., Brulte, G. B., Walls, M. L., Finley, K. D., Chatfield, D. A., Herr, D. R. and Harris, G. L. (2013). Identification of Sphingolipid Metabolites That Induce Obesity via Misregulation of Appetite, Caloric Intake and Fat Storage in Drosophila. PLoS Genet 9: e1003970. PubMed ID: 24339790

    Fyrst, H., Zhang, X., Herr, D. R., Byun, H. S., Bittman, R., Phan, V. H., Harris, G. L. and Saba, J. D. (2008). Identification and characterization by electrospray mass spectrometry of endogenous Drosophila sphingadienes. J Lipid Res 49: 597-606. PubMed ID: 18156591

  • Tony Harris Department of Cell & Systems Biology, University of Toronto
    Hunt, E. L., Rai, H. and Harris, T. J. C. (2022). SCAR/WAVE complex recruitment to a supracellular actomyosin cable by myosin activators and a junctional Arf-GEF during Drosophila dorsal closure. Mol Biol Cell 33(8): br12. PubMed ID: 35476600 Sharma, M., Jiang, T., Jiang, Z. C., Moguel-Lehmer, C. E. and Harris, T. J. (2021). Emergence of a smooth interface from growth of a dendritic network against a mechanosensitive contractile material. Elife 10. PubMed ID: 34423780

    West, J. J. and Harris, T. J. C. (2020). The Arf-GEF Steppke promotes F-actin accumulation, cell protrusions and tissue sealing during Drosophila dorsal closure. PLoS One 15(11): e0239357. PubMed ID: 33186390

    Zheng, S., West, J. J., Yu, C. G. and Harris, T. J. C. (2019). Arf-GEF localization and function at myosin-rich adherens junctions via coiled-coil hetero-dimerization with an adaptor protein. Mol Biol Cell: mbcE19100566. PubMed ID: 31693432

    Jiang, T. and Harris, T. J. C. (2019). Par-1 controls the composition and growth of cortical actin caps during Drosophila embryo cleavage. J Cell Biol. PubMed ID: 31641019

    Zhang, Y., Yu, J. C., Jiang, T., Fernandez-Gonzalez, R. and Harris, T. J. C. (2018). Collision of expanding actin caps with actomyosin borders for cortical bending and mitotic rounding in a syncytium. Dev Cell. Pubmed ID: 29804877

    Rodrigues, F. F., Shao, W. and Harris, T. J. (2016). The Arf GAP Asap promotes Arf1 function at the Golgi for cleavage furrow biosynthesis in Drosophila. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27535433

    Lee, D. M., Rodrigues, F. F., Yu, C. G., Swan, M. and Harris, T. J. (2015). PH Domain-Arf G Protein Interactions Localize the Arf-GEF Steppke for Cleavage Furrow Regulation in Drosophila. PLoS One 10: e0142562. PubMed ID: 26556630

    Jiang, T., McKinley, R. F., McGill, M. A., Angers, S. and Harris, T. J. (2015). A Par-1-Par-3-centrosome cell polarity pathway and its tuning for isotropic cell adhesion. Curr Biol 25: 2701-2708. PubMed ID: 26455305

    Hunter, M.V., Lee, D.M., Harris, T.J. and Fernandez-Gonzalez, R. (2015). Polarized E-cadherin endocytosis directs actomyosin remodeling during embryonic wound repair. J Cell Biol [Epub ahead of print]. PubMed ID: 26304727

    Lee, D. M., Wilk, R., Hu, J., Krause, H. M. and Harris, T. J. (2015). Germ Cell Segregation from the Drosophila Soma Is Controlled by an Inhibitory Threshold Set by the Arf-GEF Steppke. Genetics. PubMed ID: 25971667

  • Doug Harrison Department of Biology, University of Kentucky
    Gaskill, M. M., Gibson, T. J., Larson, E. D. and Harrison, M. M. (2021). GAF is essential for zygotic genome activation and chromatin accessibility in the early Drosophila embryo. Elife 10. PubMed ID: 33720012

    Wang, L., Sexton, T. R., Venard, C., Giedt, M., Guo, Q., Chen, Q. and Harrison, D. A. (2014). Pleiotropy of the Drosophila JAK pathway cytokine Unpaired 3 in development and aging. Dev Biol. PubMed ID: 25245869

    Hayashi, Y., Sexton, T. R., Dejima, K., Perry, D. W., Takemura, M., Kobayashi, S., Nakato, H. and Harrison, D. A. (2012). Glypicans regulate JAK/STAT signaling and distribution of the Unpaired morphogen. Development 139: 4162-4171. PubMed ID: 23093424

    Cai, W., Rudolph, J. L., Harrison, S. M., Jin, L., Frantz, A. L., Harrison, D. A. and Andres, D. A. (2011). An evolutionarily conserved Rit GTPase-p38 MAPK signaling pathway mediates oxidative stress resistance. Mol Biol Cell 22: 3231-3241. PubMed ID: 21737674

    Zhu, F., Parthasarathy, R., Bai, H., Woithe, K., Kaussmann, M., Nauen, R., Harrison, D. A. and Palli, S. R. (2010). A brain-specific cytochrome P450 responsible for the majority of deltamethrin resistance in the QTC279 strain of Tribolium castaneum. Proc Natl Acad Sci U S A 107: 8557-8562. PubMed ID: 20410462

  • Jon Harrison School of Life Sciences, Arizona State University, Tempe
    Callier, V., Hand, S. C., Campbell, J. B., Biddulph, T. and Harrison, J. F. (2015). Developmental changes in hypoxic exposure and responses to anoxia in Drosophila melanogaster. J Exp Biol. PubMed ID: 26206351

    Farzin, M., Albert, T., Pierce, N., VandenBrooks, J. M., Dodge, T. and Harrison, J. F. (2014). Acute and chronic effects of atmospheric oxygen on the feeding behavior of Drosophila melanogaster larvae. J Insect Physiol. PubMed ID: 25008193

    Callier, V., Shingleton, A. W., Brent, C. S., Ghosh, S. M., Kim, J. and Harrison, J. F. (2013). The role of reduced oxygen in the developmental physiology of growth and metamorphosis initiation in Drosophila melanogaster. J Exp Biol 216: 4334-4340. PubMed ID: 24259256

    Schilman, P. E., Waters, J. S., Harrison, J. F. and Lighton, J. R. (2011). Effects of temperature on responses to anoxia and oxygen reperfusion in Drosophila melanogaster. J Exp Biol 214: 1271-1275. PubMed ID: 21430203

    Heinrich, E. C., Farzin, M., Klok, C. J. and Harrison, J. F. (2011). The effect of developmental stage on the sensitivity of cell and body size to hypoxia in Drosophila melanogaster. J Exp Biol 214: 1419-1427. PubMed ID: 21490250

  • Melissa Harrison Biomolecular Chemistory, University of Wisconsin, Madison
    Larson, E. D., Komori, H., Gibson, T. J., Ostgaard, C. M., Hamm, D. C., Schnell, J. M., Lee, C. Y. and Harrison, M. M. (2021). Cell-type-specific chromatin occupancy by the pioneer factor Zelda drives key developmental transitions in Drosophila. Nat Commun 12(1): 7153. PubMed ID: 34887421

    McDaniel, S. L., Hollatz, A. J., Branstad, A. M., Gaskill, M. M., Fox, C. A. and Harrison, M. M. (2019). Tissue-specific DNA replication defects in Drosophila melanogaster caused by a Meier-Gorlin syndrome mutation in Orc4. Genetics. PubMed ID: 31818869

    McDaniel, S. L., Gibson, T. J., Schulz, K. N., Fernandez Garcia, M., Nevil, M., Jain, S. U., Lewis, P. W., Zaret, K. S. and Harrison, M. M. (2019). Continued activity of the pioneer factor Zelda is required to drive zygotic genome activation. Mol Cell 74(1): 185-195.e184. PubMed ID: 30797686

    Hamm, D. C., Larson, E. D., Nevil, M., Marshall, K. E., Bondra, E. R. and Harrison, M. M. (2017). A conserved maternal-specific repressive domain in Zelda revealed by Cas9-mediated mutagenesis in Drosophila melanogaster. PLoS Genet 13(12): e1007120. PubMed ID: 29261646

    Janssens, D. H., Hamm, D. C., Anhezini, L., Xiao, Q., Siller, K. H., Siegrist, S. E., Harrison, M. M. and Lee, C. Y. (2017). An Hdac1/Rpd3-poised circuit balances continual self-renewal and rapid restriction of developmental potential during asymmetric stem cell division. Dev Cell 40(4): 367-380.e367. PubMed ID: 28245922

    Nevil, M., Bondra, E. R., Schulz, K. N., Kaplan, T. and Harrison, M. M. (2016). Stable binding of the conserved transcription factor Grainy head to Its target genes throughout Drosophila melanogaster development. Genetics [Epub ahead of print]. PubMed ID: 28007888

    Schulz, K.N., Bondra, E.R., Moshe, A., Villalta, J.E., Lieb, J.D., Kaplan, T., McKay, D.J. and Harrison, M.M. (2015). Zelda is differentially required for chromatin accessibility, transcription-factor binding and gene expression in the early Drosophila embryo. Genome Res [Epub ahead of print]. PubMed ID: 26335634

    Hamm, D. C., Bondra, E. R. and Harrison, M. M. (2014). Transcriptional activation is a conserved feature of the early embryonic factor Zelda that requires a cluster of four zinc fingers for DNA binding and a low-complexity activation domain. J Biol Chem [Epub ahead of print]. PubMed ID: 25538246

    Gratz, S. J., Cummings, A. M., Nguyen, J. N., Hamm, D. C., Donohue, L. K., Harrison, M. M., Wildonger, J. and O'Connor-Giles, K. M. (2013). Genome engineering of Drosophila with the CRISPR RNA-guided Cas9 nuclease. Genetics 194: 1029-1035. PubMed ID: 23709638

    Harrison, M. M., Li, X. Y., Kaplan, T., Botchan, M. R. and Eisen, M. B. (2011). Zelda binding in the early Drosophila melanogaster embryo marks regions subsequently activated at the maternal-to-zygotic transition. PLoS Genet 7: e1002266. PubMed ID: 22028662

  • Craig Hart Department of Biological Sciences, Louisana State University, Baton Rouge
    McKowen, J. K., Avva, S., Maharjan, M., Duarte, F. M., Tome, J. M., Judd, J., Wood, J. L., Negedu, S., Dong, Y., Lis, J. T. and Hart, C. M. (2022). The Drosophila BEAF insulator protein interacts with the polybromo subunit of the PBAP chromatin remodeling complex. G3 (Bethesda) 12(11). PubMed ID: 36029240

    Maharjan, M., McKowen, J. K. and Hart, C. M. (2020). Overlapping but Distinct Sequences Play Roles in the Insulator and Promoter Activities of the Drosophila BEAF-dependent scs' Insulator. Genetics. PubMed ID: 32554599

    Dong, Y., Avva, S., Maharjan, M., Jacobi, J. and Hart, C. M. (2020). Promoter-Proximal Chromatin Domain Insulator Protein BEAF Mediates Local and Long-Range Communication with a Transcription Factor and Directly Activates a Housekeeping Promoter in Drosophila. Genetics. PubMed ID: 32179582

    Shrestha, S., Oh, D. H., McKowen, J. K., Dassanayake, M. and Hart, C. M. (2018). 4C-seq characterization of Drosophila BEAF binding regions provides evidence for highly variable long-distance interactions between active chromatin. PLoS One 13(9): e0203843. PubMed ID: 30248133

    Avva, S. V. and Hart, C. M. (2016). Characterization of the Drosophila BEAF-32A and BEAF-32B insulator proteins. PLoS One 11: e0162906. PubMed ID: 27622635

    Hart, C. M. (2013). Do the BEAF insulator proteins regulate genes involved in cell polarity and neoplastic growth? Dev Biol. PubMed ID: 24211761

    Soshnev, A. A., He, B., Baxley, R. M., Jiang, N., Hart, C. M., Tan, K. and Geyer, P. K. (2012). Genome-wide studies of the multi-zinc finger Drosophila Suppressor of Hairy-wing protein in the ovary. Nucleic Acids Res 40: 5415-5431. PubMed ID: 22406832

    Roy, S., Jiang, N. and Hart, C. M. (2011). Lack of the Drosophila BEAF insulator proteins alters regulation of genes in the Antennapedia complex. Mol Genet Genomics 285: 113-123. PubMed ID: 21132442

  • Peter Harte Department of Genetics, School of Medicine, Case Western Reserve University
    Du, J., Zhang, J., He, T., Li, Y., Su, Y., Tie, F., Liu, M., Harte, P. J. and Zhu, A. J. (2016). Stuxnet facilitates the degradation of Polycomb protein during development. Dev Cell 37: 507-519. PubMed ID: 27326929

    Tie, F., Banerjee, R., Fu, C., Stratton, C.A., Fang, M. and Harte, P.J. (2016). Polycomb inhibits histone acetylation by CBP by binding directly to its catalytic domain. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26802126

    Tie, F., Banerjee, R., Saiakhova, A. R., Howard, B., Monteith, K. E., Scacheri, P. C., Cosgrove, M. S. and Harte, P. J. (2014). Trithorax monomethylates histone H3K4 and interacts directly with CBP to promote H3K27 acetylation and antagonize Polycomb silencing. Development 141: 1129-1139. PubMed ID: 24550119

    Mason-Suares, H., Tie, F., Yan, C., Harte, P. J. (2013) Polycomb silencing of the Drosophila 4E-BP gene regulates imaginal disc cell growth. Dev Biol. PubMed ID: 23523430

    Siebold, A. P., Banerjee, R., Tie, F., Kiss, D. L., Moskowitz, J. and Harte, P. J. (2010). Polycomb Repressive Complex 2 and Trithorax modulate Drosophila longevity and stress resistance. Proc Natl Acad Sci U S A 107: 169-174. PubMed ID: 20018689

    Tie, F., Banerjee, R., Stratton, C. A., Prasad-Sinha, J., Stepanik, V., Zlobin, A., Diaz, M. O., Scacheri, P. C. and Harte, P. J. (2009). CBP-mediated acetylation of histone H3 lysine 27 antagonizes Drosophila Polycomb silencing. Development 136: 3131-3141. PubMed ID: 19700617

  • Volker Hartenstein Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles
    Farnworth, M. S., Bucher, G. and Hartenstein, V. (2022). An atlas of the developing Tribolium castaneum brain reveals conservation in anatomy and divergence in timing to Drosophila melanogaster. J Comp Neurol. PubMed ID: 35535818

    Hildebrandt, K., Kloppel, C., Gogel, J., Hartenstein, V. and Walldorf, U. (2022). Orthopedia expression during Drosophila melanogaster nervous system development and its regulation by microRNA-252. Dev Biol 492: 87-100. PubMed ID: 36179878

    Zhao, Y., Duan, J., Han, Z., Engstrom, Y. and Hartenstein, V. (2022). Identification of a GABAergic neuroblast lineage modulating sweet and bitter taste sensitivity. Curr Biol. PubMed ID: 36347251

    Hardcastle, B. J., Omoto, J. J., Kandimalla, P., Nguyen, B. M., Keleş, M. F., Boyd, N. K., Hartenstein, V. and Frye, M. A. (2021). A visual pathway for skylight polarization processing in Drosophila. Elife 10. PubMed ID: 33755020

    Hartenstein, V., Yuan, M., Younossi-Hartenstein, A., Karandikar, A., Bernardo-Garcia, F. J., Sprecher, S. and Knust, E. (2019). Serial electron microscopic reconstruction of the Drosophila larval eye: Photoreceptors with a rudimentary rhabdomere of microvillar-like processes. Dev Biol. PubMed ID: 31158364

    Andrade, I. V., Riebli, N., Nguyen, B. M., Omoto, J. J., Cardona, A. and Hartenstein, V. (2019). Developmentally arrested precursors of pontine neurons establish an embryonic blueprint of the Drosophila central complex. Curr Biol 29(3): 412-425.e413. PubMed ID: 30661802

    Omoto, J. J., Nguyen, B. M., Kandimalla, P., Lovick, J. K., Donlea, J. M. and Hartenstein, V. (2018). Neuronal constituents and putative interactions within the Drosophila ellipsoid body neuropil. Front Neural Circuits 12: 103. PubMed ID: 30546298

    Deng, H., Takashima, S., Paul, M., Guo, M. and Hartenstein, V. (2018). Mitochondrial dynamics regulates Drosophila intestinal stem cell differentiation. Cell Death Discov 5: 17. PubMed ID: 30062062

    Donlea, J. M., Pimentel, D., Talbot, C. B., Kempf, A., Omoto, J. J., Hartenstein, V. and Miesenbock, G. (2018). Recurrent circuitry for balancing sleep need and sleep. Neuron 97(2): 378-389.e374. PubMed ID: 29307711

    Lovick, J. K., Omoto, J. J., Ngo, K. T. and Hartenstein, V. (2017). Development of the anterior visual input pathway to the Drosophila central complex. J Comp Neurol. PubMed ID: 28675433

    Boyan, G., Liu, Y., Khalsa, S. K. and Hartenstein, V. (2017). A conserved plan for wiring up the fan-shaped body in the grasshopper and Drosophila. Dev Genes Evol 227(4): 253-269. PubMed ID: 28752327

    Kendroud, S., Bohra, A. A., Kuert, P. A., Nguyen, B., Guillermin, O., Sprecher, S. G., Reichert, H., VijayRaghavan, K. and Hartenstein, V. (2017). Structure and development of the subesophageal zone of the Drosophila brain. II. Sensory compartments. J Comp Neurol. PubMed ID: 28875566

  • Daniel Hartl Department of Organismic and Evolutionary Biology, Harvard University
    Lienard, M. A., Araripe, L. O. and Hartl, D. L. (2016). Neighboring genes for DNA-binding proteins rescue male sterility in Drosophila hybrids. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27357670

    Ayroles, J. F., Buchanan, S. M., O'Leary, C., Skutt-Kakaria, K., Grenier, J. K., Clark, A. G., Hartl, D. L. and de Bivort, B. L. (2015). Behavioral idiosyncrasy reveals genetic control of phenotypic variability. Proc Natl Acad Sci U S A. PubMed ID: 25953335

    Lemos, B., Branco, A. T., Jiang, P. P., Hartl, D. L. and Meiklejohn, C. D. (2013). Genome-Wide Gene Expression Effects of Sex Chromosome Imprinting in Drosophila. G3 (Bethesda). PubMed ID: 24318925

    Corbett-Detig, R. B., Zhou, J., Clark, A. G., Hartl, D. L. and Ayroles, J. F. (2013). Genetic incompatibilities are widespread within species. Nature. PubMed ID: 24196712

    Meiklejohn, C. D., Coolon, J. D., Hartl, D. L. and Wittkopp, P. J. (2013). The roles of cis- and trans-regulation in the evolution of regulatory incompatibilities and sexually dimorphic gene expression. Genome Res. PubMed ID: 24043293

    Branco, A. T., Hartl, D. L., Lemos, B. (2013) Chromatin-Associated Proteins HP1 and Mod(mdg4) Modify Y-Linked Regulatory Variation in the Drosophila Testis. Genetics. PubMed ID: 23636736

    Sackton, T., Hartl, D. (2013) Meta-analysis reveals that genes regulated by the Y chromosome in Drosophila melanogaster are preferentially localized to repressive chromatin. Genome Biol Evol. PubMed ID: 23315381

    Corbett-Detig, R. B. and Hartl, D. L. (2012). Population Genomics of Inversion Polymorphisms in Drosophila melanogaster. PLoS Genet 8: e1003056. PubMed ID: 23284285

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

  • Paul Hartley Functional Genetics, Bournemouth University
    Sivakumar, S., Miellet, S., Clarke, C. and Hartley, P. S. (2022). Insect nephrocyte function is regulated by a store operated calcium entry mechanism controlling endocytosis and Amnionless turnover. J Insect Physiol 143: 104453. PubMed ID: 36341969

    Vaughan, L., Marley, R., Miellet, S. and Hartley, P. S. (2017). The impact of SPARC on age-related cardiac dysfunction and fibrosis in Drosophila. Exp Gerontol. PubMed ID: 29032244

    Hartley, P. S., Motamedchaboki, K., Bodmer, R. and Ocorr, K. (2016). SPARC-Dependent Cardiomyopathy in Drosophila. Circ Cardiovasc Genet 9(2): 119-129. PubMed ID: 26839388

    Ivy, J. R., Drechsler, M., Catterson, J. H., Bodmer, R., Ocorr, K., Paululat, A. and Hartley, P. S. (2015). Klf15 Is Critical for the Development and Differentiation of Drosophila Nephrocytes. PLoS One 10(8): e0134620. PubMed ID: 26301956

  • Kieran Harvey Peter MacCallum Cancer Centre, East Melbourne, Victoria Australia
    Pojer, J. M., Manning, S. A., Kroeger, B., Kondo, S. and Harvey, K. F. (2021). The Hippo pathway uses different machinery to control cell fate and organ size. iScience 24(8): 102830. PubMed ID: 34355153

    Vissers, J. H. A., Dent, L. G., House, C. M., Kondo, S. and Harvey, K. F. (2020). Pits and CtBP Control Tissue Growth in Drosophila melanogaster with the Hippo Pathway Transcription Repressor, Tgi. Genetics. PubMed ID: 32122936

    Dent, L. G., Manning, S. A., Kroeger, B., Williams, A. M., Saiful Hilmi, A. J., Crea, L., Kondo, S., Horne-Badovinac, S. and Harvey, K. F. (2019). The dPix-Git complex is essential to coordinate epithelial morphogenesis and regulate myosin during Drosophila egg chamber development. PLoS Genet 15(5): e1008083. PubMed ID: 31116733

    Poon, C. L. C., Liu, W., Song, Y., Gomez, M., Kulaberoglu, Y., Zhang, X., Xu, W., Veraksa, A., Hergovich, A., Ghabrial, A. and Harvey, K. F. (2018). A Hippo-like signaling pathway controls tracheal morphogenesis in Drosophila melanogaster. Dev Cell 47(5): 564-575.e565. PubMed ID: 30458981

    Vissers, J. H. A., Froldi, F., Schroder, J., Papenfuss, A. T., Cheng, L. Y. and Harvey, K. F. (2018). The Scalloped and Nerfin-1 transcription factors cooperate to maintain neuronal cell fate. Cell Rep 25(6): 1561-1576.e1567. PubMed ID: 30404010

    Manning, S. A., Dent, L. G., Kondo, S., Zhao, Z. W., Plachta, N. and Harvey, K. F. (2018). Dynamic fluctuations in subcellular localization of the Hippo pathway effector Yorkie in vivo. Curr Biol 28(10): 1651-1660.e1654. PubMed ID: 29754899

    Poon, C.L., Mitchell, K.A., Kondo, S., Cheng, L.Y. and Harvey, K.F. (2016). The Hippo pathway regulates neuroblasts and brain size in Drosophila melanogaster. Curr Biol [Epub ahead of print]. PubMed ID: 26996505

    Vissers, J.H., Manning, S.A., Kulkarni, A. and Harvey, K.F. (2016). A Drosophila RNAi library modulates Hippo pathway-dependent tissue growth. Nat Commun 7: 10368. PubMed ID: 26766446

    Irvine, K. D. and Harvey, K. F. (2015). Control of organ growth by patterning and Hippo signaling in Drosophila. Cold Spring Harb Perspect Biol 7. PubMed ID: 26032720

    Dent, L. G., Poon, C. L., Zhang, X., Degoutin, J. L., Tipping, M., Veraksa, A. and Harvey, K. F. (2014). The GTPase regulatory proteins Pix and Git control tissue growth via the Hippo pathway. Curr Biol [Epub ahead of print]. PubMed ID: 25484297

    Milton, C. C., Grusche, F. A., Degoutin, J. L., Yu, E., Dai, Q., Lai, E. C. and Harvey, K. F. (2014). The Hippo pathway regulates hematopoiesis in Drosophila melanogaster. Curr Biol 24: 2673-2680. PubMed ID: 25454587

  • Gaiti Hasan National Center for Biological Sciences, Tata Institute of Fundamental Research, Bangalore
    Kasturacharya, N., Dhall, J. K. and Hasan, G. (2023). A STIM dependent dopamine-neuropeptide axis maintains the larval drive to feed and grow in Drosophila. PLoS Genet 19(6): e1010435. PubMed ID: 37363909

    Mitra, R., Richhariya, S., Jayakumar, S., Notani, D. and Hasan, G. (2021). IP3-mediated Ca2+ signals regulate larval to pupal transition under nutrient stress through the H3K36 methyltransferase Set2. Development 148(11). PubMed ID: 34117888

    Sharma, A. and Hasan, G. (2020). Modulation of flight and feeding behaviours requires presynaptic IP(3)Rs in dopaminergic neurons. Elife 9. PubMed ID: 33155978

    Hasan, G. (2020). Surviving nutritional deprivation during development: neuronal intracellular calcium signaling is critical. Int J Dev Biol 64(1-2-3): 249-256. PubMed ID: 32659012

    Manjila, S. B., Kuruvilla, M., Ferveur, J. F., Sane, S. P. and Hasan, G. (2018). Extended flight bouts require disinhibition from GABAergic mushroom body neurons. Curr Biol. PubMed ID: 30612904

    Jayakumar, S., Richhariya, S., Deb, B. K. and Hasan, G. (2018). A multi-component neuronal response encodes the larval decision to pupariate upon amino acid starvation. J Neurosci. PubMed ID: 30301757

    Chakraborty, S. and Hasan, G. (2018). Store-operated Ca(2+) entry in Drosophila primary neuronal cultures. Methods Mol Biol 1843: 125-136. PubMed ID: 30203283

    Ravi, P., Trivedi, D. and Hasan, G. (2018). FMRFa receptor stimulated Ca2+ signals alter the activity of flight modulating central dopaminergic neurons in Drosophila melanogaster. PLoS Genet 14(8): e1007459. PubMed ID: 30110323

    Richhariya, S., Jayakumar, S., Kumar Sukumar, S. and Hasan, G. (2018). dSTIM- and Ral/Exocyst-Mediated Synaptic Release from Pupal Dopaminergic Neurons Sustains Drosophila Flight. eNeuro 5(3). PubMed ID: 29938216

    Jayakumar, S. and Hasan, G. (2018). Neuronal calcium signaling in metabolic regulation and adaptation to nutrient stress. Front Neural Circuits 12: 25. PubMed ID: 29674958

  • Bassem Hassan VIB, Gent, Belgium
    Dutta, S. B., Linneweber, G. A., Andriatsilavo, M., Hiesinger, P. R. and Hassan, B. A. (2023). EGFR-dependent suppression of synaptic autophagy is required for neuronal circuit development. Curr Biol. PubMed ID: 36640763

    Mohylyak, I., Bengochea, M., Pascual-Caro, C., Asfogo, N., Fonseca-Topp, S., Danda, N., Atak, Z. K., De Waegeneer, M., Placais, P. Y., Preat, T., Aerts, S., Corti, O., de Juan-Sanz, J. and Hassan, B. A. (2023). Developmental transcriptional control of mitochondrial homeostasis is required for activity-dependent synaptic connectivity. bioRxiv. PubMed ID: 37333418

    Liu, T., Zhang, T., Nicolas, M., Boussicault, L., Rice, H., Soldano, A., Claeys, A., Petrova, I., Fradkin, L., De Strooper, B., Potier, M. C. and Hassan, B. A. (2021). The amyloid precursor protein is a conserved Wnt receptor. Elife 10. PubMed ID: 34515635

    Kessissoglou, I. A., Langui, D., Hasan, A., Maral, M., Dutta, S. B., Hiesinger, P. R. and Hassan, B. A. (2020). The Drosophila amyloid precursor protein homologue mediates neuronal survival and neuroglial interactions. PLoS Biol 18(12): e3000703. PubMed ID: 33290404

    Linneweber, G. A., Andriatsilavo, M., Dutta, S. B., Bengochea, M., Hellbruegge, L., Liu, G., Ejsmont, R. K., Straw, A. D., Wernet, M., Hiesinger, P. R. and Hassan, B. A. (2020). A neurodevelopmental origin of behavioral individuality in the Drosophila visual system. Science 367(6482): 1112-1119. PubMed ID: 32139539

    Ramaekers, A., Claeys, A., Kapun, M., Mouchel-Vielh, E., Potier, D., Weinberger, S., Grillenzoni, N., Dardalhon-Cumenal, D., Yan, J., Wolf, R., Flatt, T., Buchner, E. and Hassan, B. A. (2019). Altering the temporal Regulation of one transcription factor drives evolutionary trade-Offs between head sensory organs. Dev Cell. PubMed ID: 31447264

    Liu, G., Nath, T., Linneweber, G. A., Claeys, A., Guo, Z., Li, J., Bengochea, M., De Backer, S., Weyn, B., Sneyders, M., Nicasy, H., Yu, P., Scheunders, P. and Hassan, B. A. (2018). A simple computer vision pipeline reveals the effects of isolation on social interaction dynamics in Drosophila. PLoS Comput Biol 14(8): e1006410. PubMed ID: 30161262

    Mora, N., Oliva, C., Fiers, M., Ejsmont, R., Soldano, A., Zhang, T. T., Yan, J., Claeys, A., De Geest, N. and Hassan, B. A. (2018). A temporal transcriptional switch governs stem cell division, neuronal numbers, and maintenance of differentiation. Dev Cell 45(1): 53-66.e55. PubMed ID: 29576424

    Koch, M., Nicolas, M., Zschaetzsch, M., de Geest, N., Claeys, A., Yan, J., Morgan, M. J., Erfurth, M. L., Holt, M., Schmucker, D. and Hassan, B. A. (2017). A Fat-Facets-Dscam1-JNK Pathway Enhances Axonal Growth in Development and after Injury. Front Cell Neurosci 11: 416. PubMed ID: 29472843

    Lou, W. P., Mateos, A., Koch, M., Klussman, S., Yang, C., Lu, N., Kumar, S., Limpert, S., Gopferich, M., Zschaetzsch, M., Sliwinski, C., Kenzelmann, M., Seedorf, M., Maillo, C., Senis, E., Grimm, D., Puttagunta, R., Mendez, R., Liu, K., Hassan, B. A. and Martin-Villalba, A. (2017). Regulation of adult CNS axonal regeneration by the post-transcriptional regulator Cpeb1. Front Mol Neurosci 10: 445. PubMed ID: 29379413

    Weinberger, S., Topping, M. P., Yan, J., Claeys, A., De Geest, N., Ozbay, D., Hassan, T., He, X., Albert, J. T., Hassan, B. A. and Ramaekers, A. (2017). Evolutionary changes in transcription factor coding sequence quantitatively alter sensory organ development and function. Elife 6. PubMed ID: 28406397

    Hassan, B. A. and Hiesinger, P. R. (2015). Beyond Molecular Codes: Simple Rules to Wire Complex Brains. Cell 163(2): 285-291. PubMed ID: 26451480

    Oliva, C., Soldano, A., Mora, N., De Geest, N., Claeys, A., Erfurth, M. L., Sierralta, J., Ramaekers, A., Dascenco, D., Ejsmont, R. K., Schmucker, D., Sanchez-Soriano, N. and Hassan, B. A. (2016). Regulation of Drosophila Brain Wiring by Neuropil Interactions via a Slit-Robo-RPTP Signaling Complex. Dev Cell 39(2): 267-278. PubMed ID: 27780041

  • Esteban Hasson Department of Ecology, Genetics and Evolution, University of Buenos Aires
    Fanara, J. J., Beti, M. I. L., Gandini, L. and Hasson, E. (2022). Oviposition behaviour in Drosophila melanogaster: Genetic and behavioural decoupling between oviposition acceptance and preference for natural fruits. J Evol Biol. PubMed ID: 36357966

    Hurtado, J., Almeida, F. C., Belliard, S. A., Revale, S. and Hasson, E. (2021). Research gaps and new insights in the evolution of Drosophila seminal fluid proteins. Insect Mol Biol. PubMed ID: 34747062

    Iglesias, P. P., Soto, E. M., Soto, I. M., Colines, B. and Hasson, E. (2018). The influence of developmental environment on courtship song in cactophilic Drosophila. J Evol Biol. PubMed ID: 29658159

    Iglesias, P. P. and Hasson, E. (2017). The role of courtship song in female mate choice in South American Cactophilic Drosophila. PLoS One 12(5): e0176119. PubMed ID: 28467464

    Mensch, J., Hurtado, J., Zermoglio, P. F., de la Vega, G., Rolandi, C., Schilman, P. E., Markow, T. A. and Hasson, E. (2017). Enhanced fertility and chill tolerance after cold-induced reproductive arrest in females of temperate species of the Drosophila buzzatii complex. J Exp Biol 220(Pt 4): 713-721. PubMed ID: 27956482

    De Panis, D. N., Padro, J., Furio-Tari, P., Tarazona, S., Milla Carmona, P. S., Soto, I. M., Dopazo, H., Conesa, A. and Hasson, E. (2016). Transcriptome modulation during host shift is driven by secondary metabolites in desert Drosophila. Mol Ecol 25(18): 4534-4550. PubMed ID: 27483442

    Carreira, V. P., Mensch, J., Hasson, E. and Fanara, J. J. (2016). Natural Genetic Variation and Candidate Genes for Morphological Traits in Drosophila melanogaster. PLoS One 11(7): e0160069. PubMed ID: 27459710

  • Victor Hatini Sackler School of Biomedical Science, Tufts University
    Malin, J., Rosa Birriel, C., Astigarraga, S., Treisman, J. E. and Hatini, V. (2022). Sidekick dynamically rebalances contractile and protrusive forces to control tissue morphogenesis. J Cell Biol 221(5). PubMed ID: 35258563

    Del Signore, S. J., Cilla, R. and Hatini, V. (2018). The WAVE regulatory complex and branched F-actin counterbalance contractile force to control cell shape and packing in the Drosophila eye. Dev Cell 44(4): 471-483.e474. PubMed ID: 29396116

    de Madrid, B.H., Greenberg, L. and Hatini, V. (2015). RhoGAP68F controls transport of adhesion proteins in Rab4 endosomes to modulate epithelial morphogenesis of Drosophila leg discs. Dev Biol [Epub ahead of print]. PubMed ID: 25617722

    Del Signore, S. J., Hayashi, T. and Hatini, V. (2012). odd-skipped genes and lines organize the notum anterior-posterior axis using autonomous and non-autonomous mechanisms. Mech Dev 129: 147-161. PubMed ID: 22613630

    Greenberg, L. and Hatini, V. (2011). Systematic expression and loss-of-function analysis defines spatially restricted requirements for Drosophila RhoGEFs and RhoGAPs in leg morphogenesis. Mech Dev 128: 5-17. PubMed ID: 20851182

    Greenberg, L. and Hatini, V. (2009). Essential roles for lines in mediating leg and antennal proximodistal patterning and generating a stable Notch signaling interface at segment borders. Dev Biol 330: 93-104. PubMed ID: 19324031

  • Daisuke Hattori University of Texas Southwestern, Dallas
    Liu, J. Y., Inoshita, T., Shiba-Fukushima, K., Yoshida, S., Ogata, K., Ishihama, Y., Imai, Y. and Hattori, N. (2022). Ubiquitination at the lysine 27 residue of the parkin ubiquitin-like domain is suggestive of a new mechanism of parkin activation. Hum Mol Genet. PubMed ID: 35313349

    Hung, Y. C., Huang, K. L., Chen, P. L., Li, J. L., Lu, S. H., Chang, J. C., Lin, H. Y., Lo, W. C., Huang, S. Y., Lee, T. T., Lin, T. Y., Imai, Y., Hattori, N., Liu, C. S., Tsai, S. Y., Chen, C. H., Lin, C. H. and Chan, C. C. (2021). UQCRC1 engages cytochrome c for neuronal apoptotic cell death. Cell Rep 36(12): 109729. PubMed ID: 34551295

    Yamaguchi, A., Ishikawa, K. I., Inoshita, T., Shiba-Fukushima, K., Saiki, S., Hatano, T., Mori, A., Oji, Y., Okuzumi, A., Li, Y., Funayama, M., Imai, Y., Hattori, N. and Akamatsu, W. (2020). Identifying Therapeutic Agents for Amelioration of Mitochondrial Clearance Disorder in Neurons of Familial Parkinson Disease. Stem Cell Reports 14(6): 1060-1075. PubMed ID: 32470327

    Yamaguchi, A., Ishikawa, K. I., Inoshita, T., Shiba-Fukushima, K., Saiki, S., Hatano, T., Mori, A., Oji, Y., Okuzumi, A., Li, Y., Funayama, M., Imai, Y., Hattori, N. and Akamatsu, W. (2020). Identifying Therapeutic Agents for Amelioration of Mitochondrial Clearance Disorder in Neurons of Familial Parkinson Disease. Stem Cell Reports 14(6): 1060-1075. PubMed ID: 32470327

    Shiba-Fukushima, K., Inoshita, T., Sano, O., Iwata, H., Ishikawa, K. I., Okano, H., Akamatsu, W., Imai, Y. and Hattori, N. (2020). A Cell-Based High-Throughput Screening Identified Two Compounds that Enhance PINK1-Parkin Signaling. iScience 23(5): 101048. PubMed ID: 32335362

    Imai, Y., Inoshita, T., Meng, H., Shiba-Fukushima, K., Hara, K. Y., Sawamura, N. and Hattori, N. (2019). Light-driven activation of mitochondrial proton-motive force improves motor behaviors in a Drosophila model of Parkinson's disease. Commun Biol 2: 424. PubMed ID: 31799427


  • Scott Hawley Stowers Institute for Medical Research, Kansas City
    Wesley, E. R., Hawley, R. S. and Billmyre, K. K. (2020). Genetic background impacts the timing of synaptonemal complex breakdown in Drosophila melanogaster. Chromosoma 129(3-4): 243-254. PubMed ID: 33068154
    Miller, D. E., Kahsai, L., Buddika, K., Dixon, M. J., Kim, B. Y., Calvi, B. R., Sokol, N. S., Hawley, R. S. and Cook, K. R. (2020). Identification and Characterization of Breakpoints and Mutations on Drosophila melanogaster Balancer Chromosomes. G3 (Bethesda). PubMed ID: 32972999

    Bonner, A. M., Hughes, S. E. and Hawley, R. S. (2020). Regulation of Polo Kinase by Matrimony Is Required for Cohesin Maintenance during Drosophila melanogaster Female Meiosis. Curr Biol. PubMed ID: 32008903

    Billmyre, K. K., Cahoon, C. K., Heenan, G. M., Wesley, E. R., Yu, Z., Unruh, J. R., Takeo, S. and Hawley, R. S. (2019). X chromosome and autosomal recombination are differentially sensitive to disruptions in SC maintenance. Proc Natl Acad Sci U S A. PubMed ID: 31570610

    Hughes, S. E., Hemenway, E., Guo, F., Yi, K., Yu, Z. and Hawley, R. S. (2019). The E3 ubiquitin ligase Sina regulates the assembly and disassembly of the synaptonemal complex in Drosophila females. PLoS Genet 15(5): e1008161. PubMed ID: 31107865

    Lake, C. M., Nielsen, R. J., Bonner, A. M., Eche, S., White-Brown, S., McKim, K. S. and Hawley, R. S. (2019). Narya, a RING finger domain-containing protein, is required for meiotic DNA double-strand break formation and crossover maturation in Drosophila melanogaster. PLoS Genet 15(1): e1007886. PubMed ID: 30615609

    Bonner, A. M. and Hawley, R. S. (2018). Functional consequences of the evolution of matrimony, a meiosis-specific inhibitor of Polo kinase. Mol Biol Evol. PubMed ID: 30351378

    Hanlon, S. L., Miller, D. E., Eche, S. and Hawley, R. S. (2018). Origin, composition, and structure of the supernumerary B chromosome of Drosophila melanogaster. Genetics. PubMed ID: 30249684

    Cahoon, C. K., Yu, Z., Wang, Y., Guo, F., Unruh, J. R., Slaughter, B. D. and Hawley, R. S. (2017). Superresolution expansion microscopy reveals the three-dimensional organization of the Drosophila synaptonemal complex. Proc Natl Acad Sci U S A 114(33): E6857-e6866. PubMed ID: 28760978

    Miller, D. E., Cook, K. R., Arvanitakis, A. V. and Hawley, R. S. (2016). Third chromosome balancer inversions disrupt protein-coding genes and influence distal recombination events in Drosophila melanogaster. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27172211

    Miller, D. E., Cook, K. R., Yeganeh Kazemi, N., Smith, C. B., Cockrell, A. J., Hawley, R. S. and Bergman, C. M. (2016). Rare recombination events generate sequence diversity among balancer chromosomes in Drosophila melanogaster. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26903656

    Gutzwiller, F., Carmo, C. R., Miller, D. E., Rice, D. W., Newton, I. L., Hawley, R. S., Teixeira, L. and Bergman, C. M. (2015). Dynamics of Wolbachia pipientis gene expression across the Drosophila melanogaster life cycle. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26497146

  • Bruce Hay Caltech
    Oberhofer, G., Ivy, T. and Hay, B. A. (2021). Gene drive that results in addiction to a temperature-sensitive version of an essential gene triggers population collapse in Drosophila. Proc Natl Acad Sci U S A 118(49). PubMed ID: 34845012

    Oberhofer, G., Ivy, T. and Hay, B. A. (2021). Split versions of Cleave and Rescue selfish genetic elements for measured self limiting gene drive. PLoS Genet 17(2): e1009385. PubMed ID: 33600432

    Oberhofer, G., Ivy, T. and Hay, B. A. (2020). Gene drive and resilience through renewal with next generation Cleave and Rescue selfish genetic elements. Proc Natl Acad Sci U S A 117(16): 9013-9021. PubMed ID: 32245808

    Oberhofer, G., Ivy, T. and Hay, B. A. (2019). Cleave and Rescue, a novel selfish genetic element and general strategy for gene drive. Proc Natl Acad Sci U S A. PubMed ID: 30760597

    Oberhofer, G., Ivy, T. and Hay, B. A. (2018). Behavior of homing endonuclease gene drives targeting genes required for viability or female fertility with multiplexed guide RNAs. Proc Natl Acad Sci U S A 115(40): E9343-e9352. PubMed ID: 30224454

    Akbari, O. S., Papathanos, P. A., Sandler, J. E., Kennedy, K. and Hay, B. A. (2014). Identification of germline transcriptional regulatory elements in Aedes aegypti. Sci Rep 4: 3954. PubMed ID: 24492376

    Akbari, O. S., Matzen, K. D., Marshall, J. M., Huang, H., Ward, C. M. and Hay, B. A. (2013). A synthetic gene drive system for local, reversible modification and suppression of insect populations. Curr Biol 23: 671-677. PubMed ID: 23541732

    Akbari, O. S., Chen, C. H., Marshall, J. M., Huang, H., Antoshechkin, I. and Hay, B. A. (2012). Novel Synthetic Medea Selfish Genetic Elements Drive Population Replacement in Drosophila; a Theoretical Exploration of Medea-Dependent Population Suppression. ACS Synth Biol. PubMed ID: 23654248

    Marshall, J. M. and Hay, B. A. (2011). Inverse Medea as a novel gene drive system for local population replacement: a theoretical analysis. J Hered 102: 336-341. PubMed ID: 21493596

    Hay, B. A., Chen, C. H., Ward, C. M., Huang, H., Su, J. T. and Guo, M. (2010). Engineering the genomes of wild insect populations: challenges, and opportunities provided by synthetic Medea selfish genetic elements. J Insect Physiol 56: 1402-1413. PubMed ID: 20570677

  • Shigeo Hayashi Riken Center for Developmental Biology, Kobe
    Chu, W. C. and Hayashi, S. (2021). Mechano-chemical enforcement of tendon apical ECM into nano-filaments during Drosophila flight muscle development. Curr Biol. PubMed ID: 33545042

    Kondo, T. and Hayashi, S. (2019). Two-step regulation of trachealess ensures tight coupling of cell fate with morphogenesis in the Drosophila trachea. Elife 8. PubMed ID: 31439126

    Ogura, Y., Sami, M. M., Wada, H. and Hayashi, S. (2019). Automated FRET quantification reveals distinct subcellular ERK activation kinetics in response to graded EGFR signaling in Drosophila. Genes Cells. PubMed ID: 30851218

    Itakura, Y., Inagaki, S., Wada, H. and Hayashi, S. (2018). Trynity controls epidermal barrier function and respiratory tube maturation in Drosophila by modulating apical extracellular matrix nano-patterning. PLoS One 13(12): e0209058. PubMed ID: 30576352

    Hayashi, S. and Kondo, T. (2018). Development and Function of the Drosophila Tracheal System. Genetics 209(2): 367-380. PubMed ID: 29844090

    Miao, G. and Hayashi, S. (2016). Escargot controls the sequential specification of two tracheal tip cell types by suppressing FGF signaling in Drosophila. Development [Epub ahead of print]. PubMed ID: 27742749

    Otani, T., Ogura, Y., Misaki, K., Maeda, T., Kimpara, A., Yonemura, S. and Hayashi, S. (2016). IKK inhibits PKC to promote Fascin-dependent actin bundling. Development. PubMed ID: 27578797

    Kato, K., Dong, B., Wada, H., Tanaka-Matakatsu, M., Yagi, Y. and Hayashi, S. (2016). Microtubule-dependent balanced cell contraction and luminal-matrix modification accelerate epithelial tube fusion. Nat Commun 7: 11141. PubMed ID: 27067650

    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

    Hannezo, E., Dong, B., Recho, P., Joanny, J. F. and Hayashi, S. (2015). Cortical instability drives periodic supracellular actin pattern formation in epithelial tubes. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26077909

    Miao, G. and Hayashi, S. (2014). Manipulation of gene expression by infrared laser heat shock and its application to the study of tracheal development in Drosophila. Dev Dyn. PubMed ID: 25258210

  • Thomas Hays Department of Genetics, Cell Biology and Development, University of Minnisota
    Tubman, E., He, Y., Hays, T. S. and Odde, D. J. (2018). Kinesin-5 mediated chromosome congression in insect spindles. Cell Mol Bioeng 11(1): 25-36. PubMed ID: 29552234

    Mauvezin, C., Neisch, A. L., Ayala, C. I., Kim, J., Beltrame, A., Braden, C. R., Gardner, M. K., Hays, T. S. and Neufeld, T. P. (2016). Coordination of autophagosome-lysosome fusion and transport by a Klp98A-Rab14 complex. J Cell Sci [Epub ahead of print]. PubMed ID: 26763909

    Reis, G. F., Yang, G., Szpankowski, L., Weaver, C., Shah, S. B., Robinson, J. T., Hays, T. S., Danuser, G. and Goldstein, L. S. (2012). Molecular motor function in axonal transport in vivo probed by genetic and computational analysis in Drosophila. Mol Biol Cell 23: 1700-1714. PubMed ID: 22398725

    Lorenzo, D. N., Li, M. G., Mische, S. E., Armbrust, K. R., Ranum, L. P. and Hays, T. S. (2010). Spectrin mutations that cause spinocerebellar ataxia type 5 impair axonal transport and induce neurodegeneration in Drosophila. J Cell Biol 189: 143-158. PubMed ID: 20368622

  • Tulle Hazelrigg Biological Sciences, Columbia University
    Clough, E., Tedeschi, T. and Hazelrigg, T. (2014). Epigenetic regulation of oogenesis and germ stem cell maintenance by the Drosophila histone methyltransferase Eggless/dSetDB1. Dev Biol. PubMed ID: 24485852

    Hines, K. A., Cryderman, D. E., Flannery, K. M., Yang, H., Vitalini, M. W., Hazelrigg, T., Mizzen, C. A. and Wallrath, L. L. (2009). Domains of heterochromatin protein 1 required for Drosophila melanogaster heterochromatin spreading. Genetics 182: 967-977. PubMed ID: 19487560

  • Qi He Biology Department, Brooklyn College
    Roblodowski, C. and He, Q. (2017). Drosophila Dunc-115 mediates axon projection through actin binding. Invert Neurosci 17(1): 2. PubMed ID: 28124181

    Koenecke, N., Johnston, J., He, Q., Meier, S. and Zeitlinger, J. (2017). Drosophila poised enhancers are generated during tissue patterning with the help of repression. Genome Res 27(1): 64-74. PubMed ID: 27979994

    He, Q. and Roblodowski, C. (2016). Functional Analysis of Actin-Binding Proteins in the Central Nervous System of Drosophila. Methods Mol Biol 1365: 349-355. PubMed ID: 26498796

  • Urlike Heberlein Janelia Farm, Ashburn, VA
    Simon, J. C. and Heberlein, U. (2020). Social hierarchy is established and maintained with distinct acts of aggression in male Drosophila melanogaster. J Exp Biol 223(Pt 24). PubMed ID: 33268534

    Shao, L., Chung, P., Wong, A., Siwanowicz, I., Kent, C. F., Long, X. and Heberlein, U. (2019). A neural circuit encoding the experience of copulation in female Drosophila. Neuron. PubMed ID: 31072787

    Kim, Y. K., Saver, M., Simon, J., Kent, C. F., Shao, L., Eddison, M., Agrawal, P., Texada, M., Truman, J. W. and Heberlein, U. (2018). Repetitive aggressive encounters generate a long-lasting internal state in Drosophila melanogaster males. Proc Natl Acad Sci U S A 115(5): 1099-1104. PubMed ID: 29339481

    Shao, L., Saver, M., Chung, P., Ren, Q., Lee, T., Kent, C. F. and Heberlein, U. (2017). Dissection of the Drosophila neuropeptide F circuit using a high-throughput two-choice assay. Proc Natl Acad Sci U S A 114(38): E8091-e8099. PubMed ID: 28874527

    Albin, S.D., Kaun, K.R., Knapp, J.M., Chung, P., Heberlein, U. and Simpson, J.H. (2015). A subset of serotonergic neurons evokes hunger in adult Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26344091

    King, I. F., Eddison, M., Kaun, K. R. and Heberlein, U. (2014). EGFR and FGFR Pathways Have Distinct Roles in Drosophila Mushroom Body Development and Ethanol-Induced Behavior. PLoS One 9: e87714. PubMed ID: 24498174

    Azanchi, R., Kaun, K. R. and Heberlein, U. (2013). Competing dopamine neurons drive oviposition choice for ethanol in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 24324162

    Devineni, A. V., Eddison, M. and Heberlein, U. (2013). The Novel Gene tank, a Tumor Suppressor Homolog, Regulates Ethanol Sensitivity in Drosophila. J Neurosci 33: 8134-8143. PubMed ID: 23658154

    Devineni, A. V. and Heberlein, U. (2013). The Evolution of Drosophila melanogaster as a Model for Alcohol Addiction. Annu Rev Neurosci. PubMed ID: 23642133

    McClure, K. D., Heberlein, U. (2013) A Small Group of Neurosecretory Cells Expressing the Transcriptional Regulator apontic and the Neuropeptide corazonin Mediate Ethanol Sedation in Drosophila. J Neurosci 33: 4044-4054. PubMed ID: 23447613

  • Margarete Heck University/BHF, Centre for Cardiovascular Science, University of Edinburgh
    Chang, C. W., Abhinav, K., Di Cara, F., Panagakou, I., Vass, S. and Heck, M. M. (2016). A role for the metalloprotease invadolysin in insulin signaling and adipogenesis. Biol Chem. PubMed ID: 27622830

    Rao, S. G., Janiszewski, M. M., Duca, E., Nelson, B., Abhinav, K., Panagakou, I., Vass, S. and Heck, M. M. (2015). Invadolysin acts genetically via the SAGA complex to modulate chromosome structure. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 25779050

    Di Cara, F., Duca, E., Dunbar, D. R., Cagney, G. and Heck, M. M. (2013). Invadolysin, a conserved lipid droplet-associated metalloprotease, is required for mitochondrial function in Drosophila. J Cell Sci. PubMed ID: 23943867

    Catterson, J. H., Heck, M. M. and y, P. S. (2013). Fermitins, the orthologs of mammalian Kindlins, regulate the development of a functional cardiac syncytium in Drosophila melanogaster. PLoS One 8: e62958. PubMed ID: 23690969

    Bolukbasi, E., Vass, S., Cobbe, N., Nelson, B., Simossis, V., Dunbar, D. R. and Heck, M. M. (2012). Drosophila poly suggests a novel role for the Elongator complex in insulin receptor-target of rapamycin signalling. Open Biol 2: 110031. PubMed ID: 22645656

    Howell, L., Sampson, C. J., Xavier, M. J., Bolukbasi, E., Heck, M. M. and Williams, M. J. (2012). A directed miniscreen for genes involved in the Drosophila anti-parasitoid immune response. Immunogenetics 64: 155-161. PubMed ID: 21947570

  • Elizabeth Heckscher Cell and Molecular Biology Program, University of Chicago
    Greaney, M. R., Wreden, C. C. and Heckscher, E. S. (2023). Distinctive features of the central synaptic organization of Drosophila larval proprioceptors. Front Neural Circuits 17: 1223334. PubMed ID: 37564629

    Wang, Y. W., Wreden, C. C., Levy, M., Meng, J. L., Marshall, Z. D., MacLean, J. and Heckscher, E. (2022). Sequential addition of neuronal stem cell temporal cohorts generates a feed-forward circuit in the Drosophila larval nerve cord. Elife 11. PubMed ID: 35723253

    Marshall, Z. D. and Heckscher, E. S. (2022). The Role of Even-Skipped in Drosophila Larval Somatosensory Circuit Assembly. eNeuro 9(1). PubMed ID: 35031555

    Chi, W., Liu, W., Fu, W., Xia, S., Heckscher, E. S. and Zhuang, X. (2021). RNA-binding protein syncrip regulates starvation-induced hyperactivity in adult Drosophila. PLoS Genet 17(2): e1009396. PubMed ID: 33617535

    He, L., Gulyanon, S., Mihovilovic Skanata, M., Karagyozov, D., Heckscher, E. S., Krieg, M., Tsechpenakis, G., Gershow, M. and Tracey, W. D., Jr. (2019). Direction Selectivity in Drosophila Proprioceptors Requires the Mechanosensory Channel Tmc. Curr Biol 29(6): 945-956 e943. PubMed ID: 30853433

  • Yael Heifetz Department of Entomology, Hebrew University, Rehovot
    Sanchez-Lopez, J. A., Twena, S., Apel, I., Kornhaeuser, S. C., Chasnitsky, M., Miklosi, A. G., Vega-Dominguez, P. J., Shephard, A., Hefetz, A. and Heifetz, Y. (2022). Male-female communication enhances release of extracellular vesicles leading to high fertility in Drosophila. Commun Biol 5(1): 815. PubMed ID: 35963938

    Elgart, M., Stern, S., Salton, O., Gnainsky, Y., Heifetz, Y. and Soen, Y. (2016). Impact of gut microbiota on the fly's germ line. Nat Commun 7: 11280. PubMed ID: 27080728

    Heifetz, Y., Lindner, M., Garini, Y. and Wolfner, M. F. (2014). Mating regulates neuromodulator ensembles at nerve termini innervating the Drosophila reproductive tract. Curr Biol 24: 731-737. PubMed ID: 24631240

    Heifetz, Y. and Rivlin, P. K. (2010). Beyond the mouse model: using Drosophila as a model for sperm interaction with the female reproductive tract. Theriogenology 73: 723-739. PubMed ID: 20015541

  • Tapio Heino Department of Biosciences, University of Helsinki
    Walkowicz, L., Kijak, E., Krzeptowski, W., Gorska-Andrzejak, J., Stratoulias, V., Woznicka, O., Chwastek, E., Heino, T. I. and Pyza, E. M. (2017). Downregulation of DmMANF in glial cells results in neurodegeneration and affects sleep and lifespan in Drosophila melanogaster. Front Neurosci 11: 610. PubMed ID: 29163014

    Lindstrom, R., Lindholm, P., Palgi, M., Saarma, M. and Heino, T. I. (2017). In vivo screening reveals interactions between Drosophila Manf and genes involved in the mitochondria and the ubiquinone synthesis pathway. BMC Genet 18(1): 52. PubMed ID: 28578657

    Stratoulias, V. and Heino, T. I. (2015). Analysis of the conserved neurotrophic factor MANF in the Drosophila adult brain. Gene Expr Patterns. PubMed ID: 25917377

    Stratoulias, V. and Heino, T. I. (2014). MANF silencing, immunity induction or autophagy trigger an unusual cell type in metamorphosing Drosophila brain. Cell Mol Life Sci [Epub ahead of print]. PubMed ID: 25511196

    Lindstrom, R., Lindholm, P., Kallijarvi, J., Yu, L. Y., Piepponen, T. P., Arumae, U., Saarma, M. and Heino, T. I. (2013). Characterization of the Structural and Functional Determinants of MANF/CDNF in Drosophila In Vivo Model. PLoS One 8: e73928. PubMed ID: 24019940

    Kallijarvi, J., Stratoulias, V., Virtanen, K., Hietakangas, V., Heino, T. I. and Saarma, M. (2012). Characterization of Drosophila GDNF receptor-like and evidence for its evolutionarily conserved interaction with neural cell adhesion molecule (NCAM)/FasII. PLoS One 7: e51997. PubMed ID: 23284846

    Pessa, H. K., Greco, D., Kvist, J., Wahlstrom, G., Heino, T. I., Auvinen, P. and Frilander, M. J. (2010). Gene expression profiling of U12-type spliceosome mutant Drosophila reveals widespread changes in metabolic pathways. PLoS One 5: e13215. PubMed ID: 20949011

  • Stephen Helfand Department of Molecular Biology, Brown University, Providence
    Taylor, J. R., Wood, J. G., Mizerak, E., Hinthorn, S., Liu, J., Finn, M., Gordon, S., Zingas, L., Chang, C., Klein, M. A., Denu, J. M., Gorbunova, V., Seluanov, A., Boeke, J. D., Sedivy, J. M. and Helfand, S. L. (2022). Sirt6 regulates lifespan in Drosophila melanogaster. Proc Natl Acad Sci U S A 119(5). PubMed ID: 35091469

    Wood, J. G., Schwer, B., Wickremesinghe, P. C., Hartnett, D. A., Burhenn, L., Garcia, M., Li, M., Verdin, E. and Helfand, S. L. (2018). Sirt4 is a mitochondrial regulator of metabolism and lifespan in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 29378963

    Jones, B. C., Wood, J. G., Chang, C., Tam, A. D., Franklin, M. J., Siegel, E. R. and Helfand, S. L. (2016). A somatic piRNA pathway in the Drosophila fat body ensures metabolic homeostasis and normal lifespan. Nat Commun 7: 13856. PubMed ID: 28000665

    Wood, J. G., Jones, B. C., Jiang, N., Chang, C., Hosier, S., Wickremesinghe, P., Garcia, M., Hartnett, D. A., Burhenn, L., Neretti, N. and Helfand, S. L. (2016). Chromatin-modifying genetic interventions suppress age-associated transposable element activation and extend life span in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 27621458

    Whitaker, R., Gil, M., Ding, F., Tatar, M., Helfand, S. L. and Neretti, N. (2014). Dietary switch reveals fast coordinated gene expression changes in Drosophila melanogaster. Aging (Albany NY). PubMed ID: 24864304

    Ding, F., Pilar Gil, M., Franklin, M., Ferreira, J., Tatar, M., Helfand, S. L. and Neretti, N. (2014). Transcriptional response to dietary restriction in Drosophila melanogaster. J Insect Physiol. PubMed ID: 24819200

    Wood, J. G. and Helfand, S. L. (2013). Chromatin structure and transposable elements in organismal aging. Front Genet 4: 274. PubMed ID: 24363663

    Jiang, N., Du, G., Tobias, E., Wood, J. G., Whitaker, R., Neretti, N. and Helfand, S. L. (2013). Dietary and genetic effects on age-related loss of gene silencing reveal epigenetic plasticity of chromatin repression during aging. Aging (Albany NY). PubMed ID: 24243774

    Savva, Y. A., Jepson, J. E., Chang, Y. J., Whitaker, R., Jones, B. C., St Laurent, G., Tackett, M. R., Kapranov, P., Jiang, N., Du, G., Helfand, S. L. and Reenan, R. A. (2013). RNA editing regulates transposon-mediated heterochromatic gene silencing. Nat Commun 4: 2745. PubMed ID: 24201902

    Wood, J. G., Whitaker, R. and Helfand, S. L. (2013). Genetic and Biochemical Tools for Investigating Sirtuin Function in Drosophila melanogaster. Methods Mol Biol 1077: 57-67. PubMed ID: 24014399

    Rogina, B. and Helfand, S. L. (2013). Indy mutations and Drosophila longevity. Front Genet 4: 47. PubMed ID: 23580130

  • Charlotte Helfrich-Förster University of Wuerzburg, Würzburg
    Deppisch, P., Kirsch, V., Helfrich-Forster, C. and Senthilan, P. R. (2023). Contribution of cryptochromes and photolyases for insect life under sunlight. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 36609567

    Manoli, G., Zandawala, M., Yoshii, T. and Helfrich-Forster, C. (2023). Characterization of clock-related proteins and neuropeptides in Drosophila littoralis and their putative role in diapause. J Comp Neurol 531(15): 1525-1549. PubMed ID: 37493077

    Amatobi, K. M., Ozbek-Unal, A. G., Schabler, S., Deppisch, P., Helfrich-Forster, C., Mueller, M. J., Wegener, C. and Fekete, A. (2023). The circadian clock is required for rhythmic lipid transport in Drosophila in interaction with diet and photic condition. J Lipid Res: 100417. PubMed ID: 37481037

    Zurl, M., Poehn, B., Rieger, D., Krishnan, S., Rokvic, D., Veedin Rajan, V. B., Gerrard, E., Schlichting, M., Orel, L., Coric, A., Lucas, R. J., Wolf, E., Helfrich-Forster, C., Raible, F. and Tessmar-Raible, K. (2022). Two light sensors decode moonlight versus sunlight to adjust a plastic circadian/circalunidian clock to moon phase. Proc Natl Acad Sci U S A 119(22): e2115725119. PubMed ID: 35622889

    Reinhard, N., Schubert, F. K., Bertolini, E., Hagedorn, N., Manoli, G., Sekiguchi, M., Yoshii, T., Rieger, D. and Helfrich-Forster, C. (2022). The Neuronal Circuit of the Dorsal Circadian Clock Neurons in Drosophila melanogaster. Front Physiol 13: 886432. PubMed ID: 35574472

    Deppisch, P., Helfrich-Forster, C. and Senthilan, P. R. (2022). The Gain and Loss of Cryptochrome/Photolyase Family Members during Evolution. Genes (Basel) 13(9). PubMed ID: 36140781

    Reinhard, N., Bertolini, E., Saito, A., Sekiguchi, M., Yoshii, T., Rieger, D. and Helfrich-Forster, C. (2022). The lateral posterior clock neurons of Drosophila melanogaster express three neuropeptides and have multiple connections within the circadian clock network and beyond. J Comp Neurol 530(9): 1507-1529. PubMed ID: 34961936

    Deppisch, P., Prutscher, J. M., Pegoraro, M., Tauber, E., Wegener, C. and Helfrich-Forster, C. (2022). Adaptation of Drosophila melanogaster to Long Photoperiods of High-Latitude Summers Is Facilitated by the ls-Timeless Allele. J Biol Rhythms 37(2): 185-201. PubMed ID: 35301885

    Reinhard, N., Bertolini, E., Saito, A., Sekiguchi, M., Yoshii, T., Rieger, D. and Helfrich-Forster, C. (2021). The lateral posterior clock neurons of Drosophila melanogaster express three neuropeptides and have multiple connections within the circadian clock network and beyond. J Comp Neurol. PubMed ID: 34961936

    Pauls, D., Selcho, M., Räderscheidt, J., Amatobi, K. M., Fekete, A., Krischke, M., Hermann-Luibl, C., Ozbek-Unal, A. G., Ehmann, N., Itskov, P. M., Kittel, R. J., Helfrich-Forster, C., Kuhnlein, R. P., Mueller, M. J. and Wegener, C. (2021). Endocrine signals fine-tune daily activity patterns in Drosophila. Curr Biol. PubMed ID: 34329588

  • Steve Henikoff Fred Hutchinson Cancer Research Center, Seattle
    Wooten, M., Takushi, B., Ahmad, K. and Henikoff, S. (2023). Aclarubicin stimulates RNA polymerase II elongation at closely spaced divergent promoters. Sci Adv 9(24): eadg3257. PubMed ID: 37315134

    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

    Ahmad, K. and Henikoff, S. (2021). The H3.3K27M oncohistone antagonizes reprogramming in Drosophila. PLoS Genet 17(7): e1009225. PubMed ID: 34280185

    Sarthy, J. F., Meers, M. P., Janssens, D. H., Henikoff, J. G., Feldman, H., Paddison, P. J., Lockwood, C. M., Vitanza, N. A., Olson, J. M., Ahmad, K. and Henikoff, S. (2020). Histone deposition pathways determine the chromatin landscapes of H3.1 and H3.3 K27M oncohistones. Elife 9. PubMed ID: 32902381

    Chereji, R. V., Bryson, T. D. and Henikoff, S. (2019). Quantitative MNase-seq accurately maps nucleosome occupancy levels. Genome Biol 20(1): 198. PubMed ID: 31519205

    Ramachandran, S., Ahmad, K. and Henikoff, S. (2017). Transcription and remodeling produce asymmetrically unwrapped nucleosomal intermediates. Mol Cell 68(6): 1038-1053.e1034. PubMed ID: 29225036

    Talbert, P., Kasinathan, S. and Henikoff, S. (2018). Simple and complex centromeric satellites in Drosophila sibling species. Genetics [Epub ahead of print]. PubMed ID: 29305387

    Ramachandran, S. and Henikoff, S. (2016). Transcriptional regulators compete with nucleosomes post-replication. Cell 165: 580-592. PubMed ID: 27062929

    Weber, C. M., Ramachandran, S. and Henikoff, S. (2014). Nucleosomes are context-specific, H2A.Z-modulated barriers to RNA polymerase. Mol Cell 53: 819-830. PubMed ID: 24606920

    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


  • Matthias Hentze European Molecular Biology Laboratory, Heidelberg
    Moretti, F., Kaiser, C., Zdanowicz-Specht, A. and Hentze, M. W. (2012). PABP and the poly(A) tail augment microRNA repression by facilitated miRISC binding. Nat Struct Mol Biol 19: 603-608. PubMed ID: 22635249

    Medenbach, J., Seiler, M. and Hentze, M. W. (2011). Translational control via protein-regulated upstream open reading frames. Cell 145: 902-913. PubMed ID: 21663794

    Moretti, F., Thermann, R. and Hentze, M. W. (2010). Mechanism of translational regulation by miR-2 from sites in the 5' untranslated region or the open reading frame. RNA 16: 2493-2502. PubMed ID: 20966199

    Duncan, K. E., Strein, C. and Hentze, M. W. (2009). The SXL-UNR corepressor complex uses a PABP-mediated mechanism to inhibit ribosome recruitment to msl-2 mRNA. Mol Cell 36: 571-582. PubMed ID: 19941818

  • Tory Herman Institute of Molecular Biology, University of Oregon
    Spinner, M. A., Pinter, K., Drerup, C. M. and Herman, T. G. (2020). A Conserved Role for Vezatin Proteins in Cargo-Specific Regulation of Retrograde Axonal Transport. Genetics. PubMed ID: 32788307

    Spinner, M. A., Walla, D. A. and Herman, T. G. (2018). Drosophila Syd-1 has RhoGAP activity that is required for presynaptic clustering of Bruchpilot/ELKS but not Neurexin-1. Genetics 208(2): 705-716. PubMed ID: 29217522

    Finley, J. K., Miller, A. C. and Herman, T. G. (2015). Polycomb group genes are required to maintain a binary fate choice in the Drosophila eye. Neural Dev 10: 2. PubMed ID: 25636358

    Kniss, J. S., Holbrook, S., Herman, T. G. (2013) R7 Photoreceptor Axon Growth Is Temporally Controlled by the Transcription Factor Ttk69, Which Inhibits Growth in Part by Promoting Transforming Growth Factor-beta/Activin Signaling. J Neurosci 33: 1509-1520. PubMed ID: 23345225

    Holbrook, S., Finley, J. K., Lyons, E. L. and Herman, T. G. (2012). Loss of syd-1 from R7 Neurons Disrupts Two Distinct Phases of Presynaptic Development. J Neurosci 32: 18101-18111. PubMed ID: 23238725

    Miller, A. C., Lyons, E. L. and Herman, T. G. (2009). cis-Inhibition of Notch by endogenous Delta biases the outcome of lateral inhibition. Curr Biol 19: 1378-1383. PubMed ID: 19631544

  • Hector Herranz Center for Stem Cell and Developmental Biology, University of Copenhagen
    Gerlach, S. U., Sander, M., Song, S. and Herranz, H. (2019). The miRNA bantam regulates growth and tumorigenesis by repressing the cell cycle regulator tribbles. Life Sci Alliance 2(4). PubMed ID: 31331981

    Song, S., Andrejeva, D., Freitas, F. C. P., Cohen, S. M. and Herranz, H. (2019). dTcf/Pangolin suppresses growth and tumor formation in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 31235567

    Sander, M., Eichenlaub, T. and Herranz, H. (2018). Oncogenic cooperation between Yorkie and the conserved microRNA miR-8 in the wing disc of Drosophila. Development. PubMed ID: 29945869

    Gerlach, S. U., Eichenlaub, T. and Herranz, H. (2018). Yorkie and JNK Control Tumorigenesis in Drosophila Cells with Cytokinesis Failure. Cell Rep 23(5): 1491-1503. PubMed ID: 29719260

    Herranz, H. and Cohen, S. M. (2017). Drosophila as a Model to Study the Link between Metabolism and Cancer. J Dev Biol 5(4). PubMed ID: 29615570

    Song, S., Herranz, H. and Cohen, S. M. (2017). The chromatin remodeling BAP complex limits tumor-promoting activity of the Hippo pathway effector Yki to prevent neoplastic transformation in Drosophila epithelia. Dis Model Mech 10(10): 1201-1209. PubMed ID: 28754838

  • Patrick Heun Wellcome Centre for Cell Biology, University of Edinburgh
    Anselm, E., Thomae, A. W., Jeyaprakash, A. A. and Heun, P. (2018). Oligomerization of Drosophila Nucleoplasmin-Like Protein is required for its centromere localization. Nucleic Acids Res. PubMed ID: 30357352

    Barth, T. K., Schade, G. O., Schmidt, A., Vetter, I., Wirth, M., Heun, P., Imhof, A. and Thomae, A. W. (2015). Identification of Drosophila centromere associated proteins by quantitative affinity purification-mass spectrometry. Data Brief 4: 544-550. PubMed ID: 26306323

    Barth, T. K., Schade, G. O., Schmidt, A., Vetter, I., Wirth, M., Heun, P., Thomae, A. W. and Imhof, A. (2014). Identification of novel Drosophila centromere-associated proteins. Proteomics 14(19): 2167-2178. PubMed ID: 24841622

  • Randall Hewes Department of Cell Biology, University of Oklahoma, Norman
    Gu, T., Zhao, T., Kohli, U. and Hewes, R. S. (2017). The large and small SPEN family proteins stimulate axon outgrowth during neurosecretory cell remodeling in Drosophila. Dev Biol. PubMed ID: 28916169

    Chen, D., Qu, C. and Hewes, R. S. (2014). Neuronal Remodeling During Metamorphosis Is Regulated by the alan shepard (shep) gene in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 24931409

    Gu, T., Zhao, T. and Hewes, R. S. (2013). Insulin signaling regulates neurite growth during metamorphic neuronal remodeling. Biol Open. PubMed ID: 24357229

    Gauthier, S. A., VanHaaften, E., Cherbas, L., Cherbas, P. and Hewes, R. S. (2012). Cryptocephal, the Drosophila melanogaster ATF4, is a specific coactivator for ecdysone receptor isoform B2. PLoS Genet 8: e1002883. PubMed ID: 22912598

    Zhao, T., Gu, T., Rice, H. C., McAdams, K. L., Roark, K. M., Lawson, K., Gauthier, S. A., Reagan, K. L. and Hewes, R. S. (2008). A Drosophila gain-of-function screen for candidate genes involved in steroid-dependent neuroendocrine cell remodeling. Genetics 178: 883-901. PubMed ID: 18245346

  • Alicia Hidalgo School of Biosciences, University of Birmingham
    Harrison, N. J., Connolly, E., Gascon Gubieda, A., Yang, Z., Altenhein, B., Losada Perez, M., Moreira, M., Sun, J. and Hidalgo, A. (2021). Regenerative neurogenic response from glia requires insulin-driven neuron-glia communication. Elife 10. PubMed ID: 33527895

    Li, G., Forero, M. G., Wentzell, J. S., Durmus, I., Wolf, R., Anthoney, N. C., Parker, M., Jiang, R., Hasenauer, J., Strausfeld, N. J., Heisenberg, M. and Hidalgo, A. (2020). A Toll-receptor map underlies structural brain plasticity. Elife 9. PubMed ID: 32066523

    Kato, K., Losada-Perez, M. and Hidalgo, A. (2017). The gene network underlying the glial regenerative response to central nervous system injury. Dev Dyn. PubMed ID: 28791751

    Foldi, I., Anthoney, N., Harrison, N., Gangloff, M., Verstak, B., Nallasivan, M. P., AlAhmed, S., Zhu, B., Phizacklea, M., Losada-Perez, M., Moreira, M., Gay, N. J. and Hidalgo, A. (2017). Three-tier regulation of cell number plasticity by neurotrophins and Tolls in Drosophila. J Cell Biol. PubMed ID: 28373203

    Losada-Perez, M., Harrison, N. and Hidalgo, A. (2016). Molecular mechanism of central nervous system repair by the Drosophila NG2 homologue kon-tiki. J Cell Biol [Epub ahead of print]. PubMed ID: 27551055

    Sutcliffe, B., Forero, M. G., Zhu, B., Robinson, I. M. and Hidalgo, A. (2013). Neuron-Type Specific Functions of DNT1, DNT2 and Spz at the Drosophila Neuromuscular Junction. PLoS One 8: e75902. PubMed ID: 24124519

    McIlroy, G., Foldi, I., Aurikko, J., Wentzell, J. S., Lim, M. A., Fenton, J. C., Gay, N. J. and Hidalgo, A. (2013). Toll-6 and Toll-7 function as neurotrophin receptors in the Drosophila melanogaster CNS. Nat Neurosci. PubMed ID: 23892553

    Kato, K., Hidalgo, A. (2013) An injury paradigm to investigate central nervous system repair in Drosophila. J Vis Exp. PubMed ID: 23567253

    Sweeney, S. T., Hidalgo, A., de Belle, J. S. and Keshishian, H. (2012). X-gal staining of the central nervous system in adult Drosophila. Cold Spring Harb Protoc 2012: 239-241. PubMed ID: 22301646

    Kato, K., Forero, M. G., Fenton, J. C. and Hidalgo, A. (2011). The glial regenerative response to central nervous system injury is enabled by pros-notch and pros-NFkappaB feedback. PLoS Biol 9: e1001133. PubMed ID: 21912512

  • Peter Robin Hiesinger Freie Universitat Berlin
    Dutta, S. B., Linneweber, G. A., Andriatsilavo, M., Hiesinger, P. R. and Hassan, B. A. (2023). EGFR-dependent suppression of synaptic autophagy is required for neuronal circuit development. Curr Biol. PubMed ID: 36640763

    Kiral, F. R., Dutta, S. B., Linneweber, G. A., Hilgert, S., Poppa, C., Duch, C., von Kleist, M., Hassan, B. A. and Hiesinger, P. R. (2021). Brain connectivity inversely scales with developmental temperature in Drosophila. Cell Rep 37(12): 110145. PubMed ID: 34936868

    Kohrs, F. E., Daumann, I. M., Pavlovic, B., Jin, E. J., Kiral, F. R., Lin, S. C., Port, F., Wolfenberg, H., Mathejczyk, T. F., Linneweber, G. A., Chan, C. C., Boutros, M. and Hiesinger, P. R. (2021). Systematic functional analysis of Rab GTPases reveals limits of neuronal robustness to environmental challenges in flies. Elife 10. PubMed ID: 33666175

    Kiral, F. R., Linneweber, G. A., Mathejczyk, T., Georgiev, S. V., Wernet, M. F., Hassan, B. A., von Kleist, M. and Hiesinger, P. R. (2020). Autophagy-dependent filopodial kinetics restrict synaptic partner choice during Drosophila brain wiring. Nat Commun 11(1): 1325. PubMed ID: 32165611

    Ozel, M. N., Kulkarni, A., Hasan, A., Brummer, J., Moldenhauer, M., Daumann, I. M., Wolfenberg, H., Dercksen, V. J., Kiral, F. R., Weiser, M., Prohaska, S., von Kleist, M. and Hiesinger, P. R. (2019). Serial synapse formation through filopodial competition for synaptic seeding factors. Dev Cell. PubMed ID: 31353313

    Jin, E. J., Kiral, F. R., Ozel, M. N., Burchardt, L. S., Osterland, M., Epstein, D., Wolfenberg, H., Prohaska, S. and Hiesinger, P. R. (2018). Live observation of two parallel membrane degradation pathways at axon terminals. Curr Biol. PubMed ID: 29551411

    Özel, M.N., Langen, M., Hassan, B.A. and Hiesinger, P.R. (2015). Filopodial dynamics and growth cone stabilization in Drosophila visual circuit development. Elife [Epub ahead of print]. PubMed ID: 26512889

    Agi, E., Langen, M., Altschuler, S. J., Wu, L. F., Zimmermann, T. and Hiesinger, P. R. (2014). The evolution and development of neural superposition. J Neurogenet 28: 216-232. PubMed ID: 24912630

    Langen, M., Agi, E., Altschuler, D.J., Wu, L.F., Altschuler, S.J. and Hiesinger, P.R. (2015). The developmental rules of neural superposition in Drosophila. Cell [Epub ahead of print]. PubMed ID: 26119341

    Wang, D., Epstein, D., Khalaf, O., Srinivasan, S., Williamson, W. R., Fayyazuddin, A., Quiocho, F. A. and Hiesinger, P. R. (2014). Ca2+-Calmodulin regulates SNARE assembly and spontaneous neurotransmitter release via v-ATPase subunit V0a1. J Cell Biol 205: 21-31. PubMed ID: 24733584

  • Ville Hietakangas Institute of Biotechnology, University of Helsinki
    van den Berg, L., Kokki, K., Wowro, S. J., Petricek, K. M., Deniz, O., Stegmann, C. A., Robciuc, M., Teesalu, M., Melvin, R. G., Nieminen, A. I., Schupp, M. and Hietakangas, V. (2023). Sugar-responsive inhibition of Myc-dependent ribosome biogenesis by Clockwork orange. Cell Rep 42(7): 112739. PubMed ID: 37405919

    Kokki, K., Lamichane, N., Nieminen, A. I., Ruhanen, H., Morikka, J., Robciuc, M., Rovenko, B. M., Havula, E., Käkelä, R. and Hietakangas, V. (2021). Metabolic gene regulation by Drosophila GATA transcription factor Grain. PLoS Genet 17(10): e1009855. PubMed ID: 34634038

    Hasygar, K., Deniz, O., Liu, Y., Gullmets, J., Hynynen, R., Ruhanen, H., Kokki, K., Kakela, R. and Hietakangas, V. (2020). Coordinated control of adiposity and growth by anti-anabolic kinase ERK7. EMBO Rep: e201949602. PubMed ID: 33369866

    Havula, E. and Hietakangas, V. (2017). Sugar sensing by ChREBP/Mondo-Mlx-new insight into downstream regulatory networks and integration of nutrient-derived signals. Curr Opin Cell Biol 51: 89-96. PubMed ID: 29278834

    Teesalu, M., Rovenko, B. M. and Hietakangas, V. (2017). Salt-Inducible Kinase 3 Provides Sugar Tolerance by Regulating NADPH/NADP+ Redox Balance. Curr Biol 27(3): 458-464. PubMed ID: 28132818

    Mattila, J., Havula, E., Suominen, E., Teesalu, M., Surakka, I., Hynynen, R., Kilpinen, H., Väänänen, J., Hovatta, I., Käkelä, R., Ripatti, S., Sandmann, T. and Hietakangas, V. (2015). Mondo-Mlx mediates organismal sugar sensing through the Gli-similar transcription factor Sugarbabe. Cell Rep [Epub ahead of print]. PubMed ID: 26440885

    Bartok, O., Teesalu, M., Ashwall-Fluss, R., Pandey, V., Hanan, M., Rovenko, B.M., Poukkula, M., Havula, E., Moussaieff, A., Vodala, S., Nahmias, Y., Kadener, S. and Hietakangas, V. (2015). The transcription factor Cabut coordinates energy metabolism and the circadian clock in response to sugar sensing. EMBO J [Epub ahead of print]. PubMed ID: 25916830

    Hasygar, K. and Hietakangas, V. (2014). p53- and ERK7-dependent ribosome surveillance response regulates Drosophila Insulin-Like peptide secretion. PLoS Genet 10: e1004764. PubMed ID: 25393288

    Havula, E., Teesalu, M., Hyotylainen, T., Seppala, H., Hasygar, K., Auvinen, P., Oresic, M., Sandmann, T., Hietakangas, V. (2013) Mondo/ChREBP-Mlx-Regulated Transcriptional Network Is Essential for Dietary Sugar Tolerance in Drosophila. PLoS Genet 9: e1003438. PubMed ID: 23593032

    Kallijarvi, J., Stratoulias, V., Virtanen, K., Hietakangas, V., Heino, T. I. and Saarma, M. (2012). Characterization of Drosophila GDNF receptor-like and evidence for its evolutionarily conserved interaction with neural cell adhesion molecule (NCAM)/FasII. PLoS One 7: e51997. PubMed ID: 23284846

    Zhang, W., Thompson, B. J., Hietakangas, V. and Cohen, S. M. (2011). MAPK/ERK signaling regulates insulin sensitivity to control glucose metabolism in Drosophila. PLoS Genet 7: e1002429. PubMed ID: 22242005

  • Pejmun Haghighi Buck Institute for Research on Aging, Novato, CA
    Liao, E. H., Gray, L., Tsurudome, K., El Mounzer, W., Elazzouzi, F., Baim, C., Farzin, S., Calderon, M. R., Kauwe, G. and Haghighi, A. P. (2018). Kinesin Khc-73/KIF13B modulates retrograde BMP signaling by influencing endosomal dynamics at the Drosophila neuromuscular junction. PLoS Genet 14(1): e1007184. PubMed ID: 29373576

    Penney, J., Tsurudome, K., Liao, E. H., Kauwe, G., Gray, L., Yanagiya, A., M, R. C., Sonenberg, N. and Haghighi, A. P. (2016). LRRK2 regulates retrograde synaptic compensation at the Drosophila neuromuscular junction. Nat Commun 7: 12188. PubMed ID: 27432119

    Penney, J., Tsurudome, K., Liao, E. H., Elazzouzi, F., Livingstone, M., Gonzalez, M., Sonenberg, N. and Haghighi, A. P. (2012). TOR is required for the retrograde regulation of synaptic homeostasis at the Drosophila neuromuscular junction. Neuron 74(1): 166-178. PubMed ID: 22500638

  • Arthur Hilliker Department of Biology, York University, Toronto
    Cotsworth, S., Jackson, C. J., Hallson, G., Fitzpatrick, K. A., Syrzycka, M., Coulthard, A. B., Bejsovec, A., Marchetti, M., Pimpinelli, S., Wang, S. J. H., Camfield, R. G., Verheyen, E. M., Sinclair, D. A., Honda, B. M. and Hilliker, A. J. (2022). Characterization of Gfat1 (zeppelin) and Gfat2, Essential Paralogous Genes Which Encode the Enzymes That Catalyze the Rate-Limiting Step in the Hexosamine Biosynthetic Pathway in Drosophila melanogaster. Cells 11(3). PubMed ID: 35159258

    Coulthard, A. B., Taylor-Kamall, R. W., Hallson, G., Axentiev, A., Sinclair, D. A., Honda, B. M. and Hilliker, A. J. (2016). Meiotic recombination is suppressed near the histone-defined border of euchromatin and heterochromatin on chromosome 2L of Drosophila melanogaster. Genome 59: 289-294. PubMed ID: 27031007

    Belozerov, V. E., Ratkovic, S., McNeill, H., Hilliker, A. J. and McDermott, J. C. (2014). In vivo interaction proteomics reveal a novel p38 mitogen-activated protein kinase/Rack1 pathway regulating proteostasis in Drosophila muscle. Mol Cell Biol 34: 474-484. PubMed ID: 24277934

    Hilliker, A. J. and Taylor-Kamall, R. W. (2013). Heterochromatin and genome size in Drosophila. Genome 56: 473-474. PubMed ID: 24168630

  • Gary Hime School of Biomedical Sciences, Melbourne University
    Siddall, N. A., Casagranda, F., Johanson, T. M., Dominado, N., Heaney, J., Sutherland, J. M., McLaughlin, E. A. and Hime, G. R. (2022). MiMIC analysis reveals an isoform specific role for Drosophila Musashi in follicle stem cell maintenance and escort cell function. Cell Death Discov 8(1): 455. PubMed ID: 36371343

    Yang, Y., Kong, R., Goh, F. G., Somers, W. G., Hime, G. R., Li, Z. and Cai, Y. (2021). dRTEL1 is essential for the maintenance of Drosophila male germline stem cells. PLoS Genet 17(10): e1009834. PubMed ID: 34644293

    Siddall, N. A. and Hime, G. R. (2017). A Drosophila toolkit for defining gene function in spermatogenesis. Reproduction 153(4): R121-R132. PubMed ID: 28073824

    >Dominado, N., La Marca, J. E., Siddall, N. A., Heaney, J., Tran, M., Cai, Y., Yu, F., Wang, H., Somers, W. G., Quinn, L. M. and Hime, G. R. (2016). Rbf Regulates Drosophila Spermatogenesis via Control of Somatic Stem and Progenitor Cell Fate in the Larval Testis. Stem Cell Reports 7(6): 1152-1163. PubMed ID: 27974223

  • David Hipfner Institut de recherches cliniques de Montréal, University of Montreal
    Saad, F. and Hipfner, D. R. (2021). Extensive crosstalk of G protein-coupled receptors with the Hedgehog signalling pathway. Development. PubMed ID: 33653875

    De Jamblinne, C. V., Decelle, B., Dehghani, M., Joseph, M., Sriskandarajah, N., Leguay, K., Rambaud, B., Lemieux, S., Roux, P. P., Hipfner, D. R. and Carreno, S. (2020). STRIPAK regulates Slik localization to control mitotic morphogenesis and epithelial integrity. J Cell Biol 219(11). PubMed ID: 32960945

    Praktiknjo, S. D., Saad, F., Maier, D., Ip, P. and Hipfner, D. R. (2018). Activation of Smoothened in the Hedgehog pathway unexpectedly increases Galphas-dependent cAMP levels in Drosophila. J Biol Chem. PubMed ID: 30018136

    Panneton, V., Nath, A., Sader, F., Delaunay, N., Pelletier, A., Maier, D., Oh, K. and Hipfner, D. R. (2015). Regulation of Catalytic and Non-catalytic Functions of the Drosophila Ste20 Kinase Slik by Activation Segment Phosphorylation. J Biol Chem [Epub ahead of print]. PubMed ID: 26170449

    Maier, D., Cheng, S., Faubert, D. and Hipfner, D. R. (2014). A broadly conserved g-protein-coupled receptor kinase phosphorylation mechanism controls Drosophila smoothened activity. PLoS Genet 10: e1004399. PubMed ID: 25009998

    Tan, J., Oh, K., Burgess, J., Hipfner, D. R. and Brill, J. A. (2014). PI4KIIIalpha is required for cortical integrity and cell polarity during Drosophila oogenesis. J Cell Sci. PubMed ID: 24413170

    Maier, D., Cheng, S. and Hipfner, D. R. (2012). The complexities of G-protein-coupled receptor kinase function in Hedgehog signaling. Fly (Austin) 6: 135-141. PubMed ID: 22653052

    Cheng, S., Maier, D. and Hipfner, D. R. (2012). Drosophila G-protein-coupled receptor kinase 2 regulates cAMP-dependent Hedgehog signaling. Development 139: 85-94. PubMed ID: 22096079

  • Yoshio Hirabayashi Riken Brain Science Institute. Wako, Saitama Prefecture
    Kohyama-Koganeya, A., Kurosawa, M. and Hirabayashi, Y. (2017). Loss of BOSS causes shortened lifespan with mitochondrial dysfunction in Drosophila. PLoS One 12(1): e0169073. PubMed ID: 28045997

    Kohyama-Koganeya, A., Nabetani, T., Miura, M. and Hirabayashi, Y. (2011). Glucosylceramide synthase in the fat body controls energy metabolism in Drosophila. J Lipid Res 52: 1392-1399. PubMed ID: 21550991

    Kohyama-Koganeya, A. and Hirabayashi, Y. (2010). The Drosophila 7-pass transmembrane glycoprotein BOSS and metabolic regulation: What Drosophila can teach us about human energy metabolism. Methods Enzymol 480: 525-538. PubMed ID: 20816226

  • Yukinori Hirano Hong Kong University of Science and Technology
    Takakura, M., Nakagawa, R., Ota, T., Kimura, Y., Ng, M. Y., Alia, A. G., Okuno, H. and Hirano, Y. (2021). Rpd3/CoRest-mediated activity-dependent transcription regulates the flexibility in memory updating in Drosophila. Nat Commun 12(1): 628. PubMed ID: 33504795

    Awata, H., Takakura, M., Kimura, Y., Iwata, I., Masuda, T. and Hirano, Y. (2019). The neural circuit linking mushroom body parallel circuits induces memory consolidation in Drosophila. Proc Natl Acad Sci U S A 116(32): 16080-16085. PubMed ID: 31337675

    Enya, S., Yamamoto, C., Mizuno, H., Esaki, T., Lin, H. K., Iga, M., Morohashi, K., Hirano, Y., Kataoka, H., Masujima, T., Shimada-Niwa, Y. and Niwa, R. (2017). Dual Roles of Glutathione in Ecdysone Biosynthesis and Antioxidant Function During Larval Development in Drosophila. Genetics 207(4): 1519-1532. PubMed ID: 29021278

  • Yasushi Hiromi National Institute of Genetics, Mishima, Shizuoka
    Matsuoka, S., Gupta, S., Suzuki, E., Hiromi, Y. and Asaoka, M. (2014). gone early, a novel germline factor, ensures the proper size of the stem cell precursor pool in the Drosophila ovary. PLoS One 9: e113423. PubMed ID: 25420147

    Matsuoka, S., Hiromi, Y., Asaoka, M. (2013) Egfr signaling controls the size of the stem cell precursor pool in the Drosophila ovary. Mech Dev. PubMed ID: 23376160

    Katsuki, T., Ailani, D., Hiramoto, M. and Hiromi, Y. (2009). Intra-axonal patterning: intrinsic compartmentalization of the axonal membrane in Drosophila neurons. Neuron 64: 188-199. PubMed ID: 19874787

  • Susumu Hirose National Institute of Genetics, Mishima, Shizuoka
    Wang, X. F., Liu, J. X., Ma, Z. Y., Shen, Y., Zhang, H. R., Zhou, Z. Z., Suzuki, E., Liu, Q. X. and Hirose, S. (2020). Evolutionarily Conserved Roles for Apontic in Induction and Subsequent Decline of Cyclin E Expression. iScience 23(8): 101369. PubMed ID: 32736066

    Nishioka, K., Wang, X. F., Miyazaki, H., Soejima, H. and Hirose, S. (2018). Mbf1 ensures Polycomb silencing by protecting E(z) mRNA from degradation by Pacman. Development 145(5). PubMed ID: 29523653

    Wang, X. F., Shen, Y., Cheng, Q., Fu, C. L., Zhou, Z. Z., Hirose, S. and Liu, Q. X. (2017). Apontic directly activates hedgehog and cyclin E for proper organ growth and patterning. Sci Rep 7(1): 12470. PubMed ID: 28963499

    Fuse, N., Yu, F. and Hirose, S. (2013). Gprk2 adjusts Fog signaling to organize cell movements in Drosophila gastrulation. Development. PubMed ID: 24026125

    Akagi, K., Kageyama, Y., Kayashima, Y., Takakura, Y., Hirose, S. and Ueda, H. (2013). The binding of multiple nuclear receptors to a single regulatory region is important for the proper expression of EDG84A in Drosophila melanogaster. J Mol Biol 425: 71-81. PubMed ID: 23137796

    Kanesaki, T., Hirose, S., Grosshans, J. and Fuse, N. (2013). Heterotrimeric G protein signaling governs the cortical stability during apical constriction in Drosophila gastrulation. Mech Dev 130: 132-142. PubMed ID: 23085574

  • Jay Hirsh UVA Neuroscience Graduate Program, Charlottesville
    Aso, Y., Ray, R. P., Long, X., Bushey, D., Cichewicz, K., Ngo, T. T., Sharp, B., Christoforou, C., Hu, A., Lemire, A. L., Tillberg, P., Hirsh, J., Litwin-Kumar, A. and Rubin, G. M. (2019). Nitric oxide acts as a cotransmitter in a subset of dopaminergic neurons to diversify memory dynamics. Elife 8. PubMed ID: 31724947

    Niens, J., Reh, F., Coban, B., Cichewicz, K., Eckardt, J., Liu, Y. T., Hirsh, J. and Riemensperger, T. D. (2017). Dopamine Modulates Serotonin Innervation in the Drosophila Brain. Front Syst Neurosci 11: 76. PubMed ID: 29085286

    Cichewicz, K., Garren, E. J., Adiele, C., Aso, Y., Wang, Z., Wu, M., Birman, S., Rubin, G. M. and Hirsh, J. (2016). A new brain dopamine deficient Drosophila and its pharmacological and genetic Rescue. Genes Brain Behav [Epub ahead of print]. PubMed ID: 27762066

    Nall, A.H., Shakhmantsir, I., Cichewicz, K. Birman, S., Hirsh, J. and Sehgal, A. (2016). Caffeine promotes wakefulness via dopamine signaling in Drosophila. Sci Rep 6: 20938. PubMed ID: 26868675

    Kistenpfennig, C., Hirsh, J., Yoshii, T. and Helfrich-Forster, C. (2012). Phase-shifting the fruit fly clock without cryptochrome. J Biol Rhythms 27: 117-125. PubMed ID: 22476772

    Lee, P. T., Lin, H. W., Chang, Y. H., Fu, T. F., Dubnau, J., Hirsh, J., Lee, T. and Chiang, A. S. (2011). Serotonin-mushroom body circuit modulating the formation of anesthesia-resistant memory in Drosophila. Proc Natl Acad Sci U S A 108: 13794-13799. PubMed ID: 21808003

    Kaun, K. R., Azanchi, R., Maung, Z., Hirsh, J. and Heberlein, U. (2011). A Drosophila model for alcohol reward. Nat Neurosci 14: 612-619. PubMed ID: 21499254

    Riemensperger, T., Isabel, G., Coulom, H., Neuser, K., Seugnet, L., Kume, K., Iche-Torres, M., Cassar, M., Strauss, R., Preat, T., Hirsh, J. and Birman, S. (2011). Behavioral consequences of dopamine deficiency in the Drosophila central nervous system. Proc Natl Acad Sci U S A 108: 834-839. PubMed ID: 21187381

  • Frank Hirth Kings College London
    Buhl, E., Kottler, B., Hodge, J. J. L. and Hirth, F. (2021). Thermoresponsive motor behavior is mediated by ring neuron circuits in the central complex of Drosophila. Sci Rep 11(1): 155. PubMed ID: 33420240

    Bridi, J. C., Ludlow, Z. N., Kottler, B., Hartmann, B., Vanden Broeck, L., Dearlove, J., Göker, M., Strausfeld, N. J., Callaerts, P. and Hirth, F. (2020). Ancestral regulatory mechanisms specify conserved midbrain circuitry in arthropods and vertebrates. Proc Natl Acad Sci U S A 117(32): 19544-19555. PubMed ID: 32747566

    Gonçalves-Pimentel, C., Mazaud, D., Kottler, B., Proelss, S., Hirth, F. and Fanto, M. (2020). A miRNA screen procedure identifies garz as an essential factor in adult glia functions and validates Drosophila as a beneficial 3Rs model to study glial functions and GBF1 biology. F1000Res 9: 317. PubMed ID: 32595956

    Bridi, J. C., Ludlow, Z. N. and Hirth, F. (2019). Lineage-specific determination of ring neuron circuitry in the central complex of Drosophila. Biol Open 8(7). PubMed ID: 31285267

    Mazaud, D., Kottler, B., Goncalves-Pimentel, C., Proelss, S., Tuchler, N., Deneubourg, C., Yuasa, Y., Diebold, C., Jungbluth, H., Lai, E. C., Hirth, F., Giangrande, A. and Fanto, M. (2019). Transcriptional regulation of the Glutamate/GABA/Glutamine cycle in adult glia controls motor activity and seizures in Drosophila. J Neurosci. PubMed ID: 31064860
    Kottler, B., Faville, R., Bridi, J. C. and Hirth, F. (2019). Inverse control of turning behavior by Dopamine D1 receptor signaling in columnar and ring neurons of the central complex in Drosophila. Curr Biol. PubMed ID: 30713106

    Solomon, D. A., Stepto, A., Au, W. H., Adachi, Y., Diaper, D. C., Hall, R., Rekhi, A., Boudi, A., Tziortzouda, P., Lee, Y. B., Smith, B., Bridi, J. C., Spinelli, G., Dearlove, J., Humphrey, D. M., Gallo, J. M., Troakes, C., Fanto, M., Soller, M., Rogelj, B., Parsons, R. B., Shaw, C. E., Hortobagyi, T. and Hirth, F. (2018). A feedback loop between dipeptide-repeat protein, TDP-43 and karyopherin-alpha mediates C9orf72-related neurodegeneration. Brain 141(10): 2908-2924. PubMed ID: 30239641

    Shaw, R. E., Kottler, B., Ludlow, Z. N., Buhl, E., Kim, D., Morais da Silva, S., Miedzik, A., Coum, A., Hodge, J. J., Hirth, F. and Sousa-Nunes, R. (2018). In vivo expansion of functionally integrated GABAergic interneurons by targeted increase in neural progenitors. Embo j. Pubmed ID: 29728368

    Stepto, A., Gallo, J. M., Shaw, C. E. and Hirth, F. (2013). Modelling C9ORF72 hexanucleotide repeat expansion in amyotrophic lateral sclerosis and frontotemporal dementia. Acta Neuropathol. PubMed ID: 24366528

    Diaper, D. C. and Hirth, F. (2014). Immunostaining of the developing embryonic and larval Drosophila brain. Methods Mol Biol 1082: 3-17. PubMed ID: 24048923

  • Michael Hoch Life and Medical Sciences Institute, University of Bonn
    Loch, G., Zinke, I., Mori, T., Carrera, P., Schroer, J., Takeyama, H. and Hoch, M. (2017). Antimicrobial peptides extend lifespan in Drosophila. PLoS One 12(5): e0176689. PubMed ID: 28520752

    Sellin, J., Schulze, H., Paradis, M., Gosejacob, D., Papan, C., Shevchenko, A., Psathaki, O. E., Paululat, A., Thielisch, M., Sandhoff, K. and Hoch, M. (2017). Characterization of Drosophila saposin-related mutants as a model for lysosomal sphingolipid storage diseases. Dis Model Mech. PubMed ID: 28389479

    Richard, M., Bauer, R., Tavosanis, G. and Hoch, M. (2017). The gap junction protein Innexin3 is required for eye disc growth in Drosophila. Dev Biol [Epub ahead of print]. PubMed ID: 28390801

    Wakabayashi, S., Sawamura, N., Voelzmann, A., Broemer, M., Asahi, T. and Hoch, M. (2016). Ohgata, the single Drosophila ortholog of human Cereblon, regulates insulin signaling-dependent organismic growth. J Biol Chem [Epub ahead of print]. PubMed ID: 27702999

    Richard, M. and Hoch, M. (2015). Drosophila eye size is determined by Innexin 2-dependent Decapentaplegic signalling. Dev Biol. PubMed ID: 26455410

    Varma, D., Bulow, M. H., Pesch, Y. Y., Loch, G. and Hoch, M. (2014). Forkhead, a new cross regulator of metabolism and innate immunity downstream of TOR in Drosophila. J Insect Physiol. PubMed ID: 24842780

    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: e97332. PubMed ID: 24845618

    Bulow, M. H., Bulow, T. R., Hoch, M., Pankratz, M. J. and Junger, M. A. (2014). Src tyrosine kinase signaling antagonizes nuclear localization of FOXO and inhibits its transcription factor activity. Sci Rep 4: 4048. PubMed ID: 24513978

    Hahn, I., Fuss, B., Peters, A., Werner, T., Sieberg, A., Gosejacob, D., Hoch, M. (2013) The drosophila Arf GEF steppke controls MAPK activation in EGFR signaling. J Cell Sci. PubMed ID: 23549788

    Bauer, R., Voelzmann, A., Breiden, B., Schepers, U., Farwanah, H., Hahn, I., Eckardt, F., Sandhoff, K. and Hoch, M. (2009). Schlank, a member of the ceramide synthase family controls growth and body fat in Drosophila. EMBO J 28: 3706-3716. PubMed ID: 19834458

    Wingen, C., Stumpges, B., Hoch, M. and Behr, M. (2009). Expression and localization of clathrin heavy chain in Drosophila melanogaster. Gene Expr Patterns 9: 549-554. PubMed ID: 19577664

  • James Hodge Physiology and Pharmacology, University of Bristol
    Buhl, E., Kim, Y. A., Parsons, T., Zhu, B., Santa-Maria, I., Lefort, R. and Hodge, J. J. L. (2022). Effects of Eph/ephrin signalling and human Alzheimer's disease-associated EphA1 on Drosophila behaviour and neurophysiology. Neurobiol Dis 170: 105752. PubMed ID: 35569721

    Tasman, K., Hidalgo, S., Zhu, B., Rands, S. A. and Hodge, J. J. L. (2021). Neonicotinoids disrupt memory, circadian behaviour and sleep. Sci Rep 11(1): 2061. PubMed ID: 33479461

    Smith, P., Buhl, E., Tsaneva-Atanasova, K. and Hodge, J. J. L. (2019). Shaw and Shal voltage-gated potassium channels mediate circadian changes in Drosophila clock neuron excitability. J Physiol. PubMed ID: 31612994

    Higham, J. P., Hidalgo, S., Buhl, E. and Hodge, J. J. L. (2019). Restoration of olfactory memory in Drosophila overexpressing human Alzheimer's disease associated Tau by manipulation of L-type Ca(2+) channels. Front Cell Neurosci 13: 409. PubMed ID: 31551716

    Higham, J. P., Malik, B. R., Buhl, E., Dawson, J. M., Ogier, A. S., Lunnon, K. and Hodge, J. J. L. (2019). Alzheimer's disease associated genes Ankyrin and Tau cause shortened lifespan and memory loss in Drosophila. Front Cell Neurosci 13: 260. PubMed ID: 31244615

    Buhl, E., Higham, J. P. and Hodge, J. J. L. (2019). Alzheimer's disease-associated tau alters Drosophila circadian activity, sleep and clock neuron electrophysiology. Neurobiol Dis: 104507. PubMed ID: 31207389

    Lowe, S. A., Usowicz, M. M. and Hodge, J. J. L. (2019). Neuronal overexpression of Alzheimer's disease and Down's syndrome associated DYRK1A/minibrain gene alters motor decline, neurodegeneration and synaptic plasticity in Drosophila. Neurobiol Dis 125: 107-114. PubMed ID: 30703437

    Deakin, A., Mendl, M., Browne, W. J., Paul, E. S. and Hodge, J. J. L. (2018). State-dependent judgement bias in Drosophila: evidence for evolutionarily primitive affective processes. Biol Lett 14(2). PubMed ID: 29491031

    Julienne, H., Buhl, E., Leslie, D. S. and Hodge, J. J. L. (2017). Drosophila PINK1 and parkin loss-of-function mutants display a range of non-motor Parkinson's disease phenotypes. Neurobiol Dis 104: 15-23. PubMed ID: 28435104

    Buhl, E., Bradlaugh, A., Ogueta, M., Chen, K. F., Stanewsky, R. and Hodge, J. J. (2016). Quasimodo mediates daily and acute light effects on Drosophila clock neuron excitability. Proc Natl Acad Sci U S A. PubMed ID: 27821737

  • Ary Hoffmann Department of Genetics, University of Melbourne
    Kellermann, V., Overgaard, J., Sgro, C. M. and Hoffmann, A. A. (2022). Phylogenetic and environmental patterns of sex differentiation in physiological traits across Drosophila species. J Evol Biol 35(11): 1548-1557. PubMed ID: 36196885

    Gu, X., Chen, W., Perry, T., Batterham, P. and Hoffmann, A. A. (2021). Genomic knockout of hsp23 both decreases and increases fitness under opposing thermal extremes in Drosophila melanogaster. Insect Biochem Mol Biol 139: 103652. PubMed ID: 34562590

    Telonis-Scott, M. and Hoffmann, A. A. (2018). Enhancing Ebony? Common Associations With a cis-Regulatory Haplotype for Drosophila melanogaster Thoracic Pigmentation in a Japanese Population and Australian Populations. Front Physiol 9: 822. PubMed ID: 30042686

    Turelli, M., Cooper, B. S., Richardson, K. M., Ginsberg, P. S., Peckenpaugh, B., Antelope, C. X., Kim, K. J., May, M. R., Abrieux, A., Wilson, D. A., Bronski, M. J., Moore, B. R., Gao, J. J., Eisen, M. B., Chiu, J. C., Conner, W. R. and Hoffmann, A. A. (2018). Rapid Global Spread of wRi-like Wolbachia across Multiple Drosophila. Curr Biol 28(6): 963-971.e968. PubMed ID: 29526588

    Hangartner, S., Dworkin, I., DeNieu, M. and Hoffmann, A. A. (2017). Does increased heat resistance result in higher susceptibility to predation? A test using Drosophila melanogaster selection and hardening. J Evol Biol. PubMed ID: 28386918

    O'Brien, E. K., Higgie, M., Reynolds, A., Hoffmann, A. A. and Bridle, J. R. (2017). Testing for local adaptation and evolutionary potential along altitudinal gradients in rainforest Drosophila: beyond laboratory estimates. Glob Chang Biol. PubMed ID: 28070978

    Griffin, P. C., Hangartner, S. B., Fournier-Level, A. and Hoffmann, A. A. (2016). Genomic trajectories to desiccation resistance: Convergence and divergence among replicate selected Drosophila lines. Genetics [Epub ahead of print]. PubMed ID: 28007884

    Kriesner, P., Conner, W. R., Weeks, A. R., Turelli, M. and Hoffmann, A. A. (2016). Persistence of a Wolbachia infection frequency cline in Drosophila melanogaster and the possible role of reproductive dormancy. Evolution [Epub ahead of print]. PubMed ID: 27076356

    Rane, R. V., Rako, L., Kapun, M., Lee, S. F. and Hoffmann, A. A. (2015). Genomic evidence for role of inversion 3RP of Drosophila melanogaster in facilitating climate change adaptation. Mol Ecol [Epub ahead of print]. PubMed ID: 25789416

  • Jules Hoffmann Molecular and Cellular Biology Institut, Strasbourg
    Chen, D., Roychowdhury-Sinha, A., Prakash, P., Lan, X., Fan, W., Goto, A. and Hoffmann, J. A. (2021). A time course transcriptomic analysis of host and injected oncogenic cells reveals new aspects of Drosophila immune defenses. Proc Natl Acad Sci U S A 118(12). PubMed ID: 33737397

    Lamiable, O., Kellenberger, C., Kemp, C., Troxler, L., Pelte, N., Boutros, M., Marques, J. T., Daeffler, L., Hoffmann, J. A., Roussel, A. and Imler, J. L. (2016). Cytokine Diedel and a viral homologue suppress the IMD pathway in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 26739560

    Chtarbanova, S., Lamiable, O., Lee, K. Z., Galiana, D., Troxler, L., Meignin, C., Hetru, C., Hoffmann, J. A., Daeffler, L. and Imler, J. L. (2014). Drosophila C virus systemic infection leads to intestinal obstruction. J Virol. PubMed ID: 25253354

    Goto, A., Fukuyama, H., Imler, J. L. and Hoffmann, J. A. (2014). The chromatin regulator DMAP1 modulates activity of the NF-kappaB transcription factor Relish in the Drosophila innate immune response. J Biol Chem. PubMed ID: 24947515

    Fukuyama, H., Verdier, Y., Guan, Y., Makino-Okamura, C., Shilova, V., Liu, X., Maksoud, E., Matsubayashi, J., Haddad, I., Spirohn, K., Ono, K., Hetru, C., Rossier, J., Ideker, T., Boutros, M., Vinh, J. and Hoffmann, J. A. (2013). Landscape of protein-protein interactions in Drosophila immune deficiency signaling during bacterial challenge. Proc Natl Acad Sci U S A. PubMed ID: 23749869

    Kemp, C., Mueller, S., Goto, A., Barbier, V., Paro, S., Bonnay, F., Dostert, C., Troxler, L., Hetru, C., Meignin, C., Pfeffer, S., Hoffmann, J. A. and Imler, J. L. (2013). Broad RNA interference-mediated antiviral immunity and virus-specific inducible responses in Drosophila. J Immunol 190: 650-658. PubMed ID: 23255357

    Liu, X., Sano, T., Guan, Y., Nagata, S., Hoffmann, J. A. and Fukuyama, H. (2012). Drosophila EYA regulates the immune response against DNA through an evolutionarily conserved threonine phosphatase motif. PLoS One 7: e42725. PubMed ID: 22916150

    Lemaitre, B., Nicolas, E., Michaut, L., Reichhart, J. M. and Hoffmann, J. A. (2012). Pillars article: the dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996. 86: 973-983. J Immunol 188: 5210-5220. PubMed ID: 22611248

    Fukuyama, H., Ndiaye, S., Hoffmann, J., Rossier, J., Liuu, S., Vinh, J. and Verdier, Y. (2012). On-bead tryptic proteolysis: an attractive procedure for LC-MS/MS analysis of the Drosophila caspase 8 protein complex during immune response against bacteria. J Proteomics 75: 4610-4619. PubMed ID: 22450469

  • Anneli Hoikkala Department of Biological and Environmental Science, University of Jyväskylä, Finland
    Poikela, N., Kinnunen, J., Wurdack, M., Kauranen, H., Schmitt, T., Kankare, M., Snook, R. R. and Hoikkala, A. (2019). Strength of sexual and postmating prezygotic barriers varies between sympatric populations with different histories and species abundances. Evolution. PubMed ID: 30957216

    Salminen, T. S., Hoikkala, A. (2013) Effect of temperature on the duration of sensitive period and on the number of photoperiodic cycles required for the induction of reproductive diapause in Drosophila montana. J Insect Physiol. PubMed ID: 23428942

    Kauranen, H., Menegazzi, P., Costa, R., Helfrich-Forster, C., Kankainen, A. and Hoikkala, A. (2012). Flies in the north: locomotor behavior and clock neuron organization of Drosophila montana. J Biol Rhythms 27: 377-387. PubMed ID: 23010660

    Kauranen, H., Menegazzi, P., Costa, R., Helfrich-Forster, C., Kankainen, A. and Hoikkala, A. (2012). Flies in the north: locomotor behavior and clock neuron organization of Drosophila montana. J Biol Rhythms 27: 377-387. PubMed ID: 23010660

    Veltsos, P., Wicker-Thomas, C., Butlin, R. K., Hoikkala, A. and Ritchie, M. G. (2012). Sexual selection on song and cuticular hydrocarbons in two distinct populations of Drosophila montana. Ecol Evol 2: 80-94. PubMed ID: 22408728

    Tyukmaeva, V. I., Salminen, T. S., Kankare, M., Knott, K. E. and Hoikkala, A. (2011). Adaptation to a seasonally varying environment: a strong latitudinal cline in reproductive diapause combined with high gene flow in Drosophila montana. Ecol Evol 1: 160-168. PubMed ID: 22393492

  • Peter Hollenbeck Department of Biological Sciences, Purdue University, West Lafayette, IN
    Liao, P. C., Tandarich, L. C. and Hollenbeck, P. J. (2017). ROS regulation of axonal mitochondrial transport is mediated by Ca2+ and JNK in Drosophila. PLoS One 12(5): e0178105. PubMed ID: 28542430

    Sung, H., Tandarich, L. C., Nguyen, K. and Hollenbeck, P. J. (2016). Compartmentalized Regulation of Parkin-Mediated Mitochondrial Quality Control in the Drosophila Nervous System In Vivo. J Neurosci 36: 7375-7391. PubMed ID: 27413149

    Devireddy, S., Sung, H., Liao, P. C., Garland-Kuntz, E. and Hollenbeck, P. J. (2014). Analysis of mitochondrial traffic in Drosophila. Methods Enzymol 547: 131-150. PubMed ID: 25416356

    Sun, E. W., Wagner, M. L., Maize, A., Kemler, D., Garland-Kuntz, E., Xu, L., Luo, Z. Q. and Hollenbeck, P. J. (2013). Legionella pneumophila infection of Drosophila S2 cells induces only minor changes in mitochondrial dynamics. PLoS One 8: e62972. PubMed ID: 23638172

  • Todd Holmes Physiology & Biophysics, School of Medicine, University of California, Irvine
    Au, D. D., Foden, A. J., Park, S. J., Nguyen, T. H., Liu, J. C., Tran, M. D., Jaime, O. G., Yu, Z. and Holmes, T. C. (2022). Mosquito cryptochromes expressed in Drosophila confer species-specific behavioral light responses. Curr Biol 32(17): 3731-3744. PubMed ID: 35914532

    Nave, C., Roberts, L., Hwu, P., Estrella, J. D., Vo, T. C., Nguyen, T. H., Bui, T. T., Rindner, D. J., Pervolarakis, N., Shaw, P. J., Leise, T. L. and Holmes, T. C. (2021). Weekend light shifts evoke persistent Drosophila circadian neural network desynchrony. J Neurosci. PubMed ID: 33931552

    Baik, L. S., Au, D. D., Nave, C., Foden, A. J., Enrriquez-Villalva, W. K. and Holmes, T. C. (2019). Distinct mechanisms of Drosophila CRYPTOCHROME-mediated light-evoked membrane depolarization and in vivo clock resetting. Proc Natl Acad Sci U S A 116(46): 23339-23344. PubMed ID: 31659046

    Baik, L. S., Recinos, Y., Chevez, J. A., Au, D. D. and Holmes, T. C. (2019). Multiple phototransduction inputs integrate to mediate UV light-evoked avoidance/attraction behavior in Drosophila. J Biol Rhythms: 748730419847339. PubMed ID: 31140349

    Baik, L. S., Recinos, Y., Chevez, J. A. and Holmes, T. C. (2018). Circadian modulation of light-evoked avoidance/attraction behavior in Drosophila. PLoS One 13(8): e0201927. PubMed ID: 30106957

    Baik, L.S., Fogle, K.J., Roberts, L., Galschiodt, A.M., Chevez, J.A., Recinos, Y., Nguy, V. and Holmes, T.C. (2017). Cryptochrome mediates behavioral executive choice in response to UV light. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 28062690

    Barber, A. F., Erion, R., Holmes, T. C. and Sehgal, A. (2016). Circadian and feeding cues integrate to drive rhythms of physiology in Drosophila insulin-producing cells. Genes Dev 30(23): 2596-2606. PubMed ID: 27979876

    Das, A., Holmes, T. C. and Sheeba, V. (2016). dTRPA1 in Non-circadian Neurons Modulates Temperature-dependent Rhythmic Activity in Drosophila melanogaster. J Biol Rhythms 31(3): 272-288. PubMed ID: 26868037

    Das, A., Holmes, T. C. and Sheeba, V. (2015). dTRPA1 Modulates Afternoon Peak of Activity of Fruit Flies Drosophila melanogaster. PLoS One 10(7): e0134213. PubMed ID: 26226013

  • Robert Holmgren Northwestern University, Evanston, Il
    {Brigui, 2015 #4463} Roberto, N., Becam, I., Plessis, A. and Holmgren, R. A. (2022). Engrailed, Suppressor of fused and Roadkill modulate the Drosophila GLI transcription factor Cubitus interruptus at multiple levels. Development 149(6). PubMed ID: 35290435

    Bruzzone, L., Arguelles, C., Sanial, M., Miled, S., Alvisi, G., Goncalves-Antunes, M., Qasrawi, F., Holmgren, R. A., Smibert, C. A., Lipshitz, H. D., Boccaccio, G. L., Plessis, A. and Becam, I. (2020). Regulation of the RNA-binding protein Smaug by the GPCR Smoothened via the kinase Fused. EMBO Rep 21(7): e48425. PubMed ID: 32383557

    Sanial, M., Becam, I., Hofmann, L., Behague, J., Arguelles, C., Gourhand, V., Bruzzone, L., Holmgren, R. A. and Plessis, A. (2017). Dose-dependent transduction of Hedgehog relies on phosphorylation-based feedback between the G-protein-coupled receptor Smoothened and the kinase Fused. Development 144(10): 1841-1850. PubMed ID: 28360132

    Brigui, A., Hofmann, L., Arguelles, C., Sanial, M., Holmgren, R. A. and Plessis, A. (2015). Control of the dynamics and homeostasis of the Drosophila Hedgehog receptor Patched by two C2-WW-HECT-E3 Ubiquitin ligases. Open Biol 5(10). PubMed ID: 26446620

    Hurtado, R. R., Harney, A. S., Heffern, M. C., Holbrook, R. J., Holmgren, R. A. and Meade, T. J. (2012). Specific inhibition of the transcription factor Ci by a cobalt(III) Schiff base-DNA conjugate. Mol Pharm 9(2): 325-333. PubMed ID: 22214326

  • Joost Holthuis Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Utrecht University, Netherlands
    Vacaru, A. M., van den Dikkenberg, J., Ternes, P., Holthuis, J. C. (2013) Ceramide phosphoethanolamine biosynthesis in Drosophila is mediated by a unique ethanolamine phosphotransferase in the Golgi lumen. J Biol Chem. Pubmed ID: 23449981

    Vacaru, A. M., Tafesse, F. G., Ternes, P., Kondylis, V., Hermansson, M., Brouwers, J. F., Somerharju, P., Rabouille, C. and Holthuis, J. C. (2009). Sphingomyelin synthase-related protein SMSr controls ceramide homeostasis in the ER. J Cell Biol 185: 1013-1027. PubMed ID: 19506037

  • Marina Holz New York Medical College, Valhalla, NY
    Topfer, U., Bischoff, M. C., Bartkuhn, M. and Holz, A. (2019). Serpent/dGATAb regulates Laminin B1 and Laminin B2 expression during Drosophila embryogenesis. Sci Rep 9(1): 15910. PubMed ID: 31685844

    Wolfstetter, G., Dahlitz, I., Pfeifer, K., Topfer, U., Alt, J. A., Pfeifer, D. C., Lakes-Harlan, R., Baumgartner, S., Palmer, R. H. and Holz, A. (2019). Characterization of Drosophila Nidogen/entactin reveals roles in basement membrane stability, barrier function and nervous system patterning. Development 146(2). PubMed ID: 30567930

    Rudolf, A., Buttgereit, D., Jacobs, M., Wolfstetter, G., Kesper, D., Putz, M., Berger, S., Renkawitz-Pohl, R., Holz, A. and Onel, S. F. (2014). Distinct genetic programs guide Drosophila circular and longitudinal visceral myoblast fusion. BMC Cell Biol 15: 27. PubMed ID: 25000973

  • James Castelli-Gair Hombría Centro Andaluz de Biología del Desarrollo
    Sanchez-Higueras, C., Rastogi, C., Voutev, R., Bussemaker, H. J., Mann, R. S. and Hombria, J. C. (2019). In vivo Hox binding specificity revealed by systematic changes to a single cis regulatory module. Nat Commun 10(1): 3597. PubMed ID: 31399572

    Sotillos, S., Aguilar-Aragon, M. and Hombria, J. C. (2018). Functional analysis of the Drosophila RhoGAP Cv-c protein and its equivalence to the human DLC3 and DLC1 proteins. Sci Rep 8(1): 4601. PubMed ID: 29545526

    Sanchez-Higueras, C. and Hombria, J. C. (2016). Precise long-range migration results from short-range stepwise migration during ring gland organogenesis. Dev Biol. [Epub ahead of print] PubMed ID: 27063193

    Pinto, P.B., Espinosa-Vázquez, J.M., Rivas, M.L. and Hombría, J.C. (2015). JAK/STAT and Hox dynamic interactions in an organogenetic gene cascade. PLoS Genet 11: e1005412. PubMed ID: 26230388

    Sanchez-Higueras, C., Sotillos, S. and Castelli-Gair Hombria, J. (2013). Common Origin of Insect Trachea and Endocrine Organs from a Segmentally Repeated Precursor. Curr Biol. PubMed ID: 24332544

    Hombria, J. C. and Sotillos, S. (2013). JAK-STAT pathway in morphogenesis: From organ selector to cell behavior regulator. JAKSTAT 2: e26089. PubMed ID: 24069568

    Hombria, J. C. and Serras, F. (2013). Why should we care about fly tumors?: The case of JAK-STAT and EGFR cooperation in oncogenesis. JAKSTAT 2: e23203. PubMed ID: 24058803

    Sotillos, S., Aguilar, M., Hombria, J. C. (2013) Forces shaping a Hox morphogenetic gene network. Proc Natl Acad Sci U S A 110: 4303-4308. PubMed ID: 23440219

    Sotillos, S., Krahn, M., Espinosa-Vazquez, J. M., Hombria, J. C. (2013) Src kinases mediate the interaction of the apical determinant Bazooka/PAR3 with STAT92E and increase signalling efficiency in Drosophila ectodermal cells. Development. PubMed ID: 23462467

    Sotillos, S., Krahn, M., Espinosa-Vazquez, J. M. and Hombria, J. C. (2013). Src kinases mediate the interaction of the apical determinant Bazooka/PAR3 with STAT92E and increase signalling efficiency in Drosophila ectodermal cells. Development. PubMed ID: 23462467

    Sotillos, S., Espinosa-Vazquez, J. M., Foglia, F., Hu, N. and Hombria, J. C. (2010). An efficient approach to isolate STAT regulated enhancers uncovers STAT92E fundamental role in Drosophila tracheal development. Dev Biol 340: 571-582. PubMed ID: 20171201

  • Yang Hong University of Pittsburgh University of Pittsburgh, Department of Cell Biology
    Lu, J., Dong, W., Hammond, G. R. and Hong, Y. (2022). Hypoxia controls plasma membrane targeting of polarity proteins by dynamic turnover of PI4P and PI(4,5)P2. Elife 11. PubMed ID: 35678383

    Lu, J., Dong, W., Tao, Y. and Hong, Y. (2021). Electrostatic plasma membrane targeting contributes to Dlg function in cell polarity and tumorigenesis. Development. PubMed ID: 33688074

    Dong, W., Lu, J., Zhang, X., Wu, Y., Lettieri, K., Hammond, G. R. and Hong, Y. (2020). A polybasic domain in aPKC mediates Par6-dependent control of membrane targeting and kinase activity. J Cell Biol 219(7). PubMed ID: 32580209

    Chen, Y.J., Huang, J., Huang, L., Austin, E. and Hong, Y. (2017). Phosphorylation potential of Drosophila E-Cadherin intracellular domain is essential for development and regulating adherens junction biosynthetic dynamics. Development [Epub ahead of print]. PubMed ID: 28219985

    Yuva-Aydemir, Y., Xu, X.L., Aydemir, O., Gascon, E., Sayin, S., Zhou, W., Hong, Y. and Gao, F.B. (2015). Downregulation of the host gene jigr1 by miR-92 is essential for neuroblast self-renewal in Drosophila. PLoS Genet 11: e1005264. PubMed ID: 26000445

    Shieh, B. H., Kristaponyte, I. and Hong, Y. (2014). Distinct Roles of Arrestin 1 in Photoreceptors During Drosophila Development. J Biol Chem. PubMed ID: 24838243

    Zhou, W. and Hong, Y. (2012). Drosophila Patj plays a supporting role in apical-basal polarity but is essential for viability. Development 139: 2891-2896. PubMed ID: 22791898

    Zhou, W., Huang, J., Watson, A. M. and Hong, Y. (2012). W::Neo: a novel dual-selection marker for high efficiency gene targeting in Drosophila. PLoS One 7: e31997. PubMed ID: 22348139

    Huang, J., Huang, L., Chen, Y. J., Austin, E., Devor, C. E., Roegiers, F. and Hong, Y. (2011). Differential regulation of adherens junction dynamics during apical-basal polarization. J Cell Sci 124: 4001-4013. PubMed ID: 22159415

    Huang, J., Ghosh, P., Hatfull, G. F. and Hong, Y. (2011). Successive and targeted DNA integrations in the Drosophila genome by Bxb1 and phiC31 integrases. Genetics 189: 391-395. PubMed ID: 21652525

  • Jamila Horabin Florida State University, Tallahassee,
    Cabrera, J. R., Olcese, U. and Horabin, J. I. (2015). A balancing act: heterochromatin protein 1a and the Polycomb group coordinate their levels to silence chromatin in Drosophila. Epigenetics Chromatin 8: 17. PubMed ID: 25954320

    Mulvey, B. B., Olcese, U., Cabrera, J. R. and Horabin, J. I. (2014). An interactive network of long non-coding RNAs facilitates the Drosophila sex determination decision. Biochim Biophys Acta. PubMed ID: 24954180

    Li, H., Rodriguez, J., Yoo, Y., Shareef, M. M., Badugu, R., Horabin, J. I. and Kellum, R. (2011). Cooperative and antagonistic contributions of two heterochromatin proteins to transcriptional regulation of the Drosophila sex determination decision. PLoS Genet 7: e1002122. PubMed ID: 21695246

    Kappes, G., Deshpande, G., Mulvey, B. B., Horabin, J. I. and Schedl, P. (2011). The Drosophila Myc gene, diminutive, is a positive regulator of the Sex-lethal establishment promoter, Sxl-Pe. Proc Natl Acad Sci U S A 108: 1543-1548. PubMed ID: 21220321

  • Sally Horne-Badovinac Department of Molecular Genetics and Cell Biology, University of Chicago
    Williams, A. M. and Horne-Badovinac, S. (2023). Fat2 polarizes Lar and Sema5c to coordinate the motility of collectively migrating epithelial cells. J Cell Sci. PubMed ID: 37593878

    Zajac, A. L. and Horne-Badovinac, S. (2022). Kinesin-directed secretion of basement membrane proteins to a subdomain of the basolateral surface in Drosophila epithelial cells. Curr Biol. PubMed ID: 35021047

    Dent, L. G., Manning, S. A., Kroeger, B., Williams, A. M., Saiful Hilmi, A. J., Crea, L., Kondo, S., Horne-Badovinac, S. and Harvey, K. F. (2019). The dPix-Git complex is essential to coordinate epithelial morphogenesis and regulate myosin during Drosophila egg chamber development. PLoS Genet 15(5): e1008083. PubMed ID: 31116733

    Stedden, C. G., Menegas, W., Zajac, A. L., Williams, A. M., Cheng, S., Ozkan, E. and Horne-Badovinac, S. (2019). Planar-polarized Semaphorin-5c and Plexin A promote the collective migration of epithelial cells in Drosophila. Curr Biol 29(6): 908-920.e906. PubMed ID: 30827914

    Barlan, K., Cetera, M. and Horne-Badovinac, S. (2017). Fat2 and Lar define a basally localized planar signaling system controlling collective cell migration. Dev Cell 40(5): 467-477.e465. PubMed ID: 28292425

    Isabella, A.J. and Horne-Badovinac, S. (2016). Rab10-mediated secretion synergizes with tissue movement to build a polarized basement membrane architecture for organ morphogenesis. Dev Cell 38: 47-60. PubMed ID: 27404358

    Andersen, D. and Horne-Badovinac, S. (2016). Influence of ovarian muscle contraction and oocyte growth on egg chamber elongation in Drosophila. Development [Epub ahead of print]. PubMed ID: 26952985

    Isabella, A. J. and Horne-Badovinac, S. (2015). Dynamic regulation of basement membrane protein levels promotes egg chamber elongation in Drosophila. Dev Biol. PubMed ID: 26348027

    Cetera, M. and Horne-Badovinac, S. (2015). Round and round gets you somewhere: collective cell migration and planar polarity in elongating Drosophila egg chambers. Curr Opin Genet Dev 32C: 10-15. PubMed ID: 25677931

    Cetera, M., Ramirez-San Juan, G. R., Oakes, P. W., Lewellyn, L., Fairchild, M. J., Tanentzapf, G., Gardel, M. L. and Horne-Badovinac, S. (2014). Epithelial rotation promotes the global alignment of contractile actin bundles during Drosophila egg chamber elongation. Nat Commun 5: 5511. PubMed ID: 25413675

    Horne-Badovinac, S. (2014). The Drosophila Egg Chamber - A New Spin on How Tissues Elongate. Integr Comp Biol. PubMed ID: 24920751

    Lewellyn, L., Cetera, M., Horne-Badovinac, S. (2013) Misshapen decreases integrin levels to promote epithelial motility and planar polarity in Drosophila. J Cell Biol 200: 721-729. PubMed ID: 23509067

    Lerner, D. W., McCoy, D., Isabella, A. J., Mahowald, A. P., Gerlach, G. F., Chaudhry, T. A., Horne-Badovinac, S. (2013) A Rab10-Dependent Mechanism for Polarized Basement Membrane Secretion during Organ Morphogenesis. Dev Cell 24: 159-168. PubMed ID: 23369713

  • Mia Horowitz Tel Aviv University, Israel
    Cabasso, O., Paul, S., Maor, G., Pasmanik-Chor, M., Kallemeijn, W., Aerts, J. and Horowitz, M. (2021). The Uncovered Function of the Drosophila GBA1a-Encoded Protein. Cells 10(3). PubMed ID: 33809074

    Braunstein, H., Papazian, M., Maor, G., Lukas, J., Rolfs, A. and Horowitz, M. (2020). Misfolding of Lysosomal alpha-Galactosidase a in a Fly Model and Its Alleviation by the Pharmacological Chaperone Migalastat. Int J Mol Sci 21(19). PubMed ID: 33036426

    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

  • David Hosken Biosciences, University of Exeter
    House, C. M., Lewis, Z., Sharma, M. D., Hodgson, D. J., Hunt, J., Wedell, N. and Hosken, D. J. (2021). Sexual selection on the genital lobes of male Drosophila simulans. Evolution. PubMed ID: 33386741

    Tourmente, M., Archer, C. R. and Hosken, D. J. (2019). Complex interactions between sperm viability and female fertility. Sci Rep 9(1): 15366. PubMed ID: 31653962

    Archer, C. R., Recker, M., Duffy, E. and Hosken, D. J. (2018). Intralocus sexual conflict can resolve the male-female health-survival paradox. Nat Commun 9(1): 5048. PubMed ID: 30487539

    Archer, C. R., Stephens, R. M., Sharma, M. D. and Hosken, D. J. (2017). The Drosophila simulans Y chromosome interacts with the autosomes to influence male fitness. J Evol Biol [Epub ahead of print]. PubMed ID: 28703322

    Maraqa, M. S., Griffin, R., Sharma, M. D., Wilson, A. J., Hunt, J., Hosken, D. J. and House, C. M. (2016). Constrained evolution of the sex comb in Drosophila simulans. J Evol Biol [Epub ahead of print]. PubMed ID: 27859860

    Rostant, W. G., Kay, C., Wedell, N. and Hosken, D. J. (2015). Sexual conflict maintains variation at an insecticide resistance locus. BMC Biol 13: 34. PubMed ID: 26032845

    House, C. M., Lewis, Z., Hodgson, D. J., Wedell, N., Sharma, M. D., Hunt, J., Hosken, D. J. (2013) Sexual and natural selection both influence male genital evolution. PLoS One 8: e63807. PubMed ID: 23717488

    Ingleby, F. C., Hosken, D. J., Flowers, K., Hawkes, M. F., Lane, S. M., Rapkin, J., Dworkin, I. and Hunt, J. (2013). Genotype-by-environment interactions for cuticular hydrocarbon expression in Drosophila simulans. J Evol Biol 26: 94-107. PubMed ID: 23163514

    Sharma, M. D., Hunt, J. and Hosken, D. J. (2012). Antagonistic responses to natural and sexual selection and the sex-specific evolution of cuticular hydrocarbons in Drosophila simulans. Evolution 66: 665-677. PubMed ID: 22380431

    Sharma, M. D., Mitchell, C., Hunt, J., Tregenza, T. and Hosken, D. J. (2012). The genetics of cuticular hydrocarbon profiles in the Fruit Fly Drosophila simulans. J Hered 103: 230-239. PubMed ID: 22268163

  • Steve X. Hou NIH
    Aggarwal, P., Liu, Z., Cheng, G. Q., Singh, S. R., Shi, C., Chen, Y., Sun, L. V. and Hou, S. X. (2022). Disruption of the lipolysis pathway results in stem cell death through a sterile immunity-like pathway in adult Drosophila. Cell Rep 39(12): 110958. PubMed ID: 35732115Singh, S.R., Zeng, X., Zhao, J., Liu, Y., Hou, G., Liu, H. and Hou, S.X. (2016). The lipolysis pathway sustains normal and transformed stem cells in adult

    Drosophila. Nature [Epub ahead of print]. PubMed ID: 27680705

    Liu, Y., Ge, Q., Chan, B., Liu, H., Singh, S. R., Manley, J., Lee, J., Weideman, A. M., Hou, G. and Hou, S. X. (2016). Whole-animal genome-wide RNAi screen identifies networks regulating male germline stem cells in Drosophila. Nat Commun 7: 12149. PubMed ID: 27484291

    Singh, S. R., Liu, Y., Zhao, J., Zeng, X. and Hou, S. X. (2016). The novel tumour suppressor Madm regulates stem cell competition in the Drosophila testis. Nat Commun 7: 10473. PubMed ID: 26792023

    Liu, Y., Singh, S. R., Zeng, X., Zhao, J. and Hou, S. X. (2015). The nuclear matrix protein Megator regulates stem cell asymmetric division through the mitotic checkpoint complex in Drosophila testes. PLoS Genet 11: e1005750. PubMed ID: 26714316

    Zeng, X., Han, L., Singh, S. R., Liu, H., Neumuller, R. A., Yan, D., Hu, Y., Liu, Y., Liu, W., Lin, X. and Hou, S. X. (2015). Genome-wide RNAi Screen Identifies Networks Involved in Intestinal Stem Cell Regulation in Drosophila. Cell Rep 10: 1226-1238. PubMed ID: 25704823

    Zeng, X., Lin, X. and Hou, S. X. (2013). The Osa-containing SWI/SNF chromatin-remodeling complex regulates stem cell commitment in the adult Drosophila intestine. Development 140: 3532-3540. PubMed ID: 23942514

    Zeng, X. and Hou, S. X. (2012). Broad relays hormone signals to regulate stem cell differentiation in Drosophila midgut during metamorphosis. Development 139: 3917-3925. PubMed ID: 23048182

    Singh, S. R., Zeng, X., Zheng, Z. and Hou, S. X. (2011). The adult Drosophila gastric and stomach organs are maintained by a multipotent stem cell pool at the foregut/midgut junction in the cardia (proventriculus). Cell Cycle 10: 1109-1120. PubMed ID: 21403464

    Zeng, X., Chauhan, C. and Hou, S. X. (2010). Characterization of midgut stem cell- and enteroblast-specific Gal4 lines in drosophila. Genesis 48: 607-611. PubMed ID: 20681020

  • David Houle Department of Biology, Florida State University, Tallahassee
    Sztepanacz, J. L. and Houle, D. (2021). Allometry constrains the evolution of sexual dimorphism in Drosophila across 33 million years of divergence. Evolution. PubMed ID: 33638384

    Sztepanacz, J. L. and Houle, D. (2019). Cross-sex genetic covariances limit the evolvability of wing-shape within and among species of Drosophila. Evolution. PubMed ID: 31206655

    Pitchers, W., Nye, J., Marquez, E. J., Kowalski, A., Dworkin, I. and Houle, D. (2019). A Multivariate Genome-Wide Association Study of Wing Shape in Drosophila melanogaster. Genetics 211(4): 1429-1447. PubMed ID: 30792267

    Matamoro-Vidal, A., Huang, Y., Salazar-Ciudad, I., Shimmi, O. and Houle, D. (2018). Quantitative Morphological Variation in the Developing Drosophila Wing. G3 (Bethesda) 8(7): 2399-2409. PubMed ID: 29844017

    Hollis, B., Houle, D. and Kawecki, T. J. (2016). Evolution of reduced post-copulatory molecular interactions in Drosophila populations lacking sperm competition. J Evol Biol 29(1): 77-85. PubMed ID: 26395588

  • Bernhard Hovemann Ruhr-Universität Bochum, Fakultät für Chemie und Biochemie
    Hartwig, S., Dovengerds, C., Herrmann, C. and Hovemann, B. T. (2014). Drosophila Ebony: a novel type of nonribosomal peptide synthetase related enzyme with unusually fast peptide bond formation kinetics. FEBS J. PubMed ID: 25229196

    Sun, K., Westholm, J. O., Tsurudome, K., Hagen, J. W., Lu, Y., Kohwi, M., Betel, D., Gao, F. B., Haghighi, A. P., Doe, C. Q. and Lai, E. C. (2012). Neurophysiological defects and neuronal gene deregulation in Drosophila mir-124 mutants. PLoS Genet 8: e1002515. PubMed ID: 22347817

    Ziegler, A. B., Brusselbach, F. and Hovemann, B. T. (2013) Activity and coexpression of Drosophila black with ebony in fly optic lobes reveals putative cooperative tasks in vision that evade electroretinographic detection. J Comp Neurol 521: Spc1. PubMed ID: 23426638

    Efetova, M., Petereit, L., Rosiewicz, K., Overend, G., Haussig, F., Hovemann, B. T., Cabrero, P., JA, T. D. and Schwarzel, M. (2012). Separate roles of PKA and EPAC in renal function unraveled by the optogenetic control of cAMP levels in vivo. J Cell Sci. PubMed ID: 23264735

    Aust, S., Brusselbach, F., Putz, S. and Hovemann, B. T. (2010). Alternative tasks of Drosophila tan in neurotransmitter recycling versus cuticle sclerotization disclosed by kinetic properties. J Biol Chem 285: 20740-20747. PubMed ID: 20439462

  • Hwei-Jan Hsu Academia Sinica, Taiwan
    Chen, T. A., Lin, K. Y., Yang, S. M., Tseng, C. Y., Wang, Y. T., Lin, C. H., Luo, L., Cai, Y. and Hsu, H. J. (2022). Canonical Wnt Signaling Promotes Formation of Somatic Permeability Barrier for Proper Germ Cell Differentiation. Front Cell Dev Biol 10: 877047. PubMed ID: 35517512

    Lin, K. Y., Wang, W. D., Lin, C. H., Rastegari, E., Su, Y. H., Chang, Y. T., Liao, Y. F., Chang, Y. C., Pi, H., Yu, B. Y., Chen, S. H., Lin, C. Y., Lu, M. Y., Su, T. Y., Tzou, F. Y., Chan, C. C. and Hsu, H. J. (2020). Piwi reduction in the aged niche eliminates germline stem cells via Toll-GSK3 signaling. Nat Commun 11(1): 3147. PubMed ID: 32561720

    Rastegari, E., Kajal, K., Tan, B. S., Huang, F., Chen, R. H., Hsieh, T. S. and Hsu, H. J. (2020). WD40 protein Wuho controls germline homeostasis via TRIM-NHL tumor suppressor Mei-p26 in Drosophila. Development 147(2). PubMed ID: 31941704

    Ke, Y. T. and Hsu, H. J. (2019). Generation of inducible gene-switched GAL4 expressed in the Drosophila female germline stem cell niche. G3 (Bethesda). PubMed ID: 31018943

    Tseng, C. Y., Su, Y. H., Yang, S. M., Lin, K. Y., Lai, C. M., Rastegari, E., Amartuvshin, O., Cho, Y., Cai, Y. and Hsu, H. J. (2018). Smad-independent BMP signaling in somatic cells limits the size of the germline stem cell pool. Stem Cell Reports. PubMed ID: 30122445

    Cho, Y., Lai, C. M., Lin, K. Y. and Hsu, H. J. (2018). A targeted RNAi screen reveals Drosophila female-sterile genes That control the size of germline stem cell niche during development. G3 (Bethesda). PubMed ID: 29764959

    Su, Y. H., Rastegri, E., Kao, S. H., Lai, C. M., Lin, K. Y., Liao, H. Y., Wang, M. H. and Hsu, H. J. (2018). Diet regulates membrane extension and survival of niche escort cells for germline homeostasis via insulin signaling. Development. PubMed ID: 29549109

    Tseng, C. Y. and Hsu, H. J. (2017). Decreased expression of lethal giant larvae causes ovarian follicle cell outgrowth in the Drosophila Scutoid mutant. PLoS One 12(12): e0188917. PubMed ID: 29261681

    Lai, C. M., Lin, K. Y., Kao, S. H., Chen, Y. N., Huang, F. and Hsu, H. J. (2017). Hedgehog signaling establishes precursors for germline stem cell niches by regulating cell adhesion. J Cell Biol. PubMed ID: 28363970

    Tseng, C. Y., Kao, S. H. and Hsu, H. J. (2016). Snail controls proliferation of Drosophila ovarian epithelial follicle stem cells, independently of E-cadherin. Dev Biol 414(2): 142-148. PubMed ID: 27141871

    Hsu, H. J. and Drummond-Barbosa, D. (2017). A visual screen for diet-regulated proteins in the Drosophila ovary using GFP protein trap lines. Gene Expr Patterns 23-24: 13-21. PubMed ID: 28093350

  • Tien Hsu Boston University School of Medicine
    Ignesti, M., Barraco, M., Nallamothu, G., Woolworth, J. A., Duchi, S., Gargiulo, G., Cavaliere, V. and Hsu, T. (2014). Notch signaling during development requires the function of awd, the Drosophila homolog of human metastasis suppressor gene Nm23. BMC Biol 12: 12. PubMed ID: 24528630

    Hsouna, A., Nallamothu, G., Kose, N., Guinea, M., Dammai, V. and Hsu, T. (2010). Drosophila von Hippel-Lindau tumor suppressor gene function in epithelial tubule morphogenesis. Mol Cell Biol 30: 3779-3794. PubMed ID: 20516215

    Duchi, S., Fagnocchi, L., Cavaliere, V., Hsouna, A., Gargiulo, G. and Hsu, T. (2010). Drosophila VHL tumor-suppressor gene regulates epithelial morphogenesis by promoting microtubule and aPKC stability. Development 137: 1493-1503. PubMed ID: 20388653

    Woolworth, J. A., Nallamothu, G. and Hsu, T. (2009). The Drosophila metastasis suppressor gene Nm23 homolog, awd, regulates epithelial integrity during oogenesis. Mol Cell Biol 29: 4679-4690. PubMed ID: 19581292

  • Fu-De Huang Shanghai Institutes for Biological Sciences
    Zhang, X., Wang, W. A., Jiang, L. X., Liu, H. Y., Zhang, B. Z., Lim, N., Li, Q. Y. and F. D. (2017). Downregulation of RBO-PI4KIIIalpha Facilitates Abeta42 Secretion and Ameliorates Neural Deficits in Abeta42-Expressing Drosophila. J Neurosci 37(19): 4928-4941. PubMed ID: 28424219

    Liu, H., Han, M., Li, Q., Zhang, X., Wang, W. A. and Huang, F. D. (2015). Automated rapid iterative negative geotaxis assay and its use in a genetic screen for modifiers of Abeta(42)-induced locomotor decline in Drosophila. Neurosci Bull 31(5): 541-549. PubMed ID: 26077703

    Han, M., Huang, J. K., Liu, H. Y., Wang, W. A., Sun, X. J. and Huang, F. D. (2015). Neither insufficiency nor overexpression of sac1 affects the accumulation of Abeta42 in Drosophila expressing Ab42. Eur Rev Med Pharmacol Sci 19(9): 1700-1705. PubMed ID: 26004612

  • Xun Huang Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing
    Xu, M., Ding, L., Liang, J., Yang, X., Liu, Y., Wang, Y., Ding, M. and Huang, X. (2021). NAD kinase sustains lipogenesis and mitochondrial metabolism through fatty acid synthesis. Cell Rep 37(13): 110157. PubMed ID: 34965438

    Yang, X., Liang, J., Ding, L., Li, X., Lam, S. M., Shui, G., Ding, M. and Huang, X. (2019). Phosphatidylserine synthetase regulates cellular homeostasis through distinct metabolic mechanisms. PLoS Genet 15(12): e1008548. PubMed ID: 31869331

    Wang, W., Xin, J., Yang, X., Lam, S. M., Shui, G., Wang, Y. and Huang, X. (2018). Lipid-gene regulatory network reveals coregulations of triacylglycerol with phosphatidylinositol/lysophosphatidylinositol and with hexosyl-ceramide. Biochim Biophys Acta Mol Cell Biol Lipids 1864(2): 168-180. PubMed ID: 30521938

    Yao, Y., Li, X., Wang, W., Liu, Z., Chen, J., Ding, M. and Huang, X. (2018). MRT, functioning with NURF complex, regulates lipid droplet size. Cell Rep 24(11): 2972-2984. PubMed ID: 30208321

    Ding, L., Yang, X., Tian, H., Liang, J., Zhang, F., Wang, G., Wang, Y., Ding, M., Shui, G. and Huang, X. (2018). Seipin regulates lipid homeostasis by ensuring calcium-dependent mitochondrial metabolism. Embo J. PubMed ID: 30049710

    Fan, W., Lam, S. M., Xin, J., Yang, X., Liu, Z., Liu, Y., Wang, Y., Shui, G. and Huang, X. (2017). Drosophila TRF2 and TAF9 regulate lipid droplet size and phospholipid fatty acid composition. PLoS Genet 13(3): e1006664. PubMed ID: 28273089

    Bi, J., Xiang, Y., Chen, H., Liu, Z., Gronke, S., Kuhnlein, R. P. and Huang, X. (2012). Opposite and redundant roles of the two Drosophila perilipins in lipid mobilization. J Cell Sci 125: 3568-3577. PubMed ID: 22505614

    Wang, C., Liu, Z. and Huang, X. (2012). Rab32 is important for autophagy and lipid storage in Drosophila. PLoS One 7: e32086. PubMed ID: 22348149

    Ma, Z., Liu, Z. and Huang, X. (2012). Membrane phospholipid asymmetry counters the adverse effects of sterol overloading in the Golgi membrane of Drosophila. Genetics 190: 1299-1308. PubMed ID: 22234859

    Tian, Y., Bi, J., Shui, G., Liu, Z., Xiang, Y., Liu, Y., Wenk, M. R., Yang, H. and Huang, X. (2011). Tissue-autonomous function of Drosophila seipin in preventing ectopic lipid droplet formation. PLoS Genet 7: e1001364. PubMed ID: 21533227

  • Armin Huber Zentrum für Neurosensorik -, Institut für Physiologie, Tübingen University
    Wagner, K., Smylla, T. K., Zeger, M. and Huber, A. (2022). Studying Membrane Protein Trafficking in Drosophila Photoreceptor Cells Using eGFP-Tagged Proteins. J Vis Exp(179). PubMed ID: 35129173 Wagner, K., Smylla, T. K., Lampe, M., Krieg, J. and Huber, A. (2021). Phospholipase D and retromer promote recycling of TRPL ion channel via the endoplasmic reticulum. Traffic. PubMed ID: 34719094

    Voolstra, O., Strauch, L., Mayer, M. and Huber, A. (2018). Functional characterization of the three Drosophila retinal degeneration C (RDGC) protein phosphatase isoforms. PLoS One 13(9): e0204933. PubMed ID: 30265717

    Katz, B., Voolstra, O., Tzadok, H., Yasin, B., Rhodes-Modrov, E., Bartels, J. P., Strauch, L., Huber, A. and Minke, B. (2017). The latency of the light response is modulated by the phosphorylation state of Drosophila TRP at a specific site. Channels (Austin): [Epub ahead of print]. PubMed ID: 28762890

    Voolstra, O., Rhodes-Mordov, E., Katz, B., Bartels, J. P., Oberegelsbacher, C., Schotthofer, S. K., Yasin, B., Tzadok, H., Huber, A. and Minke, B. (2017). The phosphorylation state of the Drosophila TRP channel modulates the frequency response to oscillating light in vivo. J Neurosci 37(15): 4213-4224. PubMed ID: 28314815

    Schopf, K. and Huber, A. (2016). Membrane protein trafficking in Drosophila photoreceptor cells. Eur J Cell Biol. PubMed ID: 27964885

    Paulsen, R., Bahner, M. and Huber, A. (2000). The PDZ assembled "transducisome" of microvillar photoreceptors: the TRP/TRPL problem. Pflugers Arch 439: R181-R183. PubMed ID: 27757613

    Cerny, A. C., Altendorfer, A., Schopf, K., Baltner, K., Maag, N., Sehn, E., Wolfrum, U. and Huber, A. (2015). The GTP- and Phospholipid-Binding Protein TTD14 Regulates Trafficking of the TRPL Ion Channel in Drosophila Photoreceptor Cells. PLoS Genet 11: e1005578. PubMed ID: 26509977

    Voolstra, O., Spat, P., Oberegelsbacher, C., Claussen, B., Pfannstiel, J. and Huber, A. (2015). Light-dependent phosphorylation of the Drosophila Inactivation No Afterpotential D (INAD) scaffolding protein at Thr170 and Ser174 by eye-specific Protein kinase C. PLoS One 10: e0122039. PubMed ID: 25799587

    Katz, B., Oberacker, T., Richter, D., Tzadok, H., Peters, M., Minke, B., Huber, A. (2013) The Drosophila TRP and TRPL are assembled as homomultimeric channels in vivo. J Cell Sci. PubMed ID: 23687378

  • Wolf Huetteroth Genetik, University of Leipzig, Germany
    Mahishi, D., Triphan, T., Hesse, R. and Huetteroth, W. (2021). The Panopticon-Assessing the Effect of Starvation on Prolonged Fly Activity and Place Preference. Front Behav Neurosci 15: 640146. PubMed ID: 33841109

    Lerner, H., Rozenfeld, E., Rozenman, B., Huetteroth, W. and Parnas, M. (2020). Differential Role for a Defined Lateral Horn Neuron Subset in Naive Odor Valence in Drosophila. Sci Rep 10(1): 6147. PubMed ID: 32273557

    Mahishi, D. and Huetteroth, W. (2019). The prandial process in flies. Curr Opin Insect Sci 36: 157-166. PubMed ID: 31765996

    Nassel, D. R., Pauls, D. and Huetteroth, W. (2019). Neuropeptides in modulation of Drosophila behavior: how to get a grip on their pleiotropic actions. Curr Opin Insect Sci 36: 1-8. PubMed ID: 31280184

  • Sarah Hughes Department of Medical Genetics, University of Alberta
    Keegan, S. E., Haskins, J., Simmonds, A. J. and Hughes, S. C. (2023). A chromatin remodelling SWI/SNF subunit, Snr1, regulates neural stem cell determination and differentiation. Development 150(13). PubMed ID: 37294080

    Keegan, S. E. and Hughes, S. C. (2020). Role of nuclear-cytoplasmic protein localization during Drosophila neuroblast development. Genome. PubMed ID: 32526151

    Abeysundara, N., Simmonds, A. J. and Hughes, S. C. (2017). Moesin is involved in polarity maintenance and cortical remodelling during asymmetric cell division. Mol Biol Cell [Epub ahead of print]. PubMed ID: 29282284

    Germain, D. R., Li, L., Hildebrandt, M. R., Simmonds, A. J., Hughes, S. C. and Godbout, R. (2015). Loss of the Drosophila melanogaster DEAD box protein Ddx1 leads to reduced size and aberrant gametogenesis. Dev Biol 407(2): 232-245. PubMed ID: 26433063

    Abeysundara, N., Leung, A. C., Primrose, D. A. and Hughes, S. C. (2014). Regulation of cell proliferation and adhesion by means of a novel region of drosophila merlin interacting with Sip1. Dev Dyn 243(12): 1554-1570. PubMed ID: 25204795

    Li, X., Zhuo, R., Tiong, S., Di Cara, F., King-Jones, K., Hughes, S. C., Campbell, S. D. and Wevrick, R. (2013). The Smc5/Smc6/MAGE complex confers resistance to caffeine and genotoxic stress in Drosophila melanogaster. PLoS One 8(3): e59866. PubMed ID: 23555814

  • Dan Hultmark Department of Molecular Biology, Umea University
    Yang, H. and Hultmark, D. (2017). Drosophila muscles regulate the immune response against wasp infection via carbohydrate metabolism. Sci Rep 7(1): 15713. PubMed ID: 29146985

    Schmid, M. R., Anderl, I., Vo, H. T., Valanne, S., Yang, H., Kronhamn, J., Ramet, M., Rusten, T. E. and Hultmark, D. (2016). Genetic screen in Drosophila larvae links ird1 function to Toll signaling in the fat body and hemocyte motility. PLoS One 11: e0159473. PubMed ID: 27467079

    Anderl, I., Vesala, L., Ihalainen, T.O., Vanha-Aho, L.M., Andó, I., Rämet, M. and Hultmark, D. (2016). Transdifferentiation and proliferation in two distinct hemocyte lineages in Drosophila melanogaster larvae after wasp infection. PLoS Pathog 12: e1005746. PubMed ID: 27414410

    Ekstrom, J. O. and Hultmark, D. (2016). A novel strategy for live detection of viral infection in Drosophila melanogaster. Sci Rep 6: 26250. PubMed ID: 27189868

    Yang, H. and Hultmark, D. (2016). Tissue communication in a systemic immune response of Drosophila. Fly (Austin): [Epub ahead of print] PubMed ID: 27116253

    Yang, H., Kronhamn, J., Ekström, J.O., Korkut, G.G. and Hultmark, D. (2015). JAK/STAT signaling in Drosophila muscles controls the cellular immune response against parasitoid infection. EMBO Rep [Epub ahead of print]. PubMed ID: 26412855

    Vanha-Aho, L.M., Anderl, I., Vesala, L., Hultmark, D., Valanne, S and Rämet, M. (2015). Edin expression in the fat body is required in the defense against parasitic wasps in Drosophila melanogaster PLoS Pathog 11: e1004895. PubMed ID: 25965263

    Schmid, M. R., Anderl, I., Vesala, L., Vanha-Aho, L. M., Deng, X. J., Ramet, M. and Hultmark, D. (2014). Control of Drosophila Blood Cell Activation via Toll Signaling in the Fat Body. PLoS One 9: e102568. PubMed ID: 25102059

    Ramet, M. and Hultmark, D. (2014). Drosophila immunity--glorious past, dynamic present and exciting future. Dev Comp Immunol 42: 1-2. PubMed ID: 23891875

    van Mierlo, J. T., Bronkhorst, A. W., Overheul, G. J., Sadanandan, S. A., Ekstrom, J. O., Heestermans, M., Hultmark, D., Antoniewski, C. and van Rij, R. P. (2012). Convergent evolution of argonaute-2 slicer antagonism in two distinct insect RNA viruses. PLoS Pathog 8: e1002872. PubMed ID: 22916019

  • Thomas Hummel Department for Neurobiology, Universität Wien
    Timaeus, L., Geid, L., Sancer, G., Wernet, M. F. and Hummel, T. (2020). Parallel Visual Pathways with Topographic versus Nontopographic Organization Connect the Drosophila Eyes to the Central Brain. iScience 23(10): 101590. PubMed ID: 33205011

    Kaur, R., Surala, M., Hoger, S., Grossmann, N., Grimm, A., Timaeus, L., Kallina, W. and Hummel, T. (2019). Pioneer interneurons instruct bilaterality in the Drosophila olfactory sensory map. Sci Adv 5(10): eaaw5537. PubMed ID: 31681838

    Goyal, G., Zierau, A., Lattemann, M., Bergkirchner, B., Javorski, D., Kaur, R. and Hummel, T. (2019). Inter-axonal recognition organizes Drosophila olfactory map formation. Sci Rep 9(1): 11554. PubMed ID: 31399611

    Kulkarni, A., Ertekin, D., Lee, C. H. and Hummel, T. (2016). Birth order dependent growth cone segregation determines synaptic layer identity in the visual system. Elife 5. PubMed ID: 26987017

    Jafari, S., Alkhori, L., Schleiffer, A., Brochtrup, A., Hummel, T. and Alenius, M. (2012). Combinatorial activation and repression by seven transcription factors specify Drosophila odorant receptor expression. PLoS Biol 10: e1001280. PubMed ID: 22427741

    Brochtrup, A. and Hummel, T. (2011). Olfactory map formation in the Drosophila brain: genetic specificity and neuronal variability. Curr Opin Neurobiol 21: 85-92. PubMed ID: 21112768

  • Deb Hursh FDA, Bethesda
    Dai, Z., Li, D., Du, X., Ge, Y., Hursh, D. A. and Bi, X. (2020). Drosophila Caliban preserves intestinal homeostasis and lifespan through regulating mitochondrial dynamics and redox state in enterocytes. PLoS Genet 16(10): e1009140. PubMed ID: 33057338

    Thomas, J.T., Eric Dollins, D., Andrykovich, K.R., Chu, T., Stultz, B.G., Hursh, D.A. and Moos, M. (2017). SMOC can act as both an antagonist and an expander of BMP signaling. Elife 6. PubMed ID: 28323621

    Hursh, D. A., Stultz, B. G. and Park, S. Y. (2016). Jun N-terminal Kinase signaling makes a face. Fly (Austin): [Epub ahead of print]. PubMed ID: 27384866

    Park, S. Y., Stultz, B. G. and Hursh, D. A. (2015). Dual role of Jun N-terminal kinase activity in bone morphogenetic protein-mediated Drosophila ventral head development. Genetics [Epub ahead of print]. PubMed ID: 26500262

    Stultz, B. G., Park, S. Y., Mortin, M. A., Kennison, J. A. and Hursh, D. A. (2012). Hox proteins coordinate peripodial decapentaplegic expression to direct adult head morphogenesis in Drosophila. Dev Biol 369: 362-376. PubMed ID: 22824425

    Sen, A., Stultz, B. G., Lee, H. and Hursh, D. A. (2010). Odd paired transcriptional activation of decapentaplegic in the Drosophila eye/antennal disc is cell autonomous but indirect. Dev Biol 343: 167-177. PubMed ID: 20403347

  • Stephan Hutter Evolutionary Biology, Ludwig Maxmillians-Universität München
    Croze, M., Wollstein, A., Bozicevic, V., Zivkovic, D., Stephan, W. and Hutter, S. (2017). A genome-wide scan for genes under balancing selection in Drosophila melanogaster. BMC Evol Biol 17(1): 15. PubMed ID: 28086750

    von Heckel, K., Stephan, W. and Hutter, S. (2016). Canalization of gene expression is a major signature of regulatory cold adaptation in temperate Drosophila melanogaster. BMC Genomics 17: 574. PubMed ID: 27502401

    Croze, M., Zivkovic, D., Stephan, W. and Hutter, S. (2016). Balancing selection on immunity genes: review of the current literature and new analysis in Drosophila melanogaster. Zoology (Jena). PubMed ID: 27106015

    Bozicevic, V., Hutter, S., Stephan, W. and Wollstein, A. (2016). Population genetic evidence for cold adaptation in European Drosophila melanogaster populations. Mol Ecol 25: 1175-1191. PubMed ID: 26558479

    Paparazzo, F., Tellier, A., Stephan, W. and Hutter, S. (2015). Survival Rate and Transcriptional Response upon Infection with the Generalist Parasite Beauveria bassiana in a World-Wide Sample of Drosophila melanogaster. PLoS One 10: e0132129. PubMed ID: 26154519

  • Jean-René Huynh Genetique et biologie du developpement, Institut Curie
    Mathieu, J., Michel-Hissier, P., Boucherit, V. and Huynh, J. R. (2022). The deubiquitinase USP8 targets ESCRT-III to promote incomplete cell division. Science 376(6595): 818-823. PubMed ID: 35587967

    Rubin, T., Macaisne, N., Valles, A. M., Guilleman, C., Gaugue, I., Dal Toe, L. and Huynh, J. R. (2022). Premeiotic pairing of homologous chromosomes during Drosophila male meiosis. Proc Natl Acad Sci U S A 119(47): e2207660119. PubMed ID: 36375065

    Molla-Herman, A., Angelova, M. T., Ginestet, M., Carre, C., Antoniewski, C. and Huynh, J. R. (2020). tRNA Fragments Populations Analysis in Mutants Affecting tRNAs Processing and tRNA Methylation. Front Genet 11: 518949. PubMed ID: 33193603

    Christophorou, N., Rubin, T., Bonnet, I., Piolot, T., Arnaud, M. and Huynh, J. R. (2015). Microtubule-driven nuclear rotations promote meiotic chromosome dynamics. Nat Cell Biol 17: 1388-1400. PubMed ID: 26458247

    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

    Christophorou, N., Rubin, T., Bonnet, I., Piolot, T., Arnaud, M. and Huynh, J.R. (2015). Microtubule-driven nuclear rotations promote meiotic chromosome dynamics. Nat Cell Biol [Epub ahead of print]. PubMed ID: 26458247

    Christophorou, N., Rubin, T. and Huynh, J. R. (2013). Synaptonemal Complex Components Promote Centromere Pairing in Pre-meiotic Germ Cells. PLoS Genet 9: e1004012. PubMed ID: 24367278

    Mathieu, J., Cauvin, C., Moch, C., Radford, S. J., Sampaio, P., Perdigoto, C. N., Schweisguth, F., Bardin, A. J., Sunkel, C. E., McKim, K., Echard, A. and Huynh, J. R. (2013). Aurora B and cyclin B have opposite effects on the timing of cytokinesis abscission in Drosophila germ cells and in vertebrate somatic cells. Dev Cell 26: 250-265. PubMed ID: 23948252

    Jagut, M., Mihaila-Bodart, L., Molla-Herman, A., Alin, M. F., Lepesant, J. A., Huynh, J. R. (2013) A mosaic genetic screen for genes involved in the early steps of Drosophila oogenesis. G3 (Bethesda) 3: 409-425. Pubmed ID: 23450845

    Fichelson, P., Jagut, M., Lepanse, S., Lepesant, J. A. and Huynh, J. R. (2010). lethal giant larvae is required with the par genes for the early polarization of the Drosophila oocyte. Development 137: 815-824. PubMed ID: 20147382

  • Seogang Hyun School of Biological Sciences, Chung-Ang University, Seoul
    Lee, J., Yun, H. M., Han, G., Lee, G. J., Jeon, C. O. and Hyun, S. (2022). A bacteria-regulated gut peptide determines host dependence on specific bacteria to support host juvenile development and survival. BMC Biol 20(1): 258. PubMed ID: 36397042

    Lee, G. J., Han, G., Yun, H. M., Lim, J. J., Noh, S., Lee, J. and Hyun, S. (2018). Steroid signaling mediates nutritional regulation of juvenile body growth via IGF-binding protein in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 29784791

    Han, G., Lee, H. J., Jeong, S. E., Jeon, C. O. and Hyun, S. (2017). Comparative analysis of Drosophila melanogaster gut microbiota with respect to host strain, sex, and age. Microb Ecol [Epub ahead of print]. PubMed ID: 28054304

    Lee, G. J., Jun, J. W. and Hyun, S. (2014). MicroRNA miR-8 regulates multiple growth factor hormones produced from Drosophila fat cells. Insect Mol Biol. PubMed ID: 25492518

    Lee, G. J. and Hyun, S. (2014). Multiple targets of the microRNA miR-8 contribute to immune homeostasis in Drosophila. Dev Comp Immunol. PubMed ID: 24694685

    Jin, H., Kim, V. N. and Hyun, S. (2012). Conserved microRNA miR-8 controls body size in response to steroid signaling in Drosophila. Genes Dev 26: 1427-1432. PubMed ID: 22751499

    Choi, I. K. and Hyun, S. (2012). Conserved microRNA miR-8 in fat body regulates innate immune homeostasis in Drosophila. Dev Comp Immunol 37: 50-54. PubMed ID: 22210547

    Bang, S., Hyun, S., Hong, S. T., Kang, J., Jeong, K., Park, J. J., Choe, J. and Chung, J. (2011). Dopamine signalling in mushroom bodies regulates temperature-preference behaviour in Drosophila. PLoS Genet 7: e1001346. PubMed ID: 21455291


  • Return to The Interactive Fly