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


  • Thomas Raabe MSZ Universität Würzburg
    Putz, S. M., Kram, J., Rauh, E., Kaiser, S., Toews, R., Lueningschroer-Wang, Y., Rieger, D. and Raabe, T. (2021). Loss of p21-activated kinase Mbt/PAK4 causes Parkinson-like phenotypes in Drosophila. Dis Model Mech 14(6). PubMed ID: 34125184

    Dapergola, E., Menegazzi, P., Raabe, T. and Hovhanyan, A. (2021). Light Stimuli and Circadian Clock Affect Neural Development in Drosophila melanogaster. Front Cell Dev Biol 9: 595754. PubMed ID: 33763414

    Beck, K., Hovhanyan, A., Menegazzi, P., Helfrich-Forster, C. and Raabe, T. (2018). Drosophila RSK influences the pace of the circadian clock by negative regulation of protein kinase Shaggy activity. Front Mol Neurosci 11: 122. PubMed ID: 29706866

    Blumröder, R., Glunz, A., Dunkelberger, B.S., Serway, C.N., Berger, C., Mentzel, B., de Belle, J.S. and Raabe, T. (2016). Mcm3 replicative helicase mutation impairs neuroblast proliferation and memory in Drosophila. Genes Brain Behav [Epub ahead of print]. PubMed ID: 27283469

    Cate, S., Gajendra, S., Alsbury, S., Raabe, T., Tear, G. and Mitchell, K.J. (2016). Mushroom body defect is required in parallel to Netrin for midline axon guidance in Drosophila. Development [Epub ahead of print]. PubMed ID: 26893348

    Beck, K., Ehmann, N., Andlauer, T.F., Ljaschenko, D., Strecker, K., Fischer, M., Kittel, R.J. and Raabe, T. (2015). Loss of the Coffin-Lowry syndrome associated gene RSK2 alters ERK activity, synaptic function and axonal transport in Drosophilam motoneurons. Dis Model Mech [Epub ahead of print]. PubMed ID: 26398944

    Herter, E. K., Stauch, M., Gallant, M., Wolf, E., Raabe, T. and Gallant, P. (2015). snoRNAs are a novel class of biologically relevant Myc targets. BMC Biol 13: 25. PubMed ID: 25888729

    Hovhanyan, A., Herter, E. K., Pfannstiel, J., Gallant, P. and Raabe, T. (2014). Drosophila Mbm is a Nucleolar Myc and CK2 Target Required for Ribosome Biogenesis and Cell Growth of Central Brain Neuroblasts. Mol Cell Biol[Epub ahead of print] . PubMed ID: 24615015

    Szabo, A., Papin, C., Zorn, D., Ponien, P., Weber, F., Raabe, T. and Rouyer, F. (2013). The CK2 Kinase Stabilizes CLOCK and Represses Its Activity in the Drosophila Circadian Oscillator. PLoS Biol 11: e1001645. PubMed ID: 24013921

  • Leonard Rabinow CNRS UMR 8195, Université Paris Sud, Orsay
    Sun, X., Yang, H., Sturgill, D., Oliver, B., Rabinow, L. and Samson, M. L. (2015). Sxl-dependent, tra/tra2-independent alternative splicing of the Drosophila melanogaster X-Linked gene found in neurons. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26511498

    Fan, Y., Gittis, A. H., Juge, F., Qiu, C., Xu, Y. Z. and Rabinow, L. (2014). Multifunctional RNA Processing Protein SRm160 Induces Apoptosis and Regulates Eye and Genital Development in Drosophila. Genetics. PubMed ID: 24907259

    Tchankouo-Nguetcheu, S., Udinotti, M., Durand, M., Meng, T. C., Taouis, M. and Rabinow, L. (2014). Negative regulation of MAP kinase signaling in Drosophila by Ptp61F/PTP1B. Mol Genet Genomics. PubMed ID: 24752400

    Samson, M. L. and Rabinow, L. (2013). Transcriptomic Analysis of Sexual Differentiation in Somatic Tissues of Drosophila melanogaster: Successes and Caveats. Sex Dev. PubMed ID: 24356334

    Sturgill, D., Malone, J. H., Sun, X., Smith, H. E., Rabinow, L., Samson, M. L. and Oliver, B. (2013). Design of RNA splicing analysis null models for post hoc filtering of Drosophila head RNA-Seq data with the splicing analysis kit (Spanki). BMC Bioinformatics 14: 320. PubMed ID: 24209455

    Zanini, D., Jallon, J. M., Rabinow, L. and Samson, M. L. (2012). Deletion of the Drosophila neuronal gene found in neurons disrupts brain anatomy and male courtship. Genes Brain Behav 11: 819-827. PubMed ID: 22741816

  • Catherine Rabouille Hubrecht Institute, University Medical Center Utrecht
    Zhang, C., van Leeuwen, W., Blotenburg, M., Aguilera-Gomez, A., Brussee, S., Grond, R., Kampinga, H. H. and Rabouille, C. (2021). Activation of IRE1, PERK and salt-inducible kinases leads to Sec body formation in Drosophila S2 cells. J Cell Sci 134(17). PubMed ID: 34350957

    Aguilera-Gomez, A., Zacharogianni, M., van Oorschot, M. M., Genau, H., Grond, R., Veenendaal, T., Sinsimer, K. S., Gavis, E. R., Behrends, C. and Rabouille, C. (2017). Phospho-Rasputin stabilization by Sec16 is required for stress granule formation upon amino acid starvation. Cell Rep 20(4): 935-948. PubMed ID: 28746877

    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

    Zacharogianni, M. and Rabouille, C. (2013). Trafficking along the secretory pathway in Drosophila cell line and tissues: a light and electron microscopy approach. Methods Cell Biol 118: 35-49. PubMed ID: 24295299

    Giuliani, G., Giuliani, F., Volk, T. and Rabouille, C. (2013). The Drosophila RNA-binding protein HOW controls the stability of dgrasp mRNA in the follicular epithelium. Nucleic Acids Res. PubMed ID: 24217913

    Giuliani, F., Giuliani, G., Bauer, R. and Rabouille, C. (2013). Innexin 3, a new gene required for dorsal closure in Drosophila embryo. PLoS One 8: e69212. PubMed ID: 23894431

    Weil, T. T., Parton, R. M., Herpers, B., Soetaert, J., Veenendaal, T., Xanthakis, D., Dobbie, I. M., Halstead, J. M., Hayashi, R., Rabouille, C. and Davis, I. (2012). Drosophila patterning is established by differential association of mRNAs with P bodies. Nat Cell Biol 14: 1305-1313. PubMed ID: 23178881

    Kondylis, V., Tang, Y., Fuchs, F., Boutros, M. and Rabouille, C. (2011). Identification of ER proteins involved in the functional organisation of the early secretory pathway in Drosophila cells by a targeted RNAi screen. PLoS One 6: e17173. PubMed ID: 21383842

  • Svetlana Radyuk Department of Biological Sciences, Southern Methodist University, Dallas
    Odnokoz, O., Nakatsuka, K., Wright, C., Castellanos, J., Klichko, V. I., Kretzschmar, D., Orr, W. C. and Radyuk, S. N. (2021). Mitochondrial Redox Signaling Is Critical to the Normal Functioning of the Neuronal System. Front Cell Dev Biol 9: 613036. PubMed ID: 33585478

    Klichko, V. I., Chow, E. S., Kotwica-Rolinska, J., Orr, W. C., Giebultowicz, J. M. and Radyuk, S. N. (2015). Aging alters circadian regulation of redox in Drosophila. Front Genet 6: 83. PubMed ID: 25806044

    Klichko, V., Sohal, B. H., Radyuk, S. N., Orr, W. C. and Sohal, R. S. (2014). Decrease in cytochrome c oxidase reserve capacity diminishes robustness of Drosophila melanogaster and shortens lifespan. Biochem J 459: 127-135. PubMed ID: 24444354

    Orr, W. C., Radyuk, S. N. and Sohal, R. S. (2013). Involvement of redox state in the aging of Drosophila melanogaster. Antioxid Redox Signal 19: 788-803. PubMed ID: 23458359

  • Jordan Raff Sir William Dunn School of Pathology, University of Oxford
    Alvarez-Rodrigo, I., Wainman, A., Saurya, S. and Raff, J. W. (2021). Ana1 helps recruit Polo to centrioles to promote mitotic PCM assembly and centriole elongation. J Cell Sci 134(14). PubMed ID: 34156068

    Gartenmann, L., Vicente, C. C., Wainman, A., Novak, Z. A., Sieber, B., Richens, J. H. and Raff, J. W. (2020). Sas-6, Ana2 and Sas-4 self-organise into macromolecular structures that can be used to probe centriole/centrosome assembly. J Cell Sci. PubMed ID: 32409564

    Roque, H., Saurya, S., Pratt, M. B., Johnson, E. and Raff, J. W. (2018). Drosophila PLP assembles pericentriolar clouds that promote centriole stability, cohesion and MT nucleation. PLoS Genet 14(2): e1007198. PubMed ID: 29425198

    Aydogan, M. G., Wainman, A., Saurya, S., Steinacker, T. L., Caballe, A., Novak, Z. A., Baumbach, J., Muschalik, N. and Raff, J. W. (2018). A homeostatic clock sets daughter centriole size in flies. J Cell Biol. PubMed ID: 29500190

    Pratt, M. B., Titlow, J. S., Davis, I., Barker, A. R., Dawe, H. R., Raff, J. W. and Roque, H. (2016). Drosophila sensory cilia lacking MKS-proteins exhibit striking defects in development but only subtle defects in adults. J Cell Sci. PubMed ID: 27577095

    Novak, Z. A., Wainman, A., Gartenmann, L. and Raff, J. W. (2016). Cdk1 phosphorylates Drosophila Sas-4 to recruit Polo to daughter centrioles and convert them to centrosomes. Dev Cell 37: 545-557. PubMed ID: 27326932

    Conduit, P. T., Wainman, A., Novak, Z. A., Weil, T. T. and Raff, J. W. (2015). Re-examining the role of Drosophila Sas-4 in centrosome assembly using two-colour-3D-SIM FRAP. Elife 4 [Epub ahead of print]. PubMed ID: 26530814

    Conduit, P. T. and Raff, J. W. (2015). Different Drosophila cell types exhibit differences in mitotic centrosome assembly dynamics. Curr Biol 25: R650-651. PubMed ID: 26241137

    Richens, J. H., Barros, T. P., Lucas, E. P., Peel, N., Pinto, D. M., Wainman, A. and Raff, J. W. (2015). The Drosophila Pericentrin-like-protein (PLP) cooperates with Cnn to maintain the integrity of the outer PCM. Biol Open [Epub ahead of print]. PubMed ID: 26157019

    Baumbach, J., Novak, Z.A., Raff, J.W. and Wainman, A. (2015). Dissecting the function and assembly of acentriolar microtubule organizing centers in Drosophila cells in vivo. PLoS Genet 11: e1005261. PubMed ID: 26020779

  • Padinjat Raghu National Center for Biological Sciences, Tata Institute of Fundamental Research, Hyderabad
    Kumari, A., Ghosh, A., Kolay, S. and Raghu, P. (2022). Septins tune lipid kinase activity and PI(4,5)P(2) turnover during G-protein-coupled PLC signalling in vivo. Life Sci Alliance 5(6). PubMed ID: 35277468

    Panda, A., Thakur, R., Kumari, A. and Raghu, P. (2022). Analysis of Lipid Signaling in Drosophila Photoreceptors using Mass Spectrometry. J Vis Exp(181). PubMed ID: 35311809

    Trivedi, D., Cm, V., Bisht, K., Janardan, V., Pandit, A., Basak, B., H, S., Ramesh, N. and Raghu, P. (2020). A genome engineering resource to uncover principles of cellular organization and tissue architecture by lipid signaling. Elife 9. PubMed ID: 33320085

    Nath, V. R., Mishra, S., Basak, B., Trivedi, D. and Raghu, P. (2020). Extended synaptotagmin regulates membrane contact site structure and lipid transfer function in vivo. EMBO Rep: e50264. PubMed ID: 32716137

    Basu, U., Balakrishnan, S. S., Janardan, V. and Raghu, P. (2020). A PI4KIIIalpha protein complex is required for cell viability during Drosophila wing development. Dev Biol. PubMed ID: 32194035

    Janardan, V., Sharma, S., Basu, U. and Raghu, P. (2019). A genetic screen in Drosophila to identify novel regulation of cell growth by phosphoinositide signaling. G3 (Bethesda). PubMed ID: 31704710

    Sharma, S., Mathre, S., Ramya, V., Shinde, D. and Raghu, P. (2019). Phosphatidylinositol 5 phosphate 4-kinase regulates plasma-membrane PIP3 turnover and insulin signaling. Cell Rep 27(7): 1979-1990.e1977. PubMed ID: 31091438

    Balakrishnan, S. S., Basu, U., Shinde, D., Thakur, R., Jaiswal, M. and Raghu, P. (2018). Regulation of PI4P levels by PI4KIIIalpha during G-protein coupled PLC signaling in Drosophila photoreceptors. J Cell Sci. PubMed ID: 29980590

    Ashe, S., Malhotra, V. and Raghu, P. (2018). Protein kinase D regulates metabolism and growth by controlling secretion of insulin like peptide. Dev Biol 434(1): 175-185. PubMed ID: 29247620

    Yadav, S., Thakur, R., Georgiev, P., Deivasigamani, S., K, H., Ratnaparkhi, G. and Raghu, P. (2017). RDGBalpha localization and function at a membrane contact site is regulated by FFAT/VAP interactions. J Cell Sci. PubMed ID: 29180517

  • Mani Ramaswami Department of Genetics, Trinity College Dublin
    Chen, N., Zhang, Y., Adel, M., Kuklin, E. A., Reed, M. L., Mardovin, J. D., Bakthavachalu, B., VijayRaghavan, K., Ramaswami, M. and Griffith, L. C. (2022). Local translation provides the asymmetric distribution of CaMKII required for associative memory formation. Curr Biol 32(12): 2730-2738. PubMed ID: 35545085

    Trisal, S., Aranha, M., Chodankar, A., VijayRaghavan, K. and Ramaswami, M. (2022). A Drosophila Circuit for Habituation Override. J Neurosci 42(14): 2930-2941. PubMed ID: 35232763

    Singh, A., Kandi, A. R., Jayaprakashappa, D., Thuery, G., Purohit, D. J., Huelsmeier, J., Singh, R., Pothapragada, S. S., Ramaswami, M. and Bakthavachalu, B. (2022). The Transcriptional Response to Oxidative Stress is Independent of Stress-Granule Formation. Mol Biol Cell: mbcE21080418. PubMed ID: 34985933

    Huelsmeier, J., Walker, E., Bakthavachalu, B. and Ramaswami, M. (2021). A C-terminal ataxin-2 disordered region promotes Huntingtin protein aggregation and neurodegeneration in Drosophila models of Huntington's disease. G3 (Bethesda). PubMed ID: 34718534

    Singh, A., Hulsmeier, J., Kandi, A. R., Pothapragada, S. S., Hillebrand, J., Petrauskas, A., Agrawal, K., Rt, K., Thiagarajan, D., Jayaprakashappa, D., VijayRaghavan, K., Ramaswami, M. and Bakthavachalu, B. (2021). Antagonistic roles for Ataxin-2 structured and disordered domains in RNP condensation. Elife 10. PubMed ID: 33689682

    Chodankar, A., Sadanandappa, M. K., VijayRaghavan, K. and Ramaswami, M. (2020). Glomerulus-selective regulation of a critical period for interneuron plasticity in the Drosophila antennal lobe. J Neurosci. PubMed ID: 32532889

    Bakthavachalu, B., Huelsmeier, J., Sudhakaran, I. P., Hilleb Gunderson, J. T., Peppriell, A. E., Krout, I. N., Vorojeikina, D. and Rand, M. D. (2021). Neuroligin-1 is a mediator of methylmercury neuromuscular toxicity. Toxicol Sci. PubMed ID: 34546366 J., Singh, A., Petrauskas, A., Thiagarajan, D., Sankaranarayanan, M., Mizoue, L., Anderson, E. N., Pandey, U. B., Ross, E., VijayRaghavan, K., Parker, R. and Ramaswami, M. (2018). RNP-granule assembly via Ataxin-2 disordered domains is required for long-term memory and neurodegeneration. Neuron 98(4): 754-766.e754. PubMed ID: 29772202

    Kuklin, E. A., Alkins, S., Bakthavachalu, B., Genco, M. C., Sudhakaran, I., Raghavan, K. V., Ramaswami, M. and Griffith, L. C. (2017). The long 3'UTR mRNA of CaMKII is essential for translation-dependent plasticity of spontaneous release in Drosophila melanogaster. J Neurosci [Epub ahead of print]. PubMed ID: 28954869

    Carreira-Rosario, A., Bhargava, V., Hillebrand, J., Kollipara, R. K., Ramaswami, M. and Buszczak, M. (2016). Repression of Pumilio protein expression by Rbfox1 promotes germ cell differentiation. Dev Cell 36: 562-571. PubMed ID: 26954550

    Choudhury, S. D., Mushtaq, Z., Reddy-Alla, S., Balakrishnan, S. S., Thakur, R. S., Krishnan, K. S., Raghu, P., Ramaswami, M. and Kumar, V. (2016). &sigma&2-adaptin facilitates basal synaptic transmission and is required for regenerating endo-exo cycling pool under high frequency nerve stimulation in Drosophila. Genetics. PubMed ID: 26920756

    Kacsoh, B.Z., Bozler, J., Ramaswami, M. and Bosco, G. (2015). Social communication of predator-induced changes in Drosophila behavior and germline physiology. Elife [Epub ahead of print]. PubMed ID: 25970035

  • Saraf Ramesh Department of Studies in Zoology, University of Mysore
    Deepashree, S., Shivanandappa, T. and Ramesh, S. R. (2021). Genetic repression of the antioxidant enzymes reduces the lifespan in Drosophila melanogaster. J Comp Physiol B. PubMed ID: 34625818

    Deepashree, S., Niveditha, S., Shivanandappa, T. and Ramesh, S. R. (2019). Oxidative stress resistance as a factor in aging: evidence from an extended longevity phenotype of Drosophila melanogaster. Biogerontology. PubMed ID: 31054025

    Deepashree, S., Shivanandappa, T. and Ramesh, S. R. (2017). Life history traits of an extended longevity phenotype of Drosophila melanogaster. Curr Aging Sci. PubMed ID: 28183243

    Haddadi, M., Nongthomba, U., Jahromi, S. R. and Ramesh, S. R. (2015). Transgenic Drosophila model to study apolipoprotein E4-induced neurodegeneration. Behav Brain Res [Epub ahead of print]. PubMed ID: 26706888

    Haddadi, M., Jahromi, S. R., Sagar, B. K., Patil, R. K., Shivanandappa, T. and Ramesh, S. R. (2013). Brain aging, memory impairment and oxidative stress: A study in Drosophila melanogaster. Behav Brain Res. PubMed ID: 24183945

    Jahromi, S. R., Haddadi, M., Shivanandappa, T. and Ramesh, S. R. (2013). Neuroprotective Effect of Decalepis hamiltonii in Paraquat-Induced Neurotoxicity in Drosophila melanogaster: Biochemical and Behavioral Evidences. Neurochem Res. PubMed ID: 24173775

    Shruthi, B. and Ramesh, S. R. (2013). Last mated male sperm precedence in doubly mated females is not ubiquitous: evidence from sperm competition in laboratory populations of Drosophila nasuta nasuta and Drosophila nasuta albomicans. J Genet 92: 309-312. PubMed ID: 23970089

    Shruthi, B., Chayakumari, Ram, K. R. and Ramesh, S. R. (2012). Influence of mating histories and age on female remating behaviour in a few closely related species of Drosophila nasuta subgroup. Indian J Exp Biol 50: 156-163. PubMed ID: 22670479

  • Mika Rämet Experimental Immunology, University of Tampere, Finland
    Valanne, S., Jarvela-Stolting, M., Harjula, S. E., Myllymaki, H., Salminen, T. S. and Ramet, M. (2020). Osa-Containing Brahma Complex Regulates Innate Immunity and the Expression of Metabolic Genes in Drosophila. J Immunol. PubMed ID: 32198143

    Valanne, S., Salminen, T. S., Jarvela-Stolting, M., Vesala, L. and Ramet, M. (2019). Immune-inducible non-coding RNA molecule lincRNA-IBIN connects immunity and metabolism in Drosophila melanogaster. PLoS Pathog 15(1): e1007504. PubMed ID: 30633769

    Aittomaki, S., Valanne, S., Lehtinen, T., Matikainen, S., Nyman, T. A., Ramet, M. and Pesu, M. (2017). Proprotein convertase Furin1 expression in the Drosophila fat body is essential for a normal antimicrobial peptide response and bacterial host defense. Faseb J. [Epub ahead of print] PubMed ID: 28705811

    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

    Kuuluvainen, E., Hakala, H., Havula, E., Sahal Estime, M., Ramet, M., Hietakangas, V. and Makela, T. P. (2014). Cyclin-dependent kinase 8 module expression profiling reveals requirement of mediator subunits 12 and 13 for transcription of Serpent-dependent innate immunity genes in Drosophila. J Biol Chem 289: 16252-16261. PubMed ID: 24778181

    >Myllymaki, H., Valanne, S. and Ramet, M. (2014). The Drosophila imd signaling pathway. J Immunol 192: 3455-3462. PubMed ID: 24706930

  • Ricardo Guelerman P Ramos University of Sao Paulo
    Valer, F. B., Spegiorim, G. C., Espreafico, E. M. and Ramos, R. G. P. (2022). The IRM cell adhesion molecules Hibris, Kin of irre and Roughest control egg morphology by modulating ovarian muscle contraction in Drosophila. J Insect Physiol 136: 104344. PubMed ID: 34896373

    Valer, F. B., Machado, M. C. R., Silva-Junior, R. M. P. and Ramos, R. G. P. (2018). Expression of Hbs, Kirre, and Rst during Drosophila ovarian development. Genesis 56(9): e23242. PubMed ID: 30114331

    Machado, M. C. R., Valer, F. B., Couto-Lima, C. A. and Ramos, R. G. P. (2018). Transcriptional cross-regulation of Irre Cell Recognition Module (IRM) members in the Drosophila pupal retina. Mech Dev 154: 193-202. PubMed ID: 30030087

  • David Rand Ecology and Evolutionary Biology, Brown University, Providence, R. I.
    Gunderson, J. T., Peppriell, A. E., Krout, I. N., Vorojeikina, D. and Rand, M. D. (2021). Neuroligin-1 is a mediator of methylmercury neuromuscular toxicity. Toxicol Sci. PubMed ID: 34546366

    Spierer, A. N., Mossman, J. A., Smith, S. P., Crawford, L., Ramachandran, S. and Rand, D. M. (2021). Natural variation in the regulation of neurodevelopmental genes modifies flight performance in Drosophila. PLoS Genet 17(3): e1008887. PubMed ID: 33735180

    Santiago, J. C., Boylan, J. M., Lemieux, F. A., Gruppuso, P. A., Sanders, J. A. and Rand, D. M. (2021). Mitochondrial genotype alters the impact of rapamycin on the transcriptional response to nutrients in Drosophila. BMC Genomics 22(1): 213. PubMed ID: 33761878

    Spierer, A. N., Yoon, D., Zhu, C. T. and Rand, D. M. (2020). FreeClimber: Automated quantification of climbing performance in Drosophila. J Exp Biol. PubMed ID: 33188065

    Mossman, J. A., Biancani, L. M. and Rand, D. M. (2019). Mitochondrial genomic variation drives differential nuclear gene expression in discrete regions of Drosophila gene and protein interaction networks. BMC Genomics 20(1): 691. PubMed ID: 31477008

    Mossman, J. A., Mabeza, R. M. S., Blake, E., Mehta, N. and Rand, D. M. (2019). Age of both parents influences reproduction and egg dumping behavior in Drosophila melanogaster. J Hered. PubMed ID: 30753690

    Mossman, J. A., Ge, J. Y., Navarro, F. and Rand, D. M. (2019). Mitochondrial DNA fitness depends on nuclear genetic background in Drosophila. G3 (Bethesda). PubMed ID: 30745378

    Mossman, J. A., Tross, J. G., Jourjine, N. A., Li, N., Wu, Z. and Rand, D. M. (2016). Mitonuclear Interactions Mediate Transcriptional Responses to Hypoxia in Drosophila. Mol Biol Evol [Epub ahead of print]. PubMed ID: 28110272

    Mossman, J. A., Tross, J. G., Li, N., Wu, Z. and Rand, D. M. (2016). Mitochondrial-nuclear interactions mediate sex-specific transcriptional profiles in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 27558138

    Mossman, J.A., Biancani, L.M. and Rand, D.M. (2016). Mitonuclear epistasis for development time and its modification by diet in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 26966258

  • Prashanth Rangan Biological Sciences, University of Albany
    McCarthy, A., Sarkar, K., Martin, E. T., Upadhyay, M., Jang, S., Williams, N. D., Forni, P. E., Buszczak, M. and Rangan, P. (2022). Msl3 promotes germline stem cell differentiation in female Drosophila. Development 149(1). PubMed ID: 34878097

    Martin, E. T., Blatt, P., Nguyen, E., Lahr, R., Selvam, S., Yoon, H. A. M., Pocchiari, T., Emtenani, S., Siekhaus, D. E., Berman, A., Fuchs, G. and Rangan, P. (2022). A translation control module coordinates germline stem cell differentiation with ribosome biogenesis during Drosophila oogenesis. Dev Cell 57(7): 883-900. PubMed ID: 35413237

    Blatt, P., Wong-Deyrup, S. W., McCarthy, A., Breznak, S., Hurton, M. D., Upadhyay, M., Bennink, B., Camacho, J., Lee, M. T. and Rangan, P. (2021). RNA degradation is required for the germ-cell to maternal transition in Drosophila. Curr Biol. PubMed ID: 33989522

    Blatt, P., Martin, E. T., Breznak, S. M. and Rangan, P. (2020). Post-transcriptional gene regulation regulates germline stem cell to oocyte transition during Drosophila oogenesis. Curr Top Dev Biol 140: 3-34. PubMed ID: 32591078

    Flora, P., Wong-Deyrup, S. W., Martin, E. T., Palumbo, R. J., Nasrallah, M., Oligney, A., Blatt, P., Patel, D., Fuchs, G. and Rangan, P. (2018). Sequential regulation of maternal mRNAs through a conserved cis-acting element in their 3' UTRs. Cell Rep 25(13): 3828-3843.e3829. PubMed ID: 30590052

    McCarthy, A., Deiulio, A., Martin, E. T., Upadhyay, M. and Rangan, P. (2018). Tip60 complex promotes expression of a differentiation factor to regulate germline differentiation in female Drosophila. Mol Biol Cell: mbcE18060385. PubMed ID: 30230973

    Upadhyay, M., Kuna, M., Tudor, S., Martino Cortez, Y. and Rangan, P. (2018). A switch in the mode of Wnt signaling orchestrates the formation of germline stem cell differentiation niche in Drosophila. PLoS Genet 14(1): e1007154. PubMed ID: 29370168

    Flora, P., Schowalter, S., Wong-Deyrup, S., DeGennaro, M., Nasrallah, M. A. and Rangan, P. (2018). Transient transcriptional silencing alters the cell cycle to promote germline stem cell differentiation in Drosophila. Dev Biol 434(1): 84-95. PubMed ID: 29198563

    Navarro-Costa, P., McCarthy, A., Prudencio, P., Greer, C., Guilgur, L. G., Becker, J. D., Secombe, J., Rangan, P. and Martinho, R. G. (2016). Early programming of the oocyte epigenome temporally controls late prophase I transcription and chromatin remodelling. Nat Commun 7: 12331. PubMed ID: 27507044

    Upadhyay, M., Martino Cortez, Y., Wong-Deyrup, S., Tavares, L., Schowalter, S., Flora, P., Hill, C., Nasrallah, M. A., Chittur, S. and Rangan, P. (2016). Transposon Dysregulation Modulates dWnt4 Signaling to Control Germline Stem Cell Differentiation in Drosophila. PLoS Genet 12: e1005918. PubMed ID: 27019121

  • Jose Ranz Ecology & Evolutionary Biology, School of Biological Sciences, U. C. Irvine
    Jayaswal, V., Jimenez, J., Magie, R., Nguyen, K., Clifton, B., Yeh, S. and Ranz, J. M. (2018). A species-specific multigene family mediates differential sperm displacement in Drosophila melanogaster. Evolution 72(2): 399-403. PubMed ID: 29315521

    Clifton, B. D., Librado, P., Yeh, S. D., Solares, E., Real, D., Jayasekera, S., Zhang, W., Shi, M., Park, R., Magie, R., Ma, H. C., Xia, X. Q., Marco, A., Rozas, J. and Ranz, J. M. (2016). Rapid functional and sequence differentiation of a tandemly-repeated species-specific multigene family in Drosophila. Mol Biol Evol [Epub ahead of print]. PubMed ID: 27702774

    Chan, C., Jayasekera, S., Kao, B., Páramo, M., von Grotthuss, M. and Ranz, J.M. (2015). Remodelling of a homeobox gene cluster by multiple independent gene reunions in Drosophila. Nat Commun 6: 6509. PubMed ID: 25739651

    Yeh, S. D., Chan, C. and Ranz, J. M. (2013). Assessing differences in sperm competitive ability in Drosophila. J Vis Exp. PubMed ID: 23995693

    Yeh, S. D., Do, T., Abbassi, M. and Ranz, J. M. (2012). Functional relevance of the newly evolved sperm dynein intermediate chain multigene family in Drosophila melanogaster males. Commun Integr Biol 5: 462-465. PubMed ID: 23181161

    Sahagun, V. and Ranz, J. M. (2012). Characterization of genomic regulatory domains conserved across the genus Drosophila. Genome Biol Evol 4: 1054-1060. PubMed ID: 23042552

    Diaz-Castillo, C. and Ranz, J. M. (2012). Nuclear chromosome dynamics in the Drosophila male germ line contribute to the nonrandom genomic distribution of retrogenes. Mol Biol Evol 29: 2105-2108. PubMed ID: 22427708

  • Yi Rao McGovern Institute For Brain Research, Peking University
    Ren, Q. and Rao, Y. (2022). The exit of axons and glial membrane from the developing Drosophila retina requires integrins. Mol Brain 15(1): 2. PubMed ID: 34980203

    Shaw, H. S., Larkin, J. and Rao, Y. (2020). Phototactic T-maze Behavioral Assay for Comparing the Functionality of Color-sensitive Photoreceptor Subtypes in the Drosophila Visual System. Bio Protoc 10(6): e3558. PubMed ID: 33659530

    Dai, X., Zhou, E., Yang, W., Mao, R., Zhang, W. and Rao, Y. (2021). Molecular resolution of a behavioral paradox: sleep and arousal are regulated by distinct acetylcholine receptors in different neuronal types in Drosophila. Sleep. PubMed ID: 33493349

    Liu, Z., Chen, Y. and Rao, Y. (2020). An RNAi screen for secreted factors and cell-surface players in coordinating neuron and glia development in Drosophila. Mol Brain 13(1): 1. PubMed ID: 31900209

    Shaw, H. S., Cameron, S. A., Chang, W. T. and Rao, Y. (2019). The conserved IgSF9 protein Borderless regulates axonal transport of presynaptic components and color vision in Drosophila. J Neurosci. PubMed ID: 31235647

    Dai, X., Zhou, E., Yang, W., Zhang, X., Zhang, W. and Rao, Y. (2019). D-Serine made by serine racemase in Drosophila intestine plays a physiological role in sleep. Nat Commun 10(1): 1986. PubMed ID: 31064979

    Ramin, M., Li, Y., Chang, W. T., Shaw, H. and Rao, Y. (2019). The peacefulness gene promotes aggression in Drosophila. Mol Brain 12(1): 1. PubMed ID: 30606245

    Qian, Y., Cao, Y., Deng, B., Yang, G., Li, J., Xu, R., Zhang, D., Huang, J. and Rao, Y. (2017). Sleep homeostasis regulated by 5HT2b receptor in a small subset of neurons in the dorsal fan-shaped body of drosophila. Elife 6. PubMed ID: 28984573

    Chen, Y., Cameron, S., Chang, W. T. and Rao, Y. (2017). Turtle interacts with borderless in regulating glial extension and axon ensheathment. Mol Brain 10(1): 17. PubMed ID: 28535795

    Cameron, S., Chen, Y. and Rao, Y. (2016). Borderless regulates glial extension and axon ensheathment. Dev Biol [Epub ahead of print]. PubMed ID: 27131624

    Ramin, M., Domocos, C., Slawaska-Eng, D. and Rao, Y. (2014). Aggression and social experience: genetic analysis of visual circuit activity in the control of aggressiveness in. Mol Brain 7: 55. PubMed ID: 25116850

  • Tobias Rasse Hertie-Institute for Clinical Brain Research, University of Tuebingen
    Zhu, J. Y., Hannan, S. B., Drager, N. M., Vereshchagina, N., Krahl, A. C., Fu, Y., Elliott, C. J. H., Han, Z., Jahn, T. R. and Rasse, T. M. (2021). Autophagy inhibition rescues structural and functional defects caused by the loss of mitochondrial chaperone Hsc70-5 in Drosophila. Autophagy. PubMed ID: 33404278

    Zhang, Y. V., Hannan, S. B., Kern, J. V., Stanchev, D. T., Koc, B., Jahn, T. R. and Rasse, T. M. (2017). The KIF1A homolog Unc-104 is important for spontaneous release, postsynaptic density maturation and perisynaptic scaffold organization. Sci Rep 7: 38172. PubMed ID: 28344334

    Zhang, Y. V., Hannan, S. B., Stapper, Z. A., Kern, J. V., Jahn, T. R. and Rasse, T. M. (2016). The Drosophila KIF1A homolog unc-104 is important for site-specific synapse maturation. Front Cell Neurosci 10: 207. PubMed ID: 27656128

    Zhu, J. Y., Vereshchagina, N., Sreekumar, V., Burbulla, L. F., Costa, A. C., Daub, K. J., Woitalla, D., Martins, L. M., Kruger, R. and Rasse, T. M. (2013). Knockdown of Hsc70-5/mortalin Induces Loss of Synaptic Mitochondria in a Drosophila Parkinson's Disease Model. PLoS One 8: e83714. PubMed ID: 24386261

    Kern, J. V., Zhang, Y. V., Kramer, S., Brenman, J. E. and Rasse, T. M. (2013). The Kinesin-3, Unc-104 Regulates Dendrite Morphogenesis and Synaptic Development in Drosophila. Genetics. PubMed ID: 23770702

    Fuger, P., Sreekumar, V., Schule, R., Kern, J. V., Stanchev, D. T., Schneider, C. D., Karle, K. N., Daub, K. J., Siegert, V. K., Flotenmeyer, M., Schwarz, H., Schols, L. and Rasse, T. M. (2012). Spastic paraplegia mutation N256S in the neuronal microtubule motor KIF5A disrupts axonal transport in a Drosophila HSP model. PLoS Genet 8: e1003066. PubMed ID: 23209432

    Zhang, Y., Fuger, P., Hannan, S. B., Kern, J. V., Lasky, B. and Rasse, T. M. (2010). In vivo imaging of intact Drosophila larvae at sub-cellular resolution. J Vis Exp. PubMed ID: 20864927

  • Christina Rathke Entwicklungsbiologie, Philipps-Universität, Marburg, Germany
    Theofel, I., Bartkuhn, M., Boettger, T., Gartner, S. M., Kreher, J., Brehm, A. and Rathke, C. (2017). tBRD-1 and tBRD-2 regulate expression of genes necessary for spermatid differentiation. Biol Open. PubMed ID: 28235844

    Heseding, C., Saumweber, H., Rathke, C. and Ehrenhofer-Murray, A. E. (2016). Widespread colocalization of the Drosophila histone acetyltransferase homolog MYST5 with DREF and insulator proteins at active genes. Chromosoma. PubMed ID: 26894919

    Eren-Ghiani, Z., Rathke, C., Theofel, I. and Renkawitz-Pohl, R. (2015). Prtl99C Acts Together with Protamines and Safeguards Male Fertility in Drosophila. Cell Rep 13: 2327-2335. PubMed ID: 26673329

    Kost, N., Kaiser, S., Ostwal, Y., Riedel, D., Stutzer, A., Nikolov, M., Rathke, C., Renkawitz-Pohl, R. and Fischle, W. (2015). Multimerization of Drosophila sperm protein Mst77F causes a unique condensed chromatin structure. Nucleic Acids Res 43: 3033-3045. PubMed ID: 25735749

    Gartner, S. M., Rothenbusch, S., Buxa, M. K., Theofel, I., Renkawitz, R., Rathke, C. and Renkawitz-Pohl, R. (2015). The HMG-box-containing proteins tHMG-1 and tHMG-2 interact during the histone-to-protamine transition in Drosophila spermatogenesis. Eur J Cell Biol 94: 46-59. PubMed ID: 25464903

  • Kristipati Ravi Ram Embryotoxicology Division, Indian Institute of Toxicology Research, Lucknow
    Sharma, V., Pandey, A. K., Kumar, A., Misra, S., Gupta, H. P. K., Gupta, S., Singh, A., Buehner, N. A. and Ravi Ram, K. (2017). Functional male accessory glands and fertility in Drosophila require novel ecdysone receptor. PLoS Genet 13(5): e1006788. PubMed ID: 28493870

    Misra, S., Pandey, A. K., Gupta, S., Kumar, A., Khanna, P., Shankar, J. and Ravi Ram, K. (2017). Estrogen related receptor is required for the testicular development and for the normal sperm axoneme/mitochondrial derivatives in Drosophila males. Sci Rep 7: 40372. PubMed ID: 28094344

    Misra, S., Kumar, A., Ratnasekhar, C., Sharma, V., Mudiam, M. K. and Ravi Ram, K. (2014). Exposure to endosulfan influences sperm competition in Drosophila melanogaster. Sci Rep 4: 7433. PubMed ID: 25503806

    Tiwari, A. K., Pragya, P., Ravi Ram, K. and Chowdhuri, D. K. (2011). Environmental chemical mediated male reproductive toxicity: Drosophila melanogaster as an alternate animal model. Theriogenology 76(2): 197-216. PubMed ID: 21356551

  • Girish Ratnaparkhi Indian Institute of Science Education & Research (IISER), Pune
    Soory, A. and Ratnaparkhi, G. S. (2022). SUMOylation of Jun fine-tunes the Drosophila gut immune response. PLoS Pathog 18(3): e1010356. PubMed ID: 35255103

    Hegde, S., Sreejan, A., Gadgil, C. J. and Ratnaparkhi, G. S. (2022). SUMOylation of Dorsal attenuates Toll/NF-κB signaling. Genetics 221(3). PubMed ID: 35567478

    Soory, A. and Ratnaparkhi, G. S. (2022). SUMOylation of Jun fine-tunes the Drosophila gut immune response. PLoS Pathog 18(3): e1010356. PubMed ID: 35255103

    Nayak, P., Kejriwal, A. and Ratnaparkhi, G. S. (2021). SUMOylation of Arginyl tRNA Synthetase Modulates the Drosophila Innate Immune Response. Front Cell Dev Biol 9: 695630. PubMed ID: 34660574

    Shweta, K., Basargekar, A. and Ratnaparkhi, A. (2021). FGFR/Heartless and Smog interact synergistically to negatively regulate Fog mediated G-protein coupled receptor signaling in the Drosophila nervous system. G3 (Bethesda) 11(3). PubMed ID: 33729500

    Harish, R. K., Tendulkar, S., Deivasigamani, S., Ratnaparkhi, A. and Ratnaparkhi, G. S. (2019). Monensin Sensitive 1 Regulates Dendritic Arborization in Drosophila by Modulating Endocytic Flux. Front Cell Dev Biol 7: 145. PubMed ID: 31428611

    Dhiman, N., Shweta, K., Tendulkar, S., Deshpande, G., Ratnaparkhi, G. S. and Ratnaparkhi, A. (2019). Drosophila Mon1 constitutes a novel node in the brain-gonad axis that is essential for female germline maturation. Development 146(13). PubMed ID: 31292144

    Chaplot, K., Pimpale, L., Ramalingam, B., Deivasigamani, S., Kamat, S. S. and Ratnaparkhi, G. S. (2019). SOD1 activity threshold and TOR signalling modulate VAP(P58S) aggregation via reactive oxygen species-induced proteasomal degradation in a Drosophila model of amyotrophic lateral sclerosis. Dis Model Mech 12(2). PubMed ID: 30635270

    Abhyankar, V., Kaduskar, B., Kamat, S. S., Deobagkar, D. and Ratnaparkhi, G. S. (2018). Drosophila DNA/RNA methyltransferase contributes to robust host defense in aging animals by regulating sphingolipid metabolism. J Exp Biol 221(Pt 22). PubMed ID: 30254027

  • Matteo Rauzi Institut de Biologie Valrose, Parc Valrose, Nice, France
    John, A. and Rauzi, M. (2021). A two-tier junctional mechanism drives simultaneous tissue folding and extension. Dev Cell. PubMed ID: 33891900

    Popkova, A., Rauzi, M. and Wang, X. (2021). Cellular and Supracellular Planar Polarity: A Multiscale Cue to Elongate the Drosophila Egg Chamber. Front Cell Dev Biol 9: 645235. PubMed ID: 33738289

    Popkova, A., Stone, O. J., Chen, L., Qin, X., Liu, C., Liu, J., Belguise, K., Montell, D. J., Hahn, K. M., Rauzi, M. and Wang, X. (2020). A Cdc42-mediated supracellular network drives polarized forces and Drosophila egg chamber extension. Nat Commun 11(1): 1921. PubMed ID: 32317641

  • Anandasankar Ray Department of Entomology, University of Riverside
    Kumar, A., Tauxe, G. M., Perry, S., Scott, C. A., Dahanukar, A. and Ray, A. (2020). Contributions of the Conserved Insect Carbon Dioxide Receptor Subunits to Odor Detection. Cell Rep 31(2): 107510. PubMed ID: 32294446

    MacWilliam, D., Kowalewski, J., Kumar, A., Pontrello, C. and Ray, A. (2018). Signaling mode of the broad-spectrum conserved CO2 Receptor is one of the important determinants of odor valence in Drosophila. Neuron 97(5): 1153-1167.e1154. PubMed ID: 29429938

    Lee, B., Sahoo, A., Marchica, J., Holzhauser, E., Chen, X., Li, J. L., Seki, T., Govindarajan, S. S., Markey, F. B., Batish, M., Lokhande, S. J., Zhang, S., Ray, A. and Perera, R. J. (2017). The long noncoding RNA SPRIGHTLY acts as an intranuclear organizing hub for pre-mRNA molecules. Sci Adv 3(5): e1602505. PubMed ID: 28508063

    Perry, S., Kiragasi, B., Dickman, D. and Ray, A. (2017). The role of Histone Deacetylase 6 in synaptic plasticity and memory. Cell Rep 18: 1337-1345. PubMed ID: 28178513

    Boyle, S. M., McInally, S., Tharadra, S. and Ray, A. (2016). Short-term memory trace mediated by termination kinetics of olfactory receptor. Sci Rep 6: 19863. PubMed ID: 26830661

    Krause Pham, C. and Ray, A. (2015). Conservation of olfactory avoidance in Drosophila species and identification of repellents for Drosophila suzukii. Sci Rep 5: 11527. PubMed ID: 26098542

    Clark, J. T. and Ray, A. (2014). The taste of togetherness. Elife 3. PubMed ID: 25497434

    Boyle, S. M., McInally, S. and Ray, A. (2013). Expanding the olfactory code by in silico decoding of odor-receptor chemical space. Elife 2: e01120. PubMed ID: 24137542

    Boyle, S. M., McInally, S. and Ray, A. (2013). Expanding the olfactory code by in silico decoding of odor-receptor chemical space. Elife 2: e01120. PubMed ID: 24137542

    Tharadra, S. K., Medina, A. and Ray, A. (2013). Advantage of the Highly Restricted Odorant Receptor Expression Pattern in Chemosensory Neurons of. PLoS One 8: e66173. PubMed ID: 23840419

  • Krishanu Ray Tata Institute of Fundamental Research
    Gadre, P., Nitsure, N., Mazumdar, D., Gupta, S. and Ray, K. (2021). The rates of stem cell division determine the cell cycle lengths of its lineage. iScience 24(11): 103232. PubMed ID: 34746698

    Jana, S. C., Dutta, P., Jain, A., Singh, A., Adusumilli, L., Girotra, M., Kumari, D., Shirolikar, S. and Ray, K. (2021). Kinesin-2 transports Orco into the olfactory cilium of Drosophila melanogaster at specific developmental stages. PLoS Genet 17(8): e1009752. PubMed ID: 34411092

    Kapoor, T., Dubey, P., Shirolikar, S. and Ray, K. (2021). An actomyosin clamp assembled by the Amphiphysin-Rho1-Dia/DAAM-Rok pathway reinforces somatic cell membrane folded around spermatid heads. Cell Rep 34(13): 108918. PubMed ID: 33789114

    Gadre, P., Chatterjee, S., Varshney, B. and Ray, K. (2020). Cyclin E and Cdk1 regulate the termination of germline transit-amplification process in Drosophila testis. Cell Cycle 19(14): 1786-1803. PubMed ID: 32573329

    Ahmed, Z., Mazumdar, S. and Ray, K. (2019). Kinesin associated protein, DmKAP, binding harnesses the C-terminal ends of the Drosophila kinesin-2 stalk heterodimer. Biochem Biophys Res Commun. PubMed ID: 31784087

    Ahmed, Z., Doodhi, H., Bhaumik, A., Mazumdar, S. and Ray, K. (2019). The structural dynamics of the kinesin-2 stalk heterodimer and its biological relevance. Biochem Biophys Res Commun 518(1): 171-177. PubMed ID: 31420166

    Dubey, P., Kapoor, T., Gupta, S., Shirolikar, S. and Ray, K. (2019). Atypical septate junctions maintain the somatic enclosure around maturing spermatids and prevent premature sperm release in Drosophila testis. Biol Open. PubMed ID: 30635267

    Gupta, S., Varshney, B., Chatterjee, S. and Ray, K. (2018). Somatic ERK activation during transit amplification is essential for maintaining the synchrony of germline divisions in Drosophila testis. Open Biol 8(7). PubMed ID: 30045884

    Dey, S. and Ray, K. (2018). Cholinergic activity is essential for maintaining the anterograde transport of Choline Acetyltransferase in Drosophila. Sci Rep 8(1): 8028. PubMed ID: 29795337

    Gupta, S. and Ray, K. (2017). Somatic PI3K activity regulates transition to the spermatocyte stages in Drosophila testis. J Biosci 42(2): 285-297. PubMed ID: 28569252


  • Don Ready Department of Biological Sciences, Purdue University
    Ready, D. F. and Chang, H. C. (2021). Calcium waves facilitate and coordinate the contraction of endfeet actin stress fibers in Drosophila interommatidial cells. Development. PubMed ID: 34698814

    Xu, Z., Chikka, M. R., Xia, H. and Ready, D. F. (2016). Ire1 supports normal ER differentiation in developing Drosophila photoreceptors. J Cell Sci [Epub ahead of print]. PubMed ID: 26787744

    Sengupta, S., Barber, T. R., Xia, H., Ready, D. F., Hardie, R. C. (2013) PI(4,5)P2 Depletion underlies Retinal Degeneration in Drosophila trp mutants. J Cell Sci. PubMed ID: 23378018

    Xia, H. and Ready, D. F. (2011). Ectoplasm, ghost in the R cell machine? Dev Neurobiol 71: 1246-1257. PubMed ID: 21542135

    Satoh, A. K., Xia, H., Yan, L., Liu, C. H., Hardie, R. C. and Ready, D. F. (2010). Arrestin translocation is stoichiometric to rhodopsin isomerization and accelerated by phototransduction in Drosophila photoreceptors. Neuron 67: 997-1008. PubMed ID: 20869596

  • Ilaria Rebay MGCB, University of Chicago
    Wu, C., Boisclair Lachance, J. F., Ludwig, M. Z. and Rebay, I. (2020). A context-dependent bifurcation in the Pointed transcriptional effector network contributes specificity and robustness to retinal cell fate acquisition. PLoS Genet 16(11): e1009216. PubMed ID: 33253156

    Hope, C. M., Webber, J. L., Tokamov, S. A. and Rebay, I. (2018). Tuned polymerization of the transcription factor Yan limits off-DNA sequestration to confer context-specific repression. Elife 7. PubMed ID: 30412049

    Webber, J. L., Zhang, J., Massey, A., Sanchez-Luege, N. and Rebay, I. (2018). Collaborative repressive action of the antagonistic ETS transcription factors Pointed and Yan fine-tunes gene expression to confer robustness in Drosophila. Development. PubMed ID: 29848501

    Boisclair Lachance, J. F., Webber, J. L., Hong, L., Dinner, A. and Rebay, I. (2018). Cooperative recruitment of Yan via a high-affinity ETS supersite organizes repression to confer specificity and robustness to cardiac cell fate specification. Genes Dev. PubMed ID: 29535190

    Davis, T. L. and Rebay, I. (2017). Pleiotropy in Drosophila organogenesis: mechanistic insights from Combgap and the Retinal Determination Gene Network. Fly (Austin) [Epub ahead of print] PubMed ID: 29125381

    Davis, T. L., Hoi, C. S. L. and Rebay, I. (2017). Mutations that impair Eyes absent tyrosine phosphatase activity in vitro reduce robustness of retinal determination gene network output in Drosophila. PLoS One 12(11): e0187546. PubMed ID: 29108015

    Davis, T. L. and Rebay, I. (2017). Antagonistic regulation of the second mitotic wave by Eyes absent-Sine oculis and Combgap coordinates proliferation and specification in the Drosophila retina. Development. PubMed ID: 28619818

    Hope, C. M., Rebay, I. and Reinitz, J. (2017). DNA occupancy of polymerizing transcription factors: A chemical model of the ETS family factor Yan. Biophys J 112(1): 180-192. PubMed ID: 28076810

    Hoi, C.S., Xiong, W. and Rebay, I. (2016). Retinal axon guidance requires integration of Eya and the JAK/STAT pathway into phosphotyrosine-based signaling circuitries in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 27194748

    Pelaez, N., Gavalda-Miralles, A., Wang, B., Navarro, H. T., Gudjonson, H., Rebay, I., Dinner, A. R., Katsaggelos, A. K., Amaral, L. A. and Carthew, R. W. (2015). Dynamics and heterogeneity of a fate determinant during transition towards cell differentiation. Elife 4 [Epub ahead of print]. PubMed ID: 26583752

  • Mark Rebeiz Department of Biological Science, University of Pittsburgh
    Rice, G. R., David, J. R., Gompel, N., Yassin, A. and Rebeiz, M. (2021). Resolving between novelty and homology in the rapidly evolving phallus of Drosophila. J Exp Zool B Mol Dev Evol. PubMed ID: 34958528

    Hughes, J. T., Williams, M. E., Rebeiz, M. and Williams, T. M. (2021). Widespread cis- and trans-regulatory evolution underlies the origin, diversification, and loss of a sexually dimorphic fruit fly pigmentation trait. J Exp Zool B Mol Dev Evol. PubMed ID: 34254440

    Vincent, B. J., Rice, G. R., Wong, G. M., Glassford, W. J., Downs, K. I., Shastay, J. L., Charles-Obi, K., Natarajan, M., Gogol, M., Zeitlinger, J. and Rebeiz, M. (2019). An atlas of transcription factors expressed in male pupal terminalia of Drosophila melanogaster. G3 (Bethesda). PubMed ID: 31619460

    Liu, Y., Ramos-Womack, M., Han, C., Reilly, P., Brackett, K. L., Rogers, W., Williams, T. M., Andolfatto, P., Stern, D. L. and Rebeiz, M. (2019). Changes throughout a genetic network mask the contribution of Hox gene evolution. Curr Biol 29(13): 2157-2166.e2156. PubMed ID: 31257142

    Grover, S., Williams, M. E., Kaiser, R., Hughes, J. T., Gresham, L., Rebeiz, M. and Williams, T. M. (2018). Augmentation of a wound response element accompanies the origin of a Hox-regulated Drosophila abdominal pigmentation trait. Dev Biol. PubMed ID: 29981311

    Roeske, M. J., Camino, E. M., Grover, S., Rebeiz, M. and Williams, T. M. (2018). Cis-regulatory evolution integrated the Bric-a-brac transcription factors into a novel fruit fly gene regulatory network. Elife 7. PubMed ID: 29297463

    Signor, S. A., Liu, Y., Rebeiz, M. and Kopp, A. (2016). Genetic convergence in the evolution of male-specific color patterns in Drosophila. Curr Biol. PubMed ID: 27546578

    Camino, E. M., Butts, J. C., Ordway, A., Vellky, J. E., Rebeiz, M. and Williams, T. M. (2015). The evolutionary origination and diversification of a dimorphic gene regulatory network through parallel innovations in cis and trans. PLoS Genet 11: e1005136. PubMed ID: 25835988

    Johnson, W. C., Ordway, A. J., Watada, M., Pruitt, J. N., Williams, T. M. and Rebeiz, M. (2015). Genetic changes to a transcriptional silencer element confers phenotypic diversity within and between Drosophila species. PLoS Genet 11: e1005279. PubMed ID: 26115430

    Ordway, A., Hancuch, K. N., Johnson, W., Wiliams, T. M. and Rebeiz, M. (2014). The expansion of body coloration involves coordinated evolution in cis and trans within the pigmentation regulatory network of Drosophila prostipennis. Dev Biol. PubMed ID: 24907418

  • Bruce Reed University of Waterloo, Waterloo, Ontario
    Kim, M., Du, O. Y., Whitney, R. J., Wilk, R., Hu, J., Krause, H. M., Kavaler, J. and Reed, B. H. (2019). A Functional analysis of the Drosophila gene hindsight: Evidence for positive regulation of EGFR signaling. G3 (Bethesda). PubMed ID: 31649045

    Baechler, B. L., McKnight, C., Pruchnicki, P. C., Biro, N. A. and Reed, B. H. (2015). Hindsight/RREB-1 functions in both the specification and differentiation of stem cells in the adult midgut of Drosophila. Biol Open. PubMed ID: 26658272

    Ming, L., Wilk, R., Reed, B. H. and Lipshitz, H. D. (2013). Drosophila Hindsight and mammalian RREB-1 are evolutionarily conserved DNA-binding transcriptional attenuators. Differentiation 86: 159-170. PubMed ID: 24418439

    Cormier, O., Mohseni, N., Voytyuk, I. and Reed, B. H. (2012). Autophagy can promote but is not required for epithelial cell extrusion in the amnioserosa of the Drosophila embryo. Autophagy 8: 252-264. PubMed ID: 22240588

  • Robert Reenan Robert Reenan, Brown University
    Şahin, A., Held, A., Bredvik, K., Major, P., Achilli, T.M., Kerson, A.G., Wharton, K., Stilwell, G. and Reenan, R. (2016). Human SOD1 ALS mutations in a Drosophila knock-in model cause severe phenotypes and reveal dosage-sensitive gain and loss of function components. Genetics [Epub ahead of print]. PubMed ID: 27974499

    Savva, Y. A., Laurent, G. S. and Reenan, R. A. (2016). Genome-Wide Analysis of A-to-I RNA Editing. Methods Mol Biol 1358: 255-268. PubMed ID: 26463388

    Rieder, L. E., Savva, Y. A., Reyna, M. A., Chang, Y. J., Dorsky, J. S., Rezaei, A. and Reenan, R. A. (2015). Dynamic response of RNA editing to temperature in Drosophila. BMC Biol 13: 1. PubMed ID: 25555396

    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

    St Laurent, G., Tackett, M. R., Nechkin, S., Shtokalo, D., Antonets, D., Savva, Y. A., Maloney, R., Kapranov, P., Lawrence, C. E. and Reenan, R. A. (2013). Genome-wide analysis of A-to-I RNA editing by single-molecule sequencing in Drosophila. Nat Struct Mol Biol. PubMed ID: 24077224

    Rieder, L. E., Staber, C. J., Hoopengardner, B. and Reenan, R. A. (2013). Tertiary structural elements determine the extent and specificity of messenger RNA editing. Nat Commun 4: 2232. PubMed ID: 23903876

    Gell, S. L. and Reenan, R. A. (2012). Mutations to the piRNA Pathway Component aubergine Enhance Meiotic Drive of Segregation Distorter in Drosophila melanogaster. Genetics. PubMed ID: 23267055

    Ryan, M. Y., Maloney, R., Fineberg, J. D., Reenan, R. A. and Horn, R. (2012). RNA editing in eag potassium channels: Biophysical consequences of editing a conserved S6 residue. Channels (Austin) 6: 443-452. PubMed ID: 23064203

    Sun, L., Gilligan, J., Staber, C., Schutte, R. J., Nguyen, V., O'Dowd, D. K. and Reenan, R. (2012). A knock-in model of human epilepsy in Drosophila reveals a novel cellular mechanism associated with heat-induced seizure. J Neurosci 32: 14145-14155. PubMed ID: 23055484

  • Greg Reeves Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh
    Al Asafen, H., Bandodkar, P. U., Carrell-Noel, S., Schloop, A. E., Friedman, J. and Reeves, G. T. (2020). Robustness of the Dorsal morphogen gradient with respect to morphogen dosage. PLoS Comput Biol 16(4): e1007750. PubMed ID: 32251432

    Bandodkar, P. U., Al Asafen, H. and Reeves, G. T. (2020). Spatiotemporal control of gene expression boundaries using a feedforward loop. Dev Dyn. PubMed ID: 31925874

    Carrell, S. N., O'Connell, M. D., Jacobsen, T., Pomeroy, A. E., Hayes, S. M. and Reeves, G. T. (2017). A facilitated diffusion mechanism establishes the Drosophila Dorsal gradient. Development 144(23): 4450-4461. PubMed ID: 29097443

    O'Connell, M. D. and Reeves, G. T. (2015). The presence of nuclear cactus in the early Drosophila embryo may extend the dynamic range of the dorsal gradient. PLoS Comput Biol 11: e1004159. PubMed ID: 25879657

    Carrell, S. N. and Reeves, G. T. (2015). Imaging the dorsal-ventral axis of live and fixed Drosophila melanogaster embryos. Methods Mol Biol 1189: 63-78. PubMed ID: 25245687

    Garcia, M., Nahmad, M., Reeves, G. T. and Stathopoulos, A. (2013). Size-dependent regulation of dorsal-ventral patterning in the early Drosophila embryo. Dev Biol 381: 286-299. PubMed ID: 23800450


  • Marc Rehmsmeier Humboldt Universitët zu Berlin
    Bredesen, B. A. and Rehmsmeier, M. (2019). DNA sequence models of genome-wide Drosophila melanogaster Polycomb binding sites improve generalization to independent Polycomb Response Elements. Nucleic Acids Res. PubMed ID: 31340029

    Hauenschild, A., Ringrose, L., Altmutter, C., Paro, R. and Rehmsmeier, M. (2008). Evolutionary plasticity of polycomb/trithorax response elements in Drosophila species. PLoS Biol 6(10): e261. PubMed ID: 18959483

  • Heinrich Reichert University of Basel
    Cammarata-Mouchtouris, A., Nguyen, X. H., Acker, A., Bonnay, F., Goto, A., Orian, A., Fauvarque, M. O., Boutros, M., Reichhart, J. M. and Matt, N. (2020). Hyd ubiquitinates the NF-kappaB co-factor Akirin to operate an effective immune response in Drosophila. PLoS Pathog 16(4): e1008458. PubMed ID: 32339205

    Landskron, L., Steinmann, V., Bonnay, F., Burkard, T. R., Steinmann, J., Reichardt, I., Harzer, H., Laurenson, A. S., Reichert, H. and Knoblich, J. A. (2018). The asymmetrically segregating lncRNA cherub is required for transforming stem cells into malignant cells. Elife 7. PubMed ID: 29580384

    Gowda, S. B. M., Paranjpe, P. D., Reddy, O. V., Thiagarajan, D., Palliyil, S., Reichert, H. and VijayRaghavan, K. (2018). GABAergic inhibition of leg motoneurons is required for normal walking behavior in freely moving Drosophila. Proc Natl Acad Sci U S A 115(9): E2115-e2124. PubMed ID: 29440493

    Bohra, A. A., Kallman, B. R., Reichert, H. and VijayRaghavan, K. (2018). Identification of a single pair of interneurons for bitter taste processing in the Drosophila brain. Curr Biol 28(6): 847-858.e843. PubMed ID: 29502953

    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

    Abramczuk, M. K., Burkard, T. R., Rolland, V., Steinmann, V., Duchek, P., Jiang, Y., Wissel, S., Reichert, H. and Knoblich, J. A. (2017). The splicing co-factor Barricade/Tat-SF1, is required for cell cycle and lineage progression in Drosophila neural stem cells. Development. PubMed ID: 28935704

    Koltun, B., Shackelford, E., Bonnay, F., Matt, N., Reichhart, J. M. and Orian, A. (2017). The SUMO-targeted ubiquitin ligase, Dgrn, is essential for Drosophila innate immunity. Int J Dev Biol 61(3-4-5): 319-327. PubMed ID: 28621429

    Schwarz, O., Bohra, A. A., Liu, X., Reichert, H., VijayRaghavan, K. and Pielage, J. (2017). Motor control of Drosophila feeding behavior. Elife 6 [Epub ahead of print]. PubMed ID: 28211791

    Syed, D.S., Bm, S., Reddy, O.V., Reichert, H. and VijayRaghavan, K. (2016). Glial and neuronal Semaphorin signaling instruct the development of a functional myotopic map for Drosophila walking. Elife [Epub ahead of print]. PubMed ID: 26926907

    Sen, S., Cao, D., Choudhary, R., Biagini, S., Wang, J. W., Reichert, H. and VijayRaghavan, K. (2014). Genetic transformation of structural and functional circuitry rewires the brain. Elife 3. PubMed ID: 25546307

  • Jean-Marc Reichhart Genetique de la Reponse Immunitaire, IBMC, Universite de Strasbourg
    Koltun, B., Shackelford, E., Bonnay, F., Matt, N., Reichhart, J. M. and Orian, A. (2017). The SUMO-targeted ubiquitin ligase, Dgrn, is essential for Drosophila innate immunity. Int J Dev Biol 61(3-4-5): 319-327. PubMed ID: 28621429

    El-Chamy, L., Matt, N., Ntwasa, M. and Reichhart, J. M. (2015). The Multilayered Innate Immune Defense of the Gut. Biomed J [Epub ahead of print]. PubMed ID: 26068126

    Bonnay, F., Nguyen, X. H., Cohen-Berros, E., Troxler, L., Batsche, E., Camonis, J., Takeuchi, O., Reichhart, J. M. and Matt, N. (2014). Akirin specifies NF-kappaB selectivity of Drosophila innate immune response via chromatin remodeling. EMBO J. PubMed ID: 25180232

    Bonnay, F., Cohen-Berros, E., Hoffmann, M., Kim, S. Y., Boulianne, G. L., Hoffmann, J. A., Matt, N., Reichhart, J. M. (2013) big bang gene modulates gut immune tolerance in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 23378635

    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

    Reichhart, J. M., Gubb, D. and Leclerc, V. (2011). The Drosophila serpins: multiple functions in immunity and morphogenesis. Methods Enzymol 499: 205-225. PubMed ID: 21683256

  • Ingolf Reim Developmental Biology, Friedrich-Alexander-University, Erlangen-Nuremberg
    Schwarz, B., Hollfelder, D., Scharf, K., Hartmann, L. and Reim, I. (2018). Diversification of heart progenitor cells by EGF signaling and differential modulation of ETS protein activity. Elife 7. PubMed ID: 29869981

    Domsch, K., Acs, A., Obermeier, C., Nguyen, H. T. and Reim, I. (2017). Identification of the essential protein domains for Mib2 function during the development of the Drosophila larval musculature and adult flight muscles. PLoS One 12(3): e0173733. PubMed ID: 28282454

    Reim, I., Frasch, M. and Schaub, C. (2017). T-Box Genes in Drosophila Mesoderm Development. Curr Top Dev Biol 122: 161-193. PubMed ID: 28057263

    Schaub, C., Marz, J., Reim, I. and Frasch, M. (2015). Org-1-dependent lineage reprogramming generates the ventral longitudinal musculature of the Drosophila heart. Curr Biol 25(4): 488-494. PubMed ID: 25660543

  • Danny Reinberg NYU Langone Health Center
    Narendra, V., Bulajić, M., Dekker, J., Mazzoni, E.O. and Reinberg, D. (2016). CTCF-mediated topological boundaries during development foster appropriate gene regulation. Genes Dev 30: 2657-2662. PubMed ID: 28087711

    Zhang, J., Bonasio, R., Strino, F., Kluger, Y., Holloway, J. K., Modzelewski, A. J., Cohen, P. E., Reinberg, D. (2013) SFMBT1 functions with LSD1 to regulate expression of canonical histone genes and chromatin-related factors. Genes Dev 27: 749-766. PubMed ID: 23592795

    Fuda, N. J., Buckley, M. S., Wei, W., Core, L. J., Waters, C. T., Reinberg, D. and Lis, J. T. (2012). Fcp1 dephosphorylation of the RNA polymerase II C-terminal domain is required for efficient transcription of heat shock genes. Mol Cell Biol 32: 3428-3437. PubMed ID: 22733996

    Li, G., Margueron, R., Ku, M., Chambon, P., Bernstein, B. E. and Reinberg, D. (2010). Jarid2 and PRC2, partners in regulating gene expression. Genes Dev 24: 368-380. PubMed ID: 20123894

    Bonasio, R., Lecona, E. and Reinberg, D. (2010). MBT domain proteins in development and disease. Semin Cell Dev Biol 21: 221-230. PubMed ID: 19778625

  • John Reinitz Department of Statistics, University of Chicago
    Liu, Y., Barr, K. and Reinitz, J. (2020). Fully interpretable deep learning model of transcriptional control. Bioinformatics 36(Supplement_1): i499-i507. PubMed ID: 32657418

    Barr, K., Reinitz, J. and Radulescu, O. (2019). An in silico analysis of robust but fragile gene regulation links enhancer length to robustness. PLoS Comput Biol 15(11): e1007497. PubMed ID: 31730659

    Barr, K. A., Martinez, C., Moran, J. R., Kim, A. R., Ramos, A. F. and Reinitz, J. (2017). Synthetic enhancer design by in silico compensatory evolution reveals flexibility and constraint in cis-regulation. BMC Syst Biol 11(1): 116. PubMed ID: 29187214

    Barr, K. A. and Reinitz, J. (2017). A sequence level model of an intact locus predicts the location and function of nonadditive enhancers. PLoS One 12(7): e0180861. PubMed ID: 28715438

    Hope, C. M., Rebay, I. and Reinitz, J. (2017). DNA occupancy of polymerizing transcription factors: A chemical model of the ETS family factor Yan. Biophys J 112(1): 180-192. PubMed ID: 28076810

    Jiang, P., Ludwig, M. Z., Kreitman, M. and Reinitz, J. (2015). Natural variation of the expression pattern of the segmentation gene even-skipped in melanogaster. Dev Biol [Epub ahead of print]. PubMed ID: 26129990

    Martinez, C., Rest, J. S., Kim, A. R., Ludwig, M., Kreitman, M., White, K. and Reinitz, J. (2014). Ancestral resurrection of the Drosophila S2E enhancer reveals accessible evolutionary paths through compensatory change. Mol Biol Evol. PubMed ID: 24408913

    Martinez, C. A., Barr, K., Kim, A. R. and Reinitz, J. (2013). A synthetic biology approach to the development of transcriptional regulatory models and custom enhancer design. Methods. PubMed ID: 23732772

    Surkova, S., Myasnikova, E., Kozlov, K. N., Pisarev, A., Reinitz, J. and Samsonova, M. (2013). Quantitative imaging of gene expression in Drosophila embryos. Cold Spring Harb Protoc 2013. PubMed ID: 23734022

    Kim, A. R., Martinez, C., Ionides, J., Ramos, A. F., Ludwig, M. Z., Ogawa, N., Sharp, D. H., Reinitz, J. (2013) Rearrangements of 2.5 Kilobases of Noncoding DNA from the Drosophila even-skipped Locus Define Predictive Rules of Genomic cis-Regulatory Logic. PLoS Genet 9: e1003243. Pubmed ID: 23468638

    Surkova, S., Golubkova, E., Manu, Panok, L., Mamon, L., Reinitz, J., Samsonova, M. (2013) Quantitative dynamics and increased variability of segmentation gene expression in the Drosophila Kruppel and knirps mutants. Dev Biol. PubMed ID: 23333947

  • Michael Reiser HHMI, Janelia Farm
    Kind, E., Longden, K. D., Nern, A., Zhao, A., Sancer, G., Flynn, M. A., Laughland, C. W., Gezahegn, B., Ludwig, H. D., Thomson, A. G., Obrusnik, T., Alarcon, P. G., Dionne, H., Bock, D. D., Rubin, G. M., Reiser, M. B. and Wernet, M. F. (2021). Synaptic targets of photoreceptors specialized to detect color and skylight polarization in Drosophila. Elife 10. PubMed ID: 34913436

    Gruntman, E., Reimers, P., Romani, S. and Reiser, M. B. (2021). Non-preferred contrast responses in the Drosophila motion pathways reveal a receptive field structure that explains a common visual illusion. Curr Biol. PubMed ID: 34672960

    Morimoto, M. M., Nern, A., Zhao, A., Rogers, E. M., Wong, A., Isaacson, M. D., Bock, D., Rubin, G. M. and Reiser, M. B. (2020). Spatial readout of visual looming in the central brain of Drosophila. Elife 9. PubMed ID: 33205753

    Gruntman, E., Romani, S. and Reiser, M. B. (2019). The computation of directional selectivity in the Drosophila OFF motion pathway. Elife 8. PubMed ID: 31825313

    Strother, J. A., Wu, S. T., Rogers, E. M., Eliason, J. L. M., Wong, A. M., Nern, A. and Reiser, M. B. (2018). Behavioral state modulates the ON visual motion pathway of Drosophila. Proc Natl Acad Sci U S A 115(1): E102-e111. PubMed ID: 29255026

    Gruntman, E., Romani, S. and Reiser, M. B. (2018). Simple integration of fast excitation and offset, delayed inhibition computes directional selectivity in Drosophila. Nat Neurosci. PubMed ID: 29311742

    Robie, A. A., Hirokawa, J., Edwards, A. W., Umayam, L. A., Lee, A., Phillips, M. L., Card, G. M., Korff, W., Rubin, G. M., Simpson, J. H., Reiser, M. B. and Branson, K. (2017). Mapping the Neural Substrates of Behavior. Cell 170(2): 393-406.e328. PubMed ID: 28709004

    Strother, J. A., Wu, S. T., Wong, A. M., Nern, A., Rogers, E. M., Le, J. Q., Rubin, G. M. and Reiser, M. B. (2017). The emergence of directional selectivity in the visual motion pathway of Drosophila. Neuron 94(1): 168-182.e110. PubMed ID: 28384470

    Tuthill, J. C., Nern, A., Rubin, G. M. and Reiser, M. B. (2014). Wide-field feedback neurons dynamically tune early visual processing. Neuron 82: 887-895. PubMed ID: 24853944

    Strother, J. A., Nern, A. and Reiser, M. B. (2014). Direct Observation of ON and OFF Pathways in the Drosophila Visual System. Curr Biol. PubMed ID: 24704075

    Tuthill, J. C., Nern, A., Holtz, S. L., Rubin, G. M. and Reiser, M. B. (2013). Contributions of the 12 neuron classes in the fly lamina to motion vision. Neuron 79: 128-140. PubMed ID: 23849200

    Reiser, M. B. and Dickinson, M. H. (2013). Visual motion speed determines a behavioral switch from forward flight to expansion avoidance in Drosophila. J Exp Biol 216: 719-732. PubMed ID: 23197097

  • Lawrence T. Reiter Neurosciences Institute, University of Tennessee, Memphis
    Hope, K. A., Johnson, D., Miller, P. W., Lopez-Ferrer, D., Kakhniashvili, D. and Reiter, L. T. (2020). Transcriptomic and proteomic profiling of glial versus neuronal Dube3a overexpression reveals common molecular changes in gliopathic epilepsies. Neurobiol Dis: 104879. PubMed ID: 32344153

    Hope, K. A., Flatten, D., Cavitch, P., May, B., Sutcliffe, J. S., O'Donnell, J. and Reiter, L. T. (2019). The Drosophila gene Sulfateless modulates autism-like behaviors. Front Genet 10: 574. PubMed ID: 31316544

    Hope, K. A., McGinn, A. and Reiter, L. T. (2019). A genome-wide enhancer/suppressor screen for Dube3a interacting genes in Drosophila melanogaster. Sci Rep 9(1): 2382. PubMed ID: 30787400

    Hope, K. A., LeDoux, M. S. and Reiter, L. T. (2017). Glial overexpression of Dube3a causes seizures and synaptic impairments in Drosophila concomitant with down regulation of the Na+/K+ pump ATPalpha. Neurobiol Dis 108: 238-248. PubMed ID: 28888970

    Valdez, C., Scroggs, R., Chassen, R. and Reiter, L. T. (2015). Variation in Dube3a expression affects neurotransmission at the Drosophila neuromuscular junction. Biol Open. PubMed ID: 25948754

    Hatfield, I., Harvey, I., Yates, E.R., Redd, J.R., Reiter, L.T. and Bridges, D. (2015). The role of TORC1 in muscle development in Drosophila. Sci Rep 5: 9676. PubMed ID: 25866192

    Jensen, L., Farook, M. F., Reiter, L. T. (2013) Proteomic profiling in Drosophila reveals potential dube3a regulation of the actin cytoskeleton and neuronal homeostasis. PLoS One 8: e61952. PubMed ID: 23626758

    Wangler, M. F., Reiter, L. T., Zimm, G., Trimble-Morgan, J., Wu, J. and Bier, E. (2011). Antioxidant proteins TSA and PAG interact synergistically with Presenilin to modulate Notch signaling in Drosophila. Protein Cell 2: 554-563. PubMed ID: 21822800

    Ferdousy, F., Bodeen, W., Summers, K., Doherty, O., Wright, O., Elsisi, N., Hilliard, G., O'Donnell, J. M. and Reiter, L. T. (2011). Drosophila Ube3a regulates monoamine synthesis by increasing GTP cyclohydrolase I activity via a non-ubiquitin ligase mechanism. Neurobiol Dis 41: 669-677. PubMed ID: 21147225

  • Andrew Renault Faculty of Medicine & Health Sciences, University of Nottingham
    Kenwrick, K., Mukherjee, A. and Renault, A. D. (2019). Hmgcr promotes a long-range signal to attract Drosophila germ cells independently of Hedgehog. J Cell Sci 132(23). PubMed ID: 31719159

    Kenwrick, K., Mukherjee, A. and Renault, A. D. (2019). Hmgcr promotes a long-range signal to attract germ cells independent of Hedgehog. J Cell Sci. PubMed ID: 31719159

    Tripathy, R., Kunwar, P. S., Sano, H. and Renault, A. D. (2014). Transcriptional regulation of Drosophila gonad formation. Dev Biol. PubMed ID: 24927896

    Mukherjee, A., Neher, R. A. and Renault, A. D. (2013). Quantifying the range of a lipid phosphate signal in vivo. J Cell Sci. PubMed ID: 24006260

    Sano, H., Kunwar, P. S., Renault, A. D., Barbosa, V., Clark, I. B., Ishihara, S., Sugimura, K. and Lehmann, R. (2012). The Drosophila actin regulator ENABLED regulates cell shape and orientation during gonad morphogenesis. PLoS One 7: e52649. PubMed ID: 23300733

    Renault, A. D. (2012). vasa is expressed in somatic cells of the embryonic gonad in a sex-specific manner in Drosophila melanogaster. Biol Open 1: 1043-1048. PubMed ID: 23213382

    Ile, K. E., Tripathy, R., Goldfinger, V. and Renault, A. D. (2012). Wunen, a Drosophila lipid phosphate phosphatase, is required for septate junction-mediated barrier function. Development 139: 2535-2546. PubMed ID: 22675212

  • Renate Renkawitz-Pohl Philipps-Universität Marburg
    Bischoff, M. C., Lieb, S., Renkawitz-Pohl, R. and Bogdan, S. (2021). Filopodia-based contact stimulation of cell migration drives tissue morphogenesis. Nat Commun 12(1): 791. PubMed ID: 33542237

    Hundertmark, T., Kreutz, S., Merle, N., Nist, A., Lamp, B., Stiewe, T., Brehm, A., Renkawitz-Pohl, R. and Rathke, C. (2019). Drosophila melanogaster tPlus3a and tPlus3b ensure full male fertility by regulating transcription of Y-chromosomal, seminal fluid, and heat shock genes. PLoS One 14(3): e0213177. PubMed ID: 30845228

    Gartner, S. M. K., Hundertmark, T., Nolte, H., Theofel, I., Eren-Ghiani, Z., Tetzner, C., Duchow, T. B., Rathke, C., Kruger, M. and Renkawitz-Pohl, R. (2019). Stage-specific testes proteomics of Drosophila melanogaster identifies essential proteins for male fertility. Eur J Cell Biol. PubMed ID: 30679029

    Hundertmark, T., Gartner, S. M. K., Rathke, C. and Renkawitz-Pohl, R. (2018). Nejire/dCBP-mediated histone H3 acetylation during spermatogenesis is essential for male fertility in Drosophila melanogaster. PLoS One 13(9): e0203622. PubMed ID: 30192860

    Rothenbusch-Fender, S., Fritzen, K., Bischoff, M. C., Buttgereit, D., Oenel, S. F. and Renkawitz-Pohl, R. (2017). Myotube migration to cover and shape the testis of Drosophila depends on Heartless, Cadherin/Catenin, and myosin II. Biol Open. PubMed ID: 29122742

    Jox, T., Buxa, M. K., Bohla, D., Ullah, I., Macinkovic, I., Brehm, A., Bartkuhn, M. and Renkawitz, R. (2017). Drosophila CP190- and dCTCF-mediated enhancer blocking is augmented by SUMOylation. Epigenetics Chromatin 10: 32. PubMed ID: 28680483

    Ali, T., Kruger, M., Bhuju, S., Jarek, M., Bartkuhn, M. and Renkawitz, R. (2016). Chromatin binding of Gcn5 in Drosophila is largely mediated by CP190. Nucleic Acids Res. PubMed ID: 27903907 Eren-Ghiani, Z., Rathke, C., Theofel, I. and Renkawitz-Pohl, R. (2015). Prtl99C acts together with protamines and safeguards male fertility in Drosophila. Cell Rep 13: 2327-2335. PubMed ID: 26673329

    Kuckwa, J., Fritzen, K., Buttgereit, D., Rothenbusch-Fender, S. and Renkawitz-Pohl, R. (2015). A new level of plasticity: Drosophila smooth-like testes muscles compensate failure of myoblast fusion. Development. PubMed ID: 26657767

    Kost, N., Kaiser, S., Ostwal, Y., Riedel, D., Stützer, A., Nikolov, M., Rathke, C., Renkawitz-Pohl, R. and Fischle, W. (2015). Multimerization of Drosophila sperm protein Mst77F causes a unique condensed chromatin structure. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 25735749

    Gartner, S. M., Rothenbusch, S., Buxa, M. K., Theofel, I., Renkawitz, R., Rathke, C. and Renkawitz-Pohl, R. (2014). The HMG-box-containing proteins tHMG-1 and tHMG-2 interact during the histone-to-protamine transition in Drosophila spermatogenesis. Eur J Cell Biol. PubMed ID: 25464903

    Theofel, I., Bartkuhn, M., Hundertmark, T., Boettger, T., Gartner, S. M., Leser, K., Awe, S., Schipper, M., Renkawitz-Pohl, R. and Rathke, C. (2014). tBRD-1 Selectively Controls Gene Activity in the Drosophila Testis and Interacts with Two New Members of the Bromodomain and Extra-Terminal (BET) Family. PLoS One 9: e108267. PubMed ID: 25251222

  • Gunter Reuter Institute of Genetics Martin-Luther Universitat, Halle Germany
    Walther, M., Schrahn, S., Krauss, V., Lein, S., Kessler, J., Jenuwein, T. and Reuter, G. (2020). Heterochromatin formation in Drosophila requires genome-wide histone deacetylation in cleavage chromatin before mid-blastula transition in early embryogenesis. Chromosoma 129(1): 83-98. PubMed ID: 31950239

    Dorafshan, E., Kahn, T. G., Glotov, A., Savitsky, M., Walther, M., Reuter, G. and Schwartz, Y. B. (2019). Ash1 counteracts Polycomb repression independent of histone H3 lysine 36 methylation. EMBO Rep. PubMed ID: 30833342

    Ost, A., Lempradl, A., Casas, E., Weigert, M., Tiko, T., Deniz, M., Pantano, L., Boenisch, U., Itskov, P. M., Stoeckius, M., Ruf, M., Rajewsky, N., Reuter, G., Iovino, N., Ribeiro, C., Alenius, M., Heyne, S., Vavouri, T. and Pospisilik, J. A. (2014). Paternal diet defines offspring chromatin state and intergenerational obesity. Cell 159: 1352-1364. PubMed ID: 25480298

    Koch, B., Kolenko, P., Buchholz, M., Carrillo, D. R., Parthier, C., Wermann, M., Rahfeld, J. U., Reuter, G., Schilling, S., Stubbs, M. T. and Demuth, H. U. (2012). Crystal structures of glutaminyl cyclases (QCs) from Drosophila melanogaster reveal active site conservation between insect and mammalian QCs. Biochemistry 51: 7383-7392. PubMed ID: 22897232

    Krauss, V. and Reuter, G. (2011). DNA methylation in Drosophila--a critical evaluation. Prog Mol Biol Transl Sci 101: 177-191. PubMed ID: 21507351

    Koryakov, D. E., Walther, M., Ebert, A., Lein, S., Zhimulev, I. F. and Reuter, G. (2011). The SUUR protein is involved in binding of SU(VAR)3-9 and methylation of H3K9 and H3K27 in chromosomes of Drosophila melanogaster. Chromosome Res 19: 235-249. PubMed ID: 21340745

  • Max Reuter Research Department of Genetics, Evolution and Environment, Faculty of Life Sciences, University College London
    Camus, M. F., Moore, J. and Reuter, M. (2020). Nutritional geometry of mitochondrial genetic effects on male fertility. Biol Lett 16(2): 20190891. PubMed ID: 32097597

    Camus, M. F., Piper, M. D. and Reuter, M. (2019). Sex-specific transcriptomic responses to changes in the nutritional environment. Elife 8. PubMed ID: 31436529

    Ruzicka, F., Hill, M. S., Pennell, T. M., Flis, I., Ingleby, F. C., Mott, R., Fowler, K., Morrow, E. H. and Reuter, M. (2019). Genome-wide sexually antagonistic variants reveal long-standing constraints on sexual dimorphism in fruit flies. PLoS Biol 17(4): e3000244. PubMed ID: 31022179

    Camus, M. F., Fowler, K., Piper, M. W. D. and Reuter, M. (2017). Sex and genotype effects on nutrient-dependent fitness landscapes in Drosophila melanogaster. Proc Biol Sci 284(1869). PubMed ID: 29263276

    Collet, J. M., Fuentes, S., Hesketh, J., Hill, M. S., Innocenti, P., Morrow, E. H., Fowler, K. and Reuter, M. (2016). Rapid evolution of the intersexual genetic correlation for fitness in Drosophila melanogaster. Evolution 70: 781-795. PubMed ID: 27077679

    Hesketh, J., Fowler, K. and Reuter, M. (2013). Genetic drift in antagonistic genes leads to divergence in sex-specific fitness between experimental populations of Drosophila melanogaster. Evolution 67: 1503-1510. PubMed ID: 23617925

    Mullon, C., Pomiankowski, A. and Reuter, M. (2012). Molecular evolution of Drosophila Sex-lethal and related sex determining genes. BMC Evol Biol 12: 5. PubMed ID: 22244243

  • Rolf Reuter Division Animal Genetics, Universitat Tubingen
    Spahn, P. and Reuter, R. (2013). A vertex model of Drosophila ventral furrow formation. PLoS One 8: e75051. PubMed ID: 24066163

    Comber, K., Huelsmann, S., Evans, I., Sanchez-Sanchez, B. J., Chalmers, A., Reuter, R., Wood, W. and Martin-Bermudo, M. D. (2013). A dual role for the betaPS integrin myospheroid in mediating Drosophila embryonic macrophage migration. J Cell Sci 126: 3475-3484. PubMed ID: 23704353

    Spahn, P., Ott, A. and Reuter, R. (2012). The PDZ-GEF protein Dizzy regulates the establishment of adherens junctions required for ventral furrow formation in Drosophila. J Cell Sci 125: 3801-3812. PubMed ID: 22553205

  • Kim Rewitz Department of Biology, Cell and Neurobiology, Copenhaven
    Kubrak, O., Koyama, T., Ahrentløv, N., Jensen, L., Malita, A., Naseem, M. T., Lassen, M., Nagy, S., Texada, M. J., Halberg, K. V. and Rewitz, K. (2022). The gut hormone Allatostatin C/Somatostatin regulates food intake and metabolic homeostasis under nutrient stress. Nat Commun 13(1): 692. PubMed ID: 35121731

    Texada, M. J., Lassen, M., Pedersen, L. H., Koyama, T., Malita, A. and Rewitz, K. (2022). Insulin signaling couples growth and early maturation to cholesterol intake in Drosophila. Curr Biol 32(7): 1548-1562.e1546. PubMed ID: 35245460

    Christensen, C. F., Koyama, T., Nagy, S., Danielsen, E. T., Texada, M. J., Halberg, K. A. and Rewitz, K. (2020). Ecdysone-dependent feedback regulation of prothoracicotropic hormone controls the timing of developmental maturation. Development 147(14). PubMed ID: 32631830

    Texada, M. J., Jorgensen, A. F., Christensen, C. F., Koyama, T., Malita, A., Smith, D. K., Marple, D. F. M., Danielsen, E. T., Petersen, S. K., Hansen, J. L., Halberg, K. A. and Rewitz, K. F. (2019). A fat-tissue sensor couples growth to oxygen availability by remotely controlling insulin secretion. Nat Commun 10(1): 1955. PubMed ID: 31028268

    Texada, M. J., Malita, A., Christensen, C. F., Dall, K. B., Faergeman, N. J., Nagy, S., Halberg, K. A. and Rewitz, K. (2019). Autophagy-mediated cholesterol trafficking controls steroid production. Dev Cell 48(5): 659-671.e654. PubMed ID: 30799225

    Moeller, M. E., Nagy, S., Gerlach, S. U., Soegaard, K. C., Danielsen, E. T., Texada, M. J. and Rewitz, K. F. (2017). Warts signaling controls organ and body Growth through regulation of ecdysone. Curr Biol. PubMed ID: 28528906

    Danielsen, E. T., Moeller, M. E., Yamanaka, N., Ou, Q., Laursen, J. M., Soenderholm, C., Zhuo, R., Phelps, B., Tang, K., Zeng, J., Kondo, S., Nielsen, C. H., Harvald, E. B., Faergeman, N. J., Haley, M. J., O'Connor, K. A., King-Jones, K., O'Connor, M. B. and Rewitz, K. F. (2016). A Drosophila genome-wide screen identifies regulators of steroid hormone production and developmental timing. Dev Cell 37: 558-570. PubMed ID: 27326933

    Hentze, J. L., Carlsson, M. A., Kondo, S., Nassel, D. R. and Rewitz, K. F. (2015). The Neuropeptide Allatostatin A Regulates Metabolism and Feeding Decisions in Drosophila. Sci Rep 5: 11680. PubMed ID: 26123697

    Danielsen, E. T., Moeller, M. E., Dorry, E., Komura-Kawa, T., Fujimoto, Y., Troelsen, J. T., Herder, R., O'Connor, M. B., Niwa, R. and Rewitz, K. F. (2014). Transcriptional control of steroid biosynthesis genes in the Drosophila prothoracic gland by ventral veins lacking and knirps. PLoS Genet 10: e1004343. PubMed ID: 24945799

    Moeller, M. E., Danielsen, E. T., Herder, R., O'Connor, M. B. and Rewitz, K. F. (2013). Dynamic feedback circuits function as a switch for shaping a maturation-inducing steroid pulse in Drosophila. Development. PubMed ID: 24173800

  • Enrique Reynaud Genética del Desarrollo y Fisiología Molecular, Cuernavaca, México
    Carvajal-Oliveros, A., Domínguez-Baleon, C., Zarate, R. V., Campusano, J. M., Narvaez-Padilla, V. and Reynaud, E. (2021). Nicotine suppresses Parkinson's disease like phenotypes induced by Synphilin-1 overexpression in Drosophila melanogaster by increasing tyrosine hydroxylase and dopamine levels. Sci Rep 11(1): 9579. PubMed ID: 33953275

    Sanchez-Diaz, I., Rosales-Bravo, F., Reyes-Taboada, J. L., Covarrubias, A. A., Narvaez-Padilla, V. and Reynaud, E. (2015). The Esg Gene Is Involved in Nicotine Sensitivity in Drosophila melanogaster. PLoS One 10: e0133956. PubMed ID: 26222315

    Reynaud, E., Lahaye, L. L., Boulanger, A., Petrova, I. M., Marquilly, C., Flandre, A., Martianez, T., Privat, M., Noordermeer, J. N., Fradkin, L. G. and Dura, J. M. (2015). Guidance of Drosophila Mushroom Body Axons Depends upon DRL-Wnt Receptor Cleavage in the Brain Dorsomedial Lineage Precursors. Cell Rep 11: 1293-1304. PubMed ID: 25981040

    Besson, C., Bernard, F., Corson, F., Rouault, H., Reynaud, E., Keder, A., Mazouni, K. and Schweisguth, F. (2015). Planar Cell Polarity Breaks the Symmetry of PAR Protein Distribution prior to Mitosis in Drosophila Sensory Organ Precursor Cells. Curr Biol 25: 1104-1110. PubMed ID: 25843034

    Soldano, A., Okray, Z., Janovska, P., Tmejova, K., Reynaud, E., Claeys, A., Yan, J., Atak, Z. K., De Strooper, B., Dura, J. M., Bryja, V. and Hassan, B. A. (2013). The Drosophila homologue of the amyloid precursor protein is a conserved modulator of Wnt PCP signaling. PLoS Biol 11: e1001562. PubMed ID: 23690751

  • Carlos Ribeiro Champalimaud Neuroscience Programme, Lisbon
    Carvalho-Santos, Z., Cardoso-Figueiredo, R., Elias, A. P., Tastekin, I., Baltazar, C. and Ribeiro, C. (2020). Cellular metabolic reprogramming controls sugar appetite in Drosophila. Nat Metab 2(9): 958-973. PubMed ID: 32868922

    Steck, K., Walker, S. J., Itskov, P. M., Baltazar, C., Moreira, J. M. and Ribeiro, C. (2018). Internal amino acid state modulates yeast taste neurons to support protein homeostasis in Drosophila. Elife 7. PubMed ID: 29393045

    Leitão-Gonçalves, R., Carvalho-Santos, Z., Francisco, A.P., Fioreze, G.T., Anjos, M., Baltazar, C., Elias, A.P., Itskov, P.M., Piper, M.D.W. and Ribeiro, C. (2017). Commensal bacteria and essential amino acids control food choice behavior and reproduction. PLoS Biol 15: e2000862. PubMed ID: 28441450

    Walker, S.J., Corrales-Carvajal, V.M. and Ribeiro, C. (2015). Postmating circuitry modulates salt taste processing to increase reproductive output in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26412135

    Itskov, P. M. and Ribeiro, C. (2013). The dilemmas of the gourmet fly: the molecular and neuronal mechanisms of feeding and nutrient decision making in Drosophila. Front Neurosci 7: 12. PubMed ID: 23407678

    Ribeiro, C. and Dickson, B. J. (2010). Sex peptide receptor and neuronal TOR/S6K signaling modulate nutrient balancing in Drosophila. Curr Biol 20: 1000-1005. PubMed ID: 20471268

  • Rob Richards Molecular Genetics, University of Adelaide
    Choo, A., O'Keefe, L. V., Lee, C. S., Gregory, S. L., Shaukat, Z., Colella, A., Lee, K., Denton, D. and Richards, R. I. (2015). Tumor suppressor WWOX moderates the mitochondrial respiratory complex. Genes Chromosomes Cancer. PubMed ID: 26390919

    O'Keefe, L. V., Lee, C. S., Choo, A. and Richards, R. I. (2015). Tumor Suppressor WWOX Contributes to the Elimination of Tumorigenic Cells in Drosophila melanogaster. PLoS One 10: e0136356. PubMed ID: 26302329

    Samaraweera, S. E., O'Keefe, L. V., van Eyk, C. L., Lawlor, K. T., Humphreys, D. T., Suter, C. M., Richards, R. I. (2013) Modeling and Analysis of Repeat RNA Toxicity in Drosophila. Methods Mol Biol 1017: 173-192. PubMed ID: 23719916

    Samaraweera, S. E., O'Keefe, L. V., Price, G. R., Venter, D. J., Richards, R. I. (2013) Distinct roles for Toll and autophagy pathways in double-stranded RNA toxicity in a Drosophila model of expanded repeat neurodegenerative diseases. Hum Mol Genet. PubMed ID: 23525903

    Lawlor, K. T., O'Keefe, L. V., Samaraweera, S. E., van Eyk, C. L. and Richards, R. I. (2012). Ubiquitous expression of CUG or CAG trinucleotide repeat RNA causes common morphological defects in a Drosophila model of RNA-mediated pathology. PLoS One 7: e38516. PubMed ID: 22715390

    van Eyk, C. L., McLeod, C. J., O'Keefe, L. V. and Richards, R. I. (2012). Comparative toxicity of polyglutamine, polyalanine and polyleucine tracts in Drosophila models of expanded repeat disease. Hum Mol Genet 21: 536-547. PubMed ID: 22021427

  • Helena Richardson La Trobe University, Australia
    La Marca, J. E., Willoughby, L. F., Allan, K., Portela, M., Goh, P. K., Tiganis, T. and Richardson, H. E. (2021). PTP61F Mediates Cell Competition and Mitigates Tumorigenesis. Int J Mol Sci 22(23). PubMed ID: 34884538

    La Marca, J. E. and Richardson, H. E. (2020). Two-Faced: Roles of JNK Signalling During Tumourigenesis in the Drosophila Model. Front Cell Dev Biol 8: 42. PubMed ID: 32117973

    La Marca, J. E., Diepstraten, S. T., Hodge, A., Wang, H., Hart, A. H., Richardson, H. E. and Somers, W. G. (2019). Strip and Cka negatively regulate JNK signalling during Drosophila spermatogenesis. Development. PubMed ID: 31164352

    Roesley, S. N. A., La Marca, J. E., Deans, A. J., McKenzie, L., Suryadinata, R., Burke, P., Portela, M., Wang, H., Bernard, O., Sarcevic, B. and Richardson, H. E. (2018). Phosphorylation of Drosophila Brahma on CDK-phosphorylation sites is important for cell cycle regulation and differentiation. Cell Cycle: 1-20. PubMed ID: 29963966

    Portela, M., Yang, L., Paul, S., Li, X., Veraksa, A., Parsons, L. M. and Richardson, H. E. (2018). Lgl reduces endosomal vesicle acidification and Notch signaling by promoting the interaction between Vap33 and the V-ATPase complex. Sci Signal 11(533). PubMed ID: 29871910

    Poon, C. L. C., Brumby, A. M. and Richardson, H. E. (2018). Src cooperates with oncogenic Ras in tumourigenesis via the JNK and PI3K pathways in Drosophila epithelial Tissue. Int J Mol Sci 19(6). PubMed ID: 29861494

    Caria, S., Magtoto, C. M., Samiei, T., Portela, M., Lim, K. Y. B., How, J. Y., Stewart, B. Z., Humbert, P. O., Richardson, H. E. and Kvansakul, M. (2018). Drosophila melanogaster Guk-holder interacts with the Scribbled PDZ1 domain and regulates epithelial development with Scribbled and Discs Large. J Biol Chem. PubMed ID: 29378849

    Parsons, L. M., Grzeschik, N. A., Amaratunga, K., Burke, P., Quinn, L. M. and Richardson, H. E. (2017). A kinome RNAi screen in Drosophila identifies novel genes interacting with Lgl, aPKC and Crb cell polarity genes in epithelial tissues. G3 (Bethesda). PubMed ID: 28611255

    Willoughby, L. F., Manent, J., Allan, K., Lee, H., Portela, M., Wiede, F., Warr, C., Meng, T. C., Tiganis, T. and Richardson, H. E. (2017). Differential regulation of protein tyrosine kinase signaling by Dock and the PTP61F variants. FEBS J. PubMed ID: 28544778

    Doggett, K., Turkel, N., Willoughby, L. F., Ellul, J., Murray, M. J., Richardson, H. E. and Brumby, A. M. (2015). BTB-zinc finger oncogenes are required for Ras and Notch-driven tumorigenesis in Drosophila. PLoS One 10: e0132987. PubMed ID: 26207831

  • Lynn Riddiford Friday Harbor Laboratories, University of Washington, Friday Harbor, WA
    Truman, J. W. and Riddiford, L. M. (2022). Chinmo is the larval member of the molecular trinity that directs Drosophila metamorphosis. Proc Natl Acad Sci U S A 119(15): e2201071119. PubMed ID: 35377802

    Riddiford, L. M., Truman, J. W. and Nern, A. (2018). Juvenile hormone reveals mosaic developmental programs in the metamorphosing optic lobe of Drosophila melanogaster. Biol Open 7(4). PubMed ID: 29618455

    Baumann, A. A., Texada, M. J., Chen, H. M., Etheredge, J. N., Miller, D. L., Picard, S., Warner, R., Truman, J. W. and Riddiford, L. M. (2017). Genetic tools to study juvenile hormone action in Drosophila. Sci Rep 7(1): 2132. PubMed ID: 28522854

    Mirth, C. K., Tang, H. Y., Makohon-Moore, S. C., Salhadar, S., Gokhale, R. H., Warner, R. D., Koyama, T., Riddiford, L. M. and Shingleton, A. W. (2014). Juvenile hormone regulates body size and perturbs insulin signaling in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 24778227

    Bilen, J., Atallah, J., Azanchi, R., Levine, J. D. and Riddiford, L. M. (2013). Regulation of onset of female mating and sex pheromone production by juvenile hormone in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 24145432

    Harshman, L. G., Song, K. D., Casas, J., Schuurmans, A., Kuwano, E., Kachman, S. D., Riddiford, L. M. and Hammock, B. D. (2010). Bioassays of compounds with potential juvenoid activity on Drosophila melanogaster: juvenile hormone III, bisepoxide juvenile hormone III and methyl farnesoates. J Insect Physiol 56: 1465-1470. PubMed ID: 20599543

    Riddiford, L. M., Truman, J. W., Mirth, C. K. and Shen, Y. C. (2010). A role for juvenile hormone in the prepupal development of Drosophila melanogaster. Development 137: 1117-1126. PubMed ID: 20181742

  • Nicole C. Riddle Department of Biology, University of Alabama, Birmingham
    Schoelz, J. M., Feng, J. X. and Riddle, N. C. (2021). The Drosophila HP1 family is associated with active gene expression across chromatin contexts. Genetics 219(1). PubMed ID: 34849911

    Riddle, N. C. (2020). Variation in the response to exercise stimulation in Drosophila: marathon runner versus sprinter genotypes. J Exp Biol 223(Pt 18). PubMed ID: 32737212

    Watanabe, L. P., Gordon, C., Momeni, M. Y. and Riddle, N. C. (2020). Genetic Networks Underlying Natural Variation in Basal and Induced Activity Levels in Drosophila melanogaster. G3 (Bethesda) 10(4): 1247-1260. PubMed ID: 32014853

    Watanabe, L. P. and Riddle, N. C. (2019). New opportunities: Drosophila as a model system for exercise research. J Appl Physiol (1985) 127(2): 482-490. PubMed ID: 31268829

    Mills, B. B., Thomas, A. D. and Riddle, N. C. (2018). HP1B is a euchromatic Drosophila HP1 homolog with links to metabolism. PLoS One 13(10): e0205867. PubMed ID: 30346969

  • Elizabeth Rideout Dept. of Cellular and Physiological Sciences, University of British Columbia,
    Millington, J. W., Biswas, P., Chao, C., Xia, Y. H., Wat, L. W., Brownrigg, G. P., Sun, Z., Basner-Collins, P. J., Klein Geltink, R. I. and Rideout, E. J. (2022). A low-sugar diet enhances Drosophila body size in males and females via sex-specific mechanisms. Development 149(6). PubMed ID: 35195254

    Wat, L. W., Chowdhury, Z. S., Millington, J. W., Biswas, P. and Rideout, E. J. (2021). Sex determination gene transformer regulates the male-female difference in Drosophila fat storage via the adipokinetic hormone pathway. Elife 10. PubMed ID: 34672260

    Millington, J. W., Brownrigg, G. P., Basner-Collins, P. J., Sun, Z. and Rideout, E. J. (2021). Genetic manipulation of insulin/insulin-like growth factor signaling pathway activity has sex-biased effects on Drosophila body size. G3 (Bethesda) 11(3). PubMed ID: 33793746

    Wat, L. W., Chao, C., Bartlett, R., Buchanan, J. L., Millington, J. W., Chih, H. J., Chowdhury, Z. S., Biswas, P., Huang, V., Shin, L. J., Wang, L. C., Gauthier, M. L., Barone, M. C., Montooth, K. L., Welte, M. A. and Rideout, E. J. (2020). A role for triglyceride lipase brummer in the regulation of sex differences in Drosophila fat storage and breakdown. PLoS Biol 18(1): e3000595. PubMed ID: 31961851

    Rideout, E. J., Narsaiya, M. S. and Grewal, S. S. (2015). The Sex Determination Gene transformer Regulates Male-Female Differences in Drosophila Body Size. PLoS Genet 11(12): e1005683. PubMed ID: 26710087

    Ghosh, A., Rideout, E. J. and Grewal, S. S. (2014). TIF-IA-dependent regulation of ribosome synthesis in Drosophila muscle is required to maintain systemic insulin signaling and larval growth. PLoS Genet 10(10): e1004750. PubMed ID: 25356674

  • Veit Riechmann Cell and Molecular Biology, University of Heidelberg
    Laiouar, S., Berns, N., Brech, A. and Riechmann, V. (2020). RabX1 Organizes a Late Endosomal Compartment that Forms Tubular Connections to Lysosomes Consistent with a "Kiss and Run" Mechanism. Curr Biol. PubMed ID: 32059769

    Woichansky, I., Beretta, C. A., Berns, N. and Riechmann, V. (2016). Three mechanisms control E-cadherin localization to the zonula adherens. Nat Commun 7: 10834. PubMed ID: 26960923

    Berns, N., Woichansky, I., Friedrichsen, S., Kraft, N. and Riechmann, V. (2014). A genome-scale in vivo RNAi analysis of epithelial development in Drosophila identifies new proliferation domains outside of the stem cell niche. J Cell Sci. PubMed ID: 24762813

    Gomez, J. M., Wang, Y. and Riechmann, V. (2012). Tao controls epithelial morphogenesis by promoting Fasciclin 2 endocytosis. J Cell Biol 199: 1131-1143. PubMed ID: 23266957

    Berns, N., Woichansky, I., Kraft, N., Husken, U., Carl, M. and Riechmann, V. (2012). "Vacuum-assisted staining": a simple and efficient method for screening in Drosophila. Dev Genes Evol 222: 113-118. PubMed ID: 22367217

    JayaNandanan, N., Gavis, E. R., Riechmann, V. and Leptin, M. (2011). A genetic in vivo system to detect asymmetrically distributed RNA. EMBO Rep 12: 1167-1174. PubMed ID: 21921935

  • Juan Riesgo-Escovar Instituto de Neurobiologia,Universidad Nacional Autonoma de México, Querétaro, México
    Alvarez-Rendon, J. P. and Riesgo-Escovar, J. R. (2020). Circadian and Rhythmic-Related Behavioral Co-Morbidities of the Diabetic State in Drosophila melanogaster. Gen Comp Endocrinol: 113477. PubMed ID: 32240709

    Mendoza-Ortiz, M. A., Murillo-Maldonado, J. M. and Riesgo-Escovar, J. R. (2018). aaquetzalli is required for epithelial cell polarity and neural tissue formation in Drosophila. PeerJ 6: e5042. PubMed ID: 29942698

    Nazario-Yepiz, N. O. and Riesgo-Escovar, J. R. (2016). piraguaen codes a zinc finger protein required for development in Drosophila. Mech Dev [Epub ahead of print]. PubMed ID: 28011160

    González-Morales, N., Mendoza-Ortíz, M.Á., Blowes, L.M., Missirlis, F. and Riesgo-Escovar, J.R. (2015). Ferritin is required in multiple tissues during Drosophila melanogaster development. PLoS One 10: e0133499. PubMed ID: 26192321

    Ríos-Barrera, L.D., Gutiérrez-Pérez, I., Domínguez, M. and Riesgo-Escovar, J.R. (2015). acal is a long non-coding RNA in JNK signaling in epithelial shape changes during Drosophila dorsal closure. PLoS Genet 11: e1004927. PubMed ID: 25710168

    Rios-Barrera, L. D. and Riesgo-Escovar, J. R. (2012). Regulating cell morphogenesis: The Drosophila Jun n-terminal kinase pathway. Genesis. PubMed ID: 23109363

    Murillo-Maldonado, J. M., Zeineddine, F. B., Stock, R., Thackeray, J. and Riesgo-Escovar, J. R. (2011). Insulin receptor-mediated signaling via phospholipase C-gamma regulates growth and differentiation in Drosophila. PLoS One 6: e28067. PubMed ID: 22132213

    Hernandez-Vargas, R., Fonseca-Ornelas, L., Lopez-Gonzalez, I., Riesgo-Escovar, J., Zurita, M. and Reynaud, E. (2011). Synphilin suppresses alpha-synuclein neurotoxicity in a Parkinson's disease Drosophila model. Genesis 49: 392-402. PubMed ID: 21584925

  • Richa Rikhy Indian Institute of Science Education and Research (IISER), Pune
    Dubal, D., Moghe, P., Verma, R. K., Uttekar, B. and Rikhy, R. (2022). Mitochondrial fusion regulates proliferation and differentiation in the type II neuroblast lineage in Drosophila. PLoS Genet 18(2): e1010055. PubMed ID: 35157701

    Sharma, S. and Rikhy, R. (2021). Spatiotemporal recruitment of RhoGTPase protein GRAF inhibits actomyosin ring constriction in Drosophila cellularization. Elife 10. PubMed ID: 33835025

    Chowdhary, S., Madan, S., Tomer, D., Mavrakis, M. and Rikhy, R. (2020). Mitochondrial morphology and activity regulate furrow ingression and contractile ring dynamics in Drosophila cellularization. Mol Biol Cell 31(21): 2331-2347. PubMed ID: 32755438

    Sherlekar, A., Mundhe, G., Richa, P., Dey, B., Sharma, S. and Rikhy, R. (2020). F-BAR domain protein Syndapin regulates actomyosin dynamics during apical cap remodeling in syncytial Drosophila embryos. J Cell Sci. PubMed ID: 32327556

    Dey, B. and Rikhy, R. (2020). DE-cadherin and Myosin II balance regulates furrow length for onset of polygon shape in syncytial Drosophila embryos. J Cell Sci. PubMed ID: 32265269

    Tomer, D., Chippalkatti, R., Mitra, K. and Rikhy, R. (2018). ERK regulates mitochondrial membrane potential in fission deficient Drosophila follicle cells during differentiation. Dev Biol 434(1):48-62. PubMed ID: 29157562

    Chowdhary, S., Tomer, D., Dubal, D., Sambre, D. and Rikhy, R. (2017). Analysis of mitochondrial organization and function in the Drosophila blastoderm embryo. Sci Rep 7(1): 5502. PubMed ID: 27146115 Rikhy, R., Mavrakis, M. and Lippincott-Schwartz, J. (2015). Dynamin regulates metaphase furrow formation and plasma membrane compartmentalization in the syncytial Drosophila embryo. Biol Open 4(3): 301-311. PubMed ID: 25661871


  • Diego Rincon-Limas Department of Neurology, University of Florida, Gainesville
    de Mena, L. and Rincon-Limas, D. E. (2020). PhotoGal4: A Versatile Light-Dependent Switch for Spatiotemporal Control of Gene Expression in Drosophila Explants. iScience 23(7): 101308. PubMed ID: 32652492

    Fernandez-Funez, P., Sanchez-Garcia, J., de Mena, L., Zhang, Y., Levites, Y., Khare, S., Golde, T. E. and Rincon-Limas, D. E. (2016). Holdase activity of secreted Hsp70 masks amyloid-beta42 neurotoxicity in Drosophila. Proc Natl Acad Sci U S A 113: E5212-5221. PubMed ID: 27531960

    Fernandez-Funez, P., Zhang, Y., Sanchez-Garcia, J., de Mena, L., Khare, S., Golde, T. E., Levites, Y. and Rincon-Limas, D. E. (2015). Anti-Aβ single-chain variable fragment antibodies exert synergistic neuroprotective activities in Drosophila models of Alzheimer's disease. Hum Mol Genet. PubMed ID: 26253732

    Zhang, Y., Arcia, S., Perez, B., Fernandez-Funez, P., Rincon-Limas, D. E. (2013) pTubHA4C, a new versatile vector for constitutive expression in Drosophila. Mol Biol Rep. PubMed ID: 23681549

    Jensen, K., Sanchez-Garcia, J., Williams, C., Khare, S., Mathur, K., Graze, R. M., Hahn, D. A., McIntyre, L. M., Rincon-Limas, D. E. and Fernandez-Funez, P. (2013). Purification of transcripts and metabolites from Drosophila heads. J Vis Exp: e50245. PubMed ID: 23524378

    Rincon-Limas, D. E., Jensen, K. and Fernandez-Funez, P. (2012). Drosophila models of proteinopathies: the little fly that could. Curr Pharm Des 18: 1108-1122. PubMed ID: 22288402

  • Leonie Ringrose Institute of Molecular Biotechnology, Vienna
    Steffen, P. A., Altmutter, C., Dworschak, E., Junttila, S., Gyenesei, A., Zhu, X., Kockmann, T. and Ringrose, L. (2021). The Trithorax group protein ASH1 requires a combination of BAH domain and AT hooks, but not the SET domain, for mitotic chromatin binding and survival. Chromosoma. PubMed ID: 34331109

    Reinig, J., Ruge, F., Howard, M. and Ringrose, L. (2020). A theoretical model of Polycomb/Trithorax action unites stable epigenetic memory and dynamic regulation. Nat Commun 11(1): 4782. PubMed ID: 32963223

    Herzog, V. A., Lempradl, A., Trupke, J., Okulski, H., Altmutter, C., Ruge, F., Boidol, B., Kubicek, S., Schmauss, G., Aumayr, K., Ruf, M., Pospisilik, A., Dimond, A., Senergin, H. B., Vargas, M. L., Simon, J. A. and Ringrose, L. (2014). A strand-specific switch in noncoding transcription switches the function of a Polycomb/Trithorax response element. Nat Genet. PubMed ID: 25108384

    Steffen, P. A. and Ringrose, L. (2014). What are memories made of? How Polycomb and Trithorax proteins mediate epigenetic memory. Nat Rev Mol Cell Biol 15: 340-356. PubMed ID: 24755934

    Steffen, P. A., Fonseca, J. P., Ganger, C., Dworschak, E., Kockmann, T., Beisel, C., Ringrose, L. (2013) Quantitative in vivo analysis of chromatin binding of Polycomb and Trithorax group proteins reveals retention of ASH1 on mitotic chromatin. Nucleic Acids Res. PubMed ID: 23580551

    Fonseca, J. P., Steffen, P. A., Muller, S., Lu, J., Sawicka, A., Seiser, C. and Ringrose, L. (2012). In vivo Polycomb kinetics and mitotic chromatin binding distinguish stem cells from differentiated cells. Genes Dev 26: 857-871. PubMed ID: 22508729

    Strubbe, G., Popp, C., Schmidt, A., Pauli, A., Ringrose, L., Beisel, C. and Paro, R. (2011). Polycomb purification by in vivo biotinylation tagging reveals cohesin and Trithorax group proteins as interaction partners. Proc Natl Acad Sci U S A 108: 5572-5577. PubMed ID: 21415365

    Okulski, H., Druck, B., Bhalerao, S. and Ringrose, L. (2011). Quantitative analysis of polycomb response elements (PREs) at identical genomic locations distinguishes contributions of PRE sequence and genomic environment. Epigenetics Chromatin 4: 4. PubMed ID: 21410956

  • Don Rio Molecular and Cell Biology, University of California, Berkeley
    Wang, Q., Abruzzi, K. C., Rosbash, M. and Rio, D. C. (2018). Striking circadian neuron diversity and cycling of Drosophila alternative splicing. Elife 7. PubMed ID: 29863472

    Francis, M. J., Roche, S., Cho, M. J., Beall, E., Min, B., Panganiban, R. P. and Rio, D. C. (2016). Drosophila IRBP bZIP heterodimer binds P-element DNA and affects hybrid dysgenesis. Proc Natl Acad Sci U S A. PubMed ID: 27799520

    Wang, Q., Taliaferro, J.M., Klibaite, U., Hilgers, V., Shaevitz, J.W. and Rio, D.C. (2016). The PSI-U1 snRNP interaction regulates male mating behavior in Drosophila. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27114556

    Rio, D. C. (2014). In vitro splicing reactions in Drosophila kc nuclear extracts. Cold Spring Harb Protoc 2014: pdb prot080853. PubMed ID: 25086019

    Rio, D. C. (2014). Large-scale immunopurification of ribonucleoprotein complexes from Drosophila nucleoplasmic extracts for tiling microarrays. Cold Spring Harb Protoc 2014. PubMed ID: 24591694

    Taliaferro, J. M., Marwha, D., Aspden, J. L., Mavrici, D., Cheng, N. E., Kohlstaedt, L. A., Rio, D. C. (2013) The Drosophila Splicing Factor PSI Is Phosphorylated by Casein Kinase II and Tousled-Like Kinase. PLoS One 8: e56401. PubMed ID: 23437125

    Taliaferro, J. M., Aspden, J. L., Bradley, T., Marwha, D., Blanchette, M. and Rio, D. C. (2013). Two new and distinct roles for Drosophila Argonaute-2 in the nucleus: alternative pre-mRNA splicing and transcriptional repression. Genes Dev. PubMed ID: 23392611

    Brooks, A. N., Aspden, J. L., Podgornaia, A. I., Rio, D. C. and Brenner, S. E. (2011). Identification and experimental validation of splicing regulatory elements in Drosophila melanogaster reveals functionally conserved splicing enhancers in metazoans. RNA 17: 1884-1894. PubMed ID: 21865603

    Taliaferro, J. M., Alvarez, N., Green, R. E., Blanchette, M. and Rio, D. C. (2011). Evolution of a tissue-specific splicing network. Genes Dev 25: 608-620. PubMed ID: 21406555

    Hartmann, B., Castelo, R., Minana, B., Peden, E., Blanchette, M., Rio, D. C., Singh, R. and Valcarcel, J. (2011). Distinct regulatory programs establish widespread sex-specific alternative splicing in Drosophila melanogaster. RNA 17: 453-468. PubMed ID: 21233220

  • Maria Giovanna Riparbelli Dipartimento Di Scienze Della Vita, Siena University
    Persico, V., Callaini, G. and Riparbelli, M. G. (2021). Sas-4 Colocalizes with the Ciliary Rootlets of the Drosophila Sensory Organs. J Dev Biol 9(1). PubMed ID: 33466292

    The Singularity of the Drosophila Male Germ Cell Centriole: The Asymmetric Distribution of Sas4 and Sas6

    Persico, V., Callaini, G. and Riparbelli, M. G. (2019). The microtubule-depolymerizing Kinesin-13 Klp10A is enriched in the transition zone of the ciliary structures of Drosophila melanogaster. Front Cell Dev Biol 7: 173. PubMed ID: 31497602

    Persico, V., Callaini, G. and Riparbelli, M. G. (2019). The male stem cell niche of Drosophila melanogaster: Interactions between the germline stem cells and the hub. Exp Cell Res: 111489. PubMed ID: 31278899

    Riparbelli, M. G., Persico, V. and Callaini, G. (2018). A transient microtubule-based structure uncovers a new intrinsic asymmetry between the mother centrioles in the early Drosophila spermatocytes. Cytoskeleton (Hoboken). PubMed ID: 30381895

    Gottardo, M., Persico, V., Callaini, G. and Riparbelli, M. G. (2018). The "transition zone" of the cilium-like regions in the Drosophila spermatocytes and the role of the C-tubule in axoneme assembly. Exp Cell Res 371(1): 262-268. PubMed ID: 30130520

    Gottardo, M., Callaini, G. and Riparbelli, M. G. (2016). Klp10A modulates the localization of centriole-associated proteins during Drosophila male gametogenesis. Cell Cycle [Epub ahead of print] PubMed ID: 27764551

    Gottardo, M., Callaini, G. and Riparbelli, M. G. (2016). Does Unc-GFP uncover ciliary structures in the rhabdomeric eye of Drosophila? J Cell Sci [Epub ahead of print]. PubMed ID: 27235419

    Fu, J., Lipinszki, Z., Rangone, H., Min, M., Mykura, C., Chao-Chu, J., Schneider, S., Dzhindzhev, N. S., Gottardo, M., Riparbelli, M. G., Callaini, G. and Glover, D. M. (2016). Conserved molecular interactions in centriole-to-centrosome conversion. Nat Cell Biol 18: 87-99. PubMed ID: 26595382

    Gottardo, M., Pollarolo, G., Llamazares, S., Reina, J., Riparbelli, M.G., Callaini, G. and Gonzalez, C. (2015). Loss of Centrobin enables daughter centrioles to form sensory cilia in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26299513

    Gottardo, M., Callaini, G. and Riparbelli, M. G. (2015). The Drosophila centriole: conversion of doublets to triplets within the stem cell niche. J Cell Sci. PubMed ID: 26092937

  • Jens Rister Genetics and Neurobiology, University of Massachusetts, Boston
    Poupault, C., Choi, D., Lam-Kamath, K., Dewett, D., Razzaq, A., Bunker, J., Perry, A., Cho, I. and Rister, J. (2021). A combinatorial cis-regulatory logic restricts color-sensing Rhodopsins to specific photoreceptor subsets in Drosophila. PLoS Genet 17(6): e1009613. PubMed ID: 34161320

    Mishra, A. K., Tsachaki, M., Rister, J., Ng, J., Celik, A. and Sprecher, S. G. (2013). Binary cell fate decisions and fate transformation in the Drosophila larval eye. PLoS Genet 9(12): e1004027. PubMed ID: 24385925

    Thanawala, S. U., Rister, J., Goldberg, G. W., Zuskov, A., Olesnicky, E. C., Flowers, J. M., Jukam, D., Purugganan, M. D., Gavis, E. R., Desplan, C. and Johnston, R. J., Jr. (2013). Regional modulation of a stochastically expressed factor determines photoreceptor subtypes in the Drosophila retina. Dev Cell 25(1): 93-105. PubMed ID: 23597484

  • Mike Ritchie School of Biology, University of St Andrews, Fife
    Marie-Orleach, L., Bailey, N. W. and Ritchie, M. G. (2019). Social effects on fruit fly courtship song. Ecol Evol 9(1): 410-416. PubMed ID: 30680123

    Parker, D. J., Wiberg, R. A. W., Trivedi, U., Tyukmaeva, V. I., Gharbi, K., Butlin, R. K., Hoikkala, A., Kankare, M. and Ritchie, M. G. (2018). Inter- and intra-specific genomic divergence in Drosophila montana shows evidence for cold adaptation. Genome Biol Evol. PubMed ID: 30010752

    Veltsos, P., Fang, Y., Cossins, A. R., Snook, R. R. and Ritchie, M. G. (2017). Mating system manipulation and the evolution of sex-biased gene expression in Drosophila. Nat Commun 8(1): 2072. PubMed ID: 29233985

    Vigoder, F. M., Parker, D. J., Cook, N., Tourniere, O., Sneddon, T. and Ritchie, M. G. (2016). Inducing Cold-Sensitivity in the Frigophilic Fly Drosophila montana by RNAi. PLoS One 11: e0165724. PubMed ID: 27832122

    Debelle, A., Ritchie, M. G. and Snook, R. R. (2016). Sexual selection and assortative mating: an experimental test. J Evol Biol [Epub ahead of print]. PubMed ID: 26970522

    Zhong, W., McClure, C. D., Evans, C. R., Mlynski, D. T., Immonen, E., Ritchie, M. G. and Priest, N. K. (2013). Immune anticipation of mating in Drosophila: Turandot M promotes immunity against sexually transmitted fungal infections. Proc Biol Sci 280: 20132018. PubMed ID: 24174107

    Parker, D. J., Gardiner, A., Neville, M. C., Ritchie, M. G. and Goodwin, S. F. (2013). The evolution of novelty in conserved genes; evidence of positive selection in the Drosophila fruitless gene is localised to alternatively spliced exons. Heredity (Edinb). PubMed ID: 24149653

    Smith, G., Fang, Y., Liu, X., Kenny, J., Cossins, A. R., de Oliveira, C. C., Etges, W. J. and Ritchie, M. G. (2013). Transcriptome-wide expression variation associated with environmental plasticity and mating success in cactophilic Drosophila mojavensis. Evolution 67: 1950-1963. PubMed ID: 23815652

    Smith, G., Lohse, K., Etges, W. J. and Ritchie, M. G. (2012). Model-based comparisons of phylogeographic scenarios resolve the intraspecific divergence of cactophilic Drosophila mojavensis. Mol Ecol 21: 3293-3307. PubMed ID: 22571504

  • Mel Robertson Department of Biology, Queen's University, Kingston Ontario
    Xiao, C., Qiu, S. and Robertson, R. M. (2017). Persistent one-way Walking in a circular arena in Drosophila melanogaster Canton-S strain. Behav Genet. PubMed ID: 29098495

    Evans, J. J., Xiao, C. and Robertson, R. M. (2017). AMP-activated protein kinase protects against anoxia in Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol 214: 30-39. PubMed ID: 28916374

    Xiao, C., Qiu, S. and Robertson, R. M. (2017). The white gene controls copulation success in Drosophila melanogaster. Sci Rep 7(1): 7712. PubMed ID: 28794482

    Xiao, C. and Robertson, R.M. (2017). White-cGMP interaction promotes fast locomotor recovery from anoxia in adult Drosophila. PLoS One 12: e0168361. PubMed ID: 28060942

    Qiu, S., Xiao, C. and Robertson, R. M. (2016). Pulsed light stimulation increases boundary preference and periodicity of episodic motor activity in Drosophila melanogaster. PLoS One 11: e0163976. PubMed ID: 27684063

    Spong, K. E., Rodriguez, E. C. and Robertson, R. M. (2016). Spreading depolarization in the brain of Drosophila is induced by inhibition of the Na+/K+-ATPase and mitigated by a decrease in activity of protein kinase G. J Neurophysiol: jn 00353 02016. PubMed ID: 27358319

    Spong, K. E., Andrew, R. D. and Robertson, R. M. (2016). Mechanisms of spreading depolarization in vertebrate and insect central nervous systems. J Neurophysiol: jn 00352 02016. PubMed ID: 27334953

    Xiao, C. and Robertson, R. M. (2016). Timing of locomotor recovery from anoxia modulated by the white gene in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 27029736

    Xiao, C. and Robertson, R. M. (2015). Locomotion Induced by Spatial Restriction in Adult Drosophila. PLoS One 10: e0135825. PubMed ID: 26351842

    Rodriguez, E. C. and Robertson, R. M. (2012). Protective effect of hypothermia on brain potassium homeostasis during repetitive anoxia in Drosophila melanogaster. J Exp Biol 215: 4157-4165. PubMed ID: 22899531

  • Alain Robichon Institut Sophia Agrobiotech, Inra, CNRS, Université Nice, France
    Pasquier, C. and Robichon, A. (2022). Temporal and sequential order of nonoverlapping gene networks unraveled in mated female Drosophila. Life Sci Alliance 5(2). PubMed ID: 34844981

    Raad, H. and Robichon, A. (2019). The pleiotropic effects of Innexin genes expressed in Drosophila glia encompass wing chemosensory sensilla. J Neurosci Res. PubMed ID: 31257643

    Pasquier, C., Agnel, S. and Robichon, A. (2017). The Mapping of Predicted Triplex DNA:RNA in the Drosophila Genome Reveals a Prominent Location in Development- and Morphogenesis-Related Genes. G3 (Bethesda). PubMed ID: 28515050

    Capovilla, M., Robichon, A. and Rassoulzadegan, M. (2017). A new paramutation-like example at the Delta gene of Drosophila. PLoS One 12(3): e0172780. PubMed ID: 28355214

    Houot, B., Gigot, V., Robichon, A. and Ferveur, J. F. (2017). Free flight odor tracking in Drosophila: Effect of wing chemosensors, sex and pheromonal gene regulation. Sci Rep 7: 40221. PubMed ID: 28067325

    Raad, H., Ferveur, J. F., Ledger, N., Capovilla, M. and Robichon, A. (2016). Functional Gustatory Role of Chemoreceptors in Drosophila Wings. Cell Rep 15(7): 1442-1454. PubMed ID: 27160896

  • Charles Robin The University of Melbourne
    Scanlan, J. L., Gledhill-Smith, R. S., Battlay, P. and Robin, C. (2020). Genomic and transcriptomic analyses in Drosophila suggest that the ecdysteroid kinase-like (EcKL) gene family encodes the 'detoxification-by-phosphorylation' enzymes of insects. Insect Biochem Mol Biol 123: 103429. PubMed ID: 32540344

    Green, L., Battlay, P., Fournier-Level, A., Good, R. T. and Robin, C. (2019). Cis- and trans-acting variants contribute to survivorship in a naive Drosophila melanogaster population exposed to ryanoid insecticides. Proc Natl Acad Sci U S A 116(21): 10424-10429. PubMed ID: 31064874

    Battlay, P., Leblanc, P. B., Green, L., Garud, N. R., Schmidt, J. M., Fournier-Level, A. and Robin, C. (2018). Structural variants and selective sweep foci contribute to insecticide resistance in the Drosophila genetic reference panel. G3 (Bethesda). PubMed ID: 30190421

    Schmidt, J. M., Battlay, P., Gledhill-Smith, R. S., Good, R. T., Lumb, C., Fournier-Level, A. and Robin, C. (2017). Insights into DDT Resistance from the Drosophila melanogaster Genetic Reference Panel. Genetics [Epub ahead of print]. PubMed ID: 28935691

    Battlay, P., Schmidt, J. M., Fournier-Level, A. and Robin, C. (2016). Genomic and Transcriptomic Associations Identify a New Insecticide Resistance Phenotype for the Selective Sweep at the Cyp6g1 Locus of Drosophila melanogaster. G3 (Bethesda). PubMed ID: 27317781

    Fournier-Level, A., Neumann-Mondlak, A., Good, R. T., Green, L. M., Schmidt, J. M. and Robin, C. (2016). Behavioural response to combined insecticide and temperature stress in natural populations of Drosophila melanogaster. J Evol Biol 29: 1030-1044. PubMed ID: 26864706

    Good, R. T., Gramzow, L., Battlay, P., Sztal, T., Batterham, P. and Robin, C. (2014). The molecular evolution of cytochrome P450 genes within and between drosophila species. Genome Biol Evol 6: 1118-1134. PubMed ID: 24751979

  • Steven Robinow Department of Zoology, University of Hawaii at Manoa, Honolulu
    Bates, K. E., Molnar, J. and Robinow, S. (2014). The unfulfilled gene and nervous system development in Drosophila. Biochim Biophys Acta. PubMed ID: 24953188

    Bates, K. E., Sung, C., Hilson, L. and Robinow, S. (2014). unfulfilled interacting genes display branch-specific roles in the development of mushroom body axons in Drosophila melanogaster. G3 (Bethesda) 4: 693-706. PubMed ID: 24558265

    Bates, K. E., Sung, C. S. and Robinow, S. (2010). The unfulfilled gene is required for the development of mushroom body neuropil in Drosophila. Neural Dev 5: 4. PubMed ID: 20122139

  • Howard Rockman Cardiology, Duke University, Durham, NC
    Lee, T. E., Yu, L., Wolf, M. J. and Rockman, H. A. (2014). Galactokinase Is a Novel Modifier of Calcineurin-Induced Cardiomyopathy in Drosophila. Genetics. PubMed ID: 25081566

    Casad, M. E., Yu, L., Daniels, J. P., Wolf, M. J. and Rockman, H. A. (2012). Deletion of Siah-interacting protein gene in Drosophila causes cardiomyopathy. Mol Genet Genomics 287: 351-360. PubMed ID: 22398840

    Wolf, M. J. and Rockman, H. A. (2011). Drosophila, genetic screens, and cardiac function. Circ Res 109: 794-806. PubMed ID: 21921272

    Casad, M. E., Abraham, D., Kim, I. M., Frangakis, S., Dong, B., Lin, N., Wolf, M. J. and Rockman, H. A. (2011). Cardiomyopathy is associated with ribosomal protein gene haplo-insufficiency in Drosophila melanogaster. Genetics 189: 861-870. PubMed ID: 21890737

  • Avi Rodal Brandeis University
    Del Signore, S. J., Kelley, C. F., Messelaar, E. M., Lemos, T., Marchan, M. F., Ermanoska, B., Mund, M., Fai, T. G., Kaksonen, M. and Rodal, A. A. (2021). An autoinhibitory clamp of actin assembly constrains and directs synaptic endocytosis. Elife 10. PubMed ID: 34324418

    Wang, S., Zhao, Z. and Rodal, A. A. (2019). Higher-order assembly of Sorting Nexin 16 controls tubulation and distribution of neuronal endosomes. J Cell Biol. PubMed ID: 31253649

    Del Signore, S. J., Biber, S. A., Lehmann, K. S., Heimler, S. R., Rosenfeld, B. H., Eskin, T. L., Sweeney, S. T. and Rodal, A. A. (2017). dOCRL maintains immune cell quiescence by regulating endosomal traffic. PLoS Genet 13(10): e1007052. PubMed ID: 29028801

    Stanishneva-Konovalova, T. B., Kelley, C. F., Eskin, T. L., Messelaar, E. M., Wasserman, S. A., Sokolova, O. S. and Rodal, A. A. (2016). Coordinated autoinhibition of F-BAR domain membrane binding and WASp activation by Nervous Wreck. Proc Natl Acad Sci U S A 113: E5552-5561. PubMed ID: 27601635

    Deshpande, M., Feiger, Z., Shilton, A. K., Luo, C. C., Silverman, E. and Rodal, A. A. (2016). Role of BMP receptor traffic in synaptic growth defects in an ALS model. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27535427

    Kelley, C. F., Becalska, A. N., Berciu, C., Nicastro, D. and Rodal, A. A. (2015). Assembly of actin filaments and microtubules in Nwk F-BAR-induced membrane deformations. Commun Integr Biol 8: e1000703. PubMed ID: 26478768

    Koles, K., Messelaar, E. M., Feiger, Z., Yu, C. J., Frank, C. A. and Rodal, A. A. (2015). The EHD protein Past1 controls postsynaptic membrane elaboration and synaptic function. Mol Biol Cell. PubMed ID: 26202464

    Frank, C. A., Wang, X., Collins, C. A., Rodal, A. A., Yuan, Q., Verstreken, P. and Dickman, D. K. (2013). New approaches for studying synaptic development, function, and plasticity using Drosophila as a model system. J Neurosci 33: 17560-17568. PubMed ID: 24198346

    Becalska, A. N., Kelley, C. F., Berciu, C., Stanishneva-Konovalova, T. B., Fu, X., Wang, S., Sokolova, O. S., Nicastro, D. and Rodal, A. A. (2013). Formation of membrane ridges and scallops by the F-BAR protein Nervous Wreck. Mol Biol Cell 24: 2406-2418. PubMed ID: 23761074

    Zhao, L., Wang, D., Wang, Q., Rodal, A. A. and Zhang, Y. Q. (2013). Drosophila cyfip regulates synaptic development and endocytosis by suppressing filamentous actin assembly. PLoS Genet 9: e1003450. PubMed ID: 23593037

  • Aylin Rodan Internal Medicine, University of Texas Southwestern Medical Center, Dallas
    Schellinger, J. N., Sun, Q., Pleinis, J. M., An, S. W., Hu, J., Mercenne, G., Titos, I., Huang, C. L., Rothenfluh, A. and Rodan, A. R. (2022). Chloride oscillation in pacemaker neurons regulates circadian rhythms through a chloride-sensing WNK kinase signaling cascade. Curr Biol 32(6): 1429-1438.e1426. PubMed ID: 35303418

    Acevedo, S. F., Peru, Y. C. d. P. R. L., Gonzalez, D. A., Rodan, A. R. and Rothenfluh, A. (2015). S6 Kinase Reflects and Regulates Ethanol-Induced Sedation. J Neurosci 35: 15396-15402. PubMed ID: 26586826

    Wu, Y., Baum, M., Huang, C. L. and Rodan, A. R. (2015). Two inwardly rectifying potassium channels, Irk1 and Irk2, play redundant roles in Drosophila renal tubule function. Am J Physiol Regul Integr Comp Physiol: ajpregu 00148 02015. PubMed ID: 26224687

    Wu, Y., Schellinger, J. N., Huang, C. L. and Rodan, A. R. (2014). Hypotonicity Stimulates Potassium Flux Through the WNK-SPAK/OSR1 Kinase Cascade and the Ncc69 Sodium-Potassium-2-Chloride Cotransporter in the Drosophila Renal Tubule. J Biol Chem. PubMed ID: 25086033

    Peru, Y. C. d. P. R. L., Ojelade, S. A., Penninti, P. S., Dove, R. J., Nye, M. J., Acevedo, S. F., Lopez, A., Rodan, A. R. and Rothenfluh, A. (2014). Long-lasting, experience-dependent alcohol preference in Drosophila. Addict Biol 19: 392-401. PubMed ID: 24164972

    Peru, Y. C. d. P. R. L., Acevedo, S. F., Rodan, A. R., Chang, L. Y., Eaton, B. A. and Rothenfluh, A. (2012). Adult neuronal Arf6 controls ethanol-induced behavior with Arfaptin downstream of Rac1 and RhoGAP18B. J Neurosci 32: 17706-17713. PubMed ID: 23223291

    Rodan, A. R., Baum, M. and Huang, C. L. (2012). The Drosophila NKCC Ncc69 is required for normal renal tubule function. Am J Physiol Cell Physiol 303: C883-894. PubMed ID: 22914641

  • Thomas Roeder Zoologisches Institut, Christian-Albrechts-Universität zu Kiel
    Proske, A., Bossen, J., von Frieling, J. and Roeder, T. (2021). Low-protein diet applied as part of combination therapy or stand-alone normalizes lifespan and tumor proliferation in a model of intestinal cancer. Aging (Albany NY) 13(undefined). PubMed ID: 34766923

    Wagner, C., Uliczka, K., Bossen, J., Niu, X., Fink, C., Thiedmann, M., Knop, M., Vock, C., Abdelsadik, A., Zissler, U. M., Isermann, K., Garn, H., Pieper, M., Wegmann, M., Koczulla, A. R., Vogelmeier, C. F., Schmidt-Weber, C. B., Fehrenbach, H., Konig, P., Silverman, N., Renz, H., Pfefferle, P., Heine, H. and Roeder, T. (2021). Constitutive immune activity promotes JNK- and FoxO-dependent remodeling of Drosophila airways. Cell Rep 35(1): 108956. PubMed ID: 33826881

    von Frieling, J., Faisal, M. N., Sporn, F., Pfefferkorn, R., Nolte, S. S., Sommer, F., Rosenstiel, P. and Roeder, T. (2020). A high-fat diet induces a microbiota-dependent increase in stem cell activity in the Drosophila intestine. PLoS Genet 16(5): e1008789. PubMed ID: 32453733

    Li, Y., Romey-Glusing, R., Tahan Zadeh, N., von Frieling, J., Hoffmann, J., Huebbe, P., Bruchhaus, I., Rimbach, G., Fink, C. and Roeder, T. (2020). Furbellow (Brown Algae) Extract Increases Lifespan in Drosophila by Interfering with TOR-Signaling. Nutrients 12(4). PubMed ID: 32331413

    Xu, Y., Borcherding, A. F., Heier, C., Tian, G., Roeder, T. and Kuhnlein, R. P. (2019). Chronic dysfunction of Stromal interaction molecule by pulsed RNAi induction in fat tissue impairs organismal energy homeostasis in Drosophila. Sci Rep 9(1): 6989. PubMed ID: 31061470

    Jishage, M., Yu, X., Shi, Y., Ganesan, S. J., Chen, W. Y., Sali, A., Chait, B. T., Asturias, F. J. and Roeder, R. G. (2018). Architecture of Pol II(G) and molecular mechanism of transcription regulation by Gdown1. Nat Struct Mol Biol 25(9): 859-867. PubMed ID: 30190596

    Stephano, F., Nolte, S., Hoffmann, J., El-Kholy, S., von Frieling, J., Bruchhaus, I., Fink, C. and Roeder, T. (2018). Impaired Wnt signaling in dopamine containing neurons is associated with pathogenesis in a rotenone triggered Drosophila Parkinson's disease model. Sci Rep 8(1): 2372. PubMed ID: 29403026

    Romey-Glusing, R., Li, Y., Hoffmann, J., von Frieling, J., Knop, M., Pfefferkorn, R., Bruchhaus, I., Fink, C. and Roeder, T. (2017). Nutritional regimens with periodically recurring phases of dietary restriction extend lifespan in Drosophila. Faseb j. PubMed ID: 29196499

    Li, Y., Hoffmann, J., Li, Y., Stephano, F., Bruchhaus, I., Fink, C. and Roeder, T. (2016). Octopamine controls starvation resistance, life span and metabolic traits in Drosophila. Sci Rep 6: 35359. PubMed ID: 27759117

    Li, Y., Fink, C., El-Kholy, S. and Roeder, T. (2014). The octopamine receptor octss2R is essential for ovulation and fertilization in the fruit fly Drosophila melanogaster. Arch Insect Biochem Physiol. PubMed ID: 25353988

    Faisal, M. N., Hoffmann, J., El-Kholy, S., Kallsen, K., Wagner, C., Bruchhaus, I., Fink, C. and Roeder, T. (2014). Transcriptional Regionalization of the Fruit Fly's Airway Epithelium. PLoS One 9: e102534. PubMed ID: 25020150

  • Fabrice Roegiers Fox Chase Cancer Center, Temple Health, Philadelphia
    Johnson, S. A., Zitserman, D. and Roegiers, F. (2016). Numb regulates the balance between Notch recycling and late endosome targeting in Drosophila neural progenitor cells. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27466320

    Upadhyay, A., Kandachar, V., Zitserman, D., Tong, X., Roegiers, F. (2013) Sanpodo controls sensory organ precursor fate by directing Notch trafficking and binding gamma-secretase. J Cell Biol. PubMed ID: 23609534

    Zitserman, D. and Roegiers, F. (2011). Live-cell imaging of sensory organ precursor cells in intact Drosophila pupae. J Vis Exp. PubMed ID: 21654627

    Tong, X., Zitserman, D., Serebriiskii, I., Andrake, M., Dunbrack, R. and Roegiers, F. (2010). Numb independently antagonizes Sanpodo membrane targeting and Notch signaling in Drosophila sensory organ precursor cells. Mol Biol Cell 21: 802-810. PubMed ID: 20053677

    Karbowniczek, M., Zitserman, D., Khabibullin, D., Hartman, T., Yu, J., Morrison, T., Nicolas, E., Squillace, R., Roegiers, F. and Henske, E. P. (2010). The evolutionarily conserved TSC/Rheb pathway activates Notch in tuberous sclerosis complex and Drosophila external sensory organ development. J Clin Invest 120: 93-102. PubMed ID: 20038815

  • Gregory Rogers University of Arizona Cancer Center, Tucson
    Boese, C. J., Nye, J., Buster, D. W., McLamarrah, T. A., Byrnes, A. E., Slep, K. C., Rusan, N. M. and Rogers, G. C. (2018). Asterless is a Polo-like kinase 4 substrate that both activates and inhibits kinase activity depending on its phosphorylation state. Mol Biol Cell: mbcE18070445. PubMed ID: 30256714

    Galletta, B. J., Fagerstrom, C. J., Schoborg, T. A., McLamarrah, T. A., Ryniawec, J. M., Buster, D. W., Slep, K. C., Rogers, G. C. and Rusan, N. M. (2016). A centrosome interactome provides insight into organelle assembly and reveals a non-duplication role for Plk4. Nat Commun 7: 12476. PubMed ID: 27558293

    Klebba, J. E., Buster, D. W., McLamarrah, T. A., Rusan, N. M. and Rogers, G. C. (2015). Autoinhibition and relief mechanism for Polo-like kinase 4. Proc Natl Acad Sci U S A 112: E657-666. PubMed ID: 25646492

    Nguyen, H. Q., Nye, J., Buster, D. W., Klebba, J. E., Rogers, G. C. and Bosco, G. (2015). Drosophila Casein Kinase I alpha regulates homolog pairing and genome organization by modulating Condensin II subunit Cap-H2 levels. PLoS Genet 11: e1005014. PubMed ID: 25723539

    Klebba, J.E., Galletta, B.J., Nye, J., Plevock, K.M., Buster, D.W., Hollingsworth, N.A., Slep, K.C., Rusan, N.M. and Rogers, G.C. (2015). Two Polo-like kinase 4 binding domains in Asterless perform distinct roles in regulating kinase stability. J Cell Biol 208: 401-414. PubMed ID: 25688134

    Bozler, J., Nguyen, H. Q., Rogers, G. C. and Bosco, G. (2014). Condensins exert force on chromatin-nuclear envelope tethers to mediate nucleoplasmic reticulum formation in Drosophila melanogaster. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 25552604

    Nye, J., Buster, D. W. and Rogers, G. C. (2014). The use of cultured Drosophila cells for studying the microtubule cytoskeleton. Methods Mol Biol 1136: 81-101. PubMed ID: 24633795

    Klebba, J. E., Buster, D. W., Nguyen, A. L., Swatkoski, S., Gucek, M., Rusan, N. M. and Rogers, G. C. (2013). Polo-like Kinase 4 Autodestructs by Generating Its Slimb-Binding Phosphodegron. Curr Biol. PubMed ID: 24184097

    Smith, H. F., Roberts, M. A., Nguyen, H. Q., Peterson, M., Hartl, T. A., Wang, X. J., Klebba, J. E., Rogers, G. C. and Bosco, G. (2013). Maintenance of interphase chromosome compaction and homolog pairing in Drosophila is regulated by the condensin cap-h2 and its partner Mrg15. Genetics 195: 127-146. PubMed ID: 23821596

    Buster, D. W., Nye, J., Klebba, J. E. and Rogers, G. C. (2010). Preparation of Drosophila S2 cells for light microscopy. J Vis Exp. PubMed ID: 20543772

  • Steve Rogers Department of Biology, University of North Carolina at Chapel Hill
    Sanchez-Corrales, Y. E., Blanchard, G. B. and Roper, K. (2021). Correct regionalization of a tissue primordium is essential for coordinated morphogenesis. Elife 10. PubMed ID: 34723792

    Gillard, G., Girdler, G. and Roper, K. (2021). A release-and-capture mechanism generates an essential non-centrosomal microtubule array during tube budding. Nat Commun 12(1): 4096. PubMed ID: 34215746

    Girdler, G. C., Applewhite, D. A., Perry, W. M., Rogers, S. L. and Roper, K. (2015). The Gas2 family protein Pigs is a microtubule +TIP that affects cytoskeleton organisation. J Cell Sci [Epub ahead of print]. PubMed ID: 26585311

    Trogden, K. P. and Rogers, S. L. (2015). TOG Proteins Are Spatially Regulated by Rac-GSK3beta to Control Interphase Microtubule Dynamics. PLoS One 10: e0138966. PubMed ID: 26406596

    Grode, K. D. and Rogers, S. L. (2015). The non-catalytic domains of Drosophila katanin regulate its abundance and microtubule-disassembly activity. PLoS One 10: e0123912. PubMed ID: 25886649

    Peters, K. A. and Rogers, S. L. (2013). Drosophila Ric-8 interacts with the Galpha12/13 subunit, Concertina, during activation of the Folded gastrulation pathway. Mol Biol Cell 24: 3460-3471. PubMed ID: 24006487

    Applewhite, D. A., Grode, K. D., Duncan, M. C. and Rogers, S. L. (2013). The actin-microtubule cross-linking activity of Drosophila Short stop is regulated by intramolecular inhibition. Mol Biol Cell 24: 2885-2893. PubMed ID: 23885120

  • Blanka Rogina Department of Genetics & Developmental Biology, School of Medicine, University of Connecticut Health Center
    Woods, J. K., Ziafazeli, T. and Rogina, B. (2016). Rpd3 interacts with insulin signaling in Drosophila longevity extension. Aging (Albany NY) [Epub ahead of print]. PubMed ID: 27852975

    Frankel, S., Woods, J., Ziafazeli, T. and Rogina, B. (2015). RPD3 histone deacetylase and nutrition have distinct but interacting effects on Drosophila longevity. Aging (Albany NY) [Epub ahead of print]. PubMed ID: 26647291

    Rogers, R. P. and Rogina, B. (2015). The role of INDY in metabolism, health and longevity. Front Genet 6: 204. PubMed ID: 26106407

    Rogers, R. P. and Rogina, B. (2014). Increased mitochondrial biogenesis preserves intestinal stem cell homeostasis and contributes to longevity in Indy mutant flies. Aging (Albany NY) 6: 335-350. PubMed ID: 24827528

    Woods, J. K., Kowalski, S. and Rogina, B. (2014). Determination of the Spontaneous Locomotor Activity in Drosophila melanogaster. J Vis Exp. PubMed ID: 24747955

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

    Garber, G., Smith, L. A., Reenan, R. A. and Rogina, B. (2012). Effect of sodium channel abundance on Drosophila development, reproductive capacity and aging. Fly (Austin) 6: 57-67. PubMed ID: 22513411

  • Jean-Yves Roignant Institute of Molecular Biology, Mainz, Germany
    Bawankar, P., Lence, T., Paolantoni, C., Haussmann, I. U., Kazlauskiene, M., Jacob, D., Heidelberger, J. B., Richter, F. M., Nallasivan, M. P., Morin, V., Kreim, N., Beli, P., Helm, M., Jinek, M., Soller, M. and Roignant, J. Y. (2021). Hakai is required for stabilization of core components of the m(6)A mRNA methylation machinery. Nat Commun 12(1): 3778. PubMed ID: 34145251

    Worpenberg, L., Paolantoni, C., Longhi, S., Mulorz, M. M., Lence, T., Wessels, H. H., Dassi, E., Aiello, G., Sutandy, F. X. R., Scheibe, M., Edupuganti, R. R., Busch, A., Möckel, M. M., Vermeulen, M., Butter, F., König, J., Notarangelo, M., Ohler, U., Dieterich, C., Quattrone, A., Soldano, A. and Roignant, J. Y. (2021). Ythdf is a N6-methyladenosine reader that modulates Fmr1 target mRNA selection and restricts axonal growth in Drosophila. Embo j: e104975. PubMed ID: 33428246

    Leismann, J., Spagnuolo, M., Pradhan, M., Wacheul, L., Vu, M. A., Musheev, M., Mier, P., Andrade-Navarro, M. A., Graille, M., Niehrs, C., Lafontaine, D. L. and Roignant, J. Y. (2020). The 18S ribosomal RNA m(6) A methyltransferase Mettl5 is required for normal walking behavior in Drosophila. EMBO Rep: e49443. PubMed ID: 32350990

    Knuckles, P., Lence, T., Haussmann, I. U., Jacob, D., Kreim, N., Carl, S. H., Masiello, I., Hares, T., Villasenor, R., Hess, D., Andrade-Navarro, M. A., Biggiogera, M., Helm, M., Soller, M., Buhler, M. and Roignant, J. Y. (2018). Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m(6)A machinery component Wtap/Fl(2)d. Genes Dev. PubMed ID: 29535189

    Dinges, N., Morin, V., Kreim, N., Southall, T. D. and Roignant, J. Y. (2017). Comprehensive characterization of the complex lola locus reveals a novel role in the octopaminergic pathway via tyramine beta-Hydroxylase regulation. Cell Rep 21(10): 2911-2925. PubMed ID: 29212035

    Casas-Vila, N., Bluhm, A., Sayols, S., Dinges, N., Dejung, M., Altenhein, T., Kappei, D., Altenhein, B., Roignant, J. Y. and Butter, F. (2017). The developmental proteome of Drosophila melanogaster. Genome Res 27(7): 1273-1285. PubMed ID: 28381612

    Lence, T., Akhtar, J., Bayer, M., Schmid, K., Spindler, L., Ho, C. H., Kreim, N., Andrade-Navarro, M. A., Poeck, B., Helm, M. and Roignant, J. Y. (2016). m(6)A modulates neuronal functions and sex determination in Drosophila. Nature 540(7632): 242-247. PubMed ID: 27919077

  • Stephanie Rollmann University of Cincinnati
    Brown, E., Layne, J. E., Elchert, A. R. and Rollmann, S. M. (2020). Behavioral and Transcriptional Response to Selection for Olfactory Behavior in Drosophila. G3 (Bethesda). PubMed ID: 32024668

    Brown, E. B., Rayens, E. and Rollmann, S. M. (2019). The gene CG6767 affects olfactory behavior in Drosophila melanogaster. Behav Genet. PubMed ID: 30710192

    Brown, E. B., Patterson, C., Pancoast, R. and Rollmann, S. M. (2017). Artificial selection for odor-guided behavior in Drosophila reveals changes in food consumption. BMC Genomics 18(1): 867. PubMed ID: 29132294

    Brown, E. B., Layne, J. E., Zhu, C., Jegga, A. G., Rollmann, S. M. (2013) Genome-wide Association Mapping of Natural Variation in Odor-guided Behavior in Drosophila. Genes Brain Behav. PubMed ID: 23682951

    Richgels, P. K. and Rollmann, S. M. (2012). Genetic variation in odorant receptors contributes to variation in olfactory behavior in a natural population of Drosophila melanogaster. Chem Senses 37: 229-240. PubMed ID: 22038943

    Rollmann, S. M., Wang, P., Date, P., West, S. A., Mackay, T. F. and Anholt, R. R. (2010). Odorant receptor polymorphisms and natural variation in olfactory behavior in Drosophila melanogaster. Genetics 186: 687-697. PubMed ID: 20628035

  • Melissa Rolls Biochemistry and Molecular Biology, Penn State University
    Feng, C., Cleary, J. M., Kothe, G. O., Stone, M. C., Weiner, A. T., Hertzler, J. I., Hancock, W. O. and Rolls, M. M. (2021). Trim9 and Klp61F promote polymerization of new dendritic microtubules along parallel microtubules. J Cell Sci. PubMed ID: 33988240

    Shorey, M., Stone, M. C., Mandel, J. and Rolls, M. M. (2020). Neurons survive simultaneous injury to axons and dendrites and regrow both types of processes in vivo. Dev Biol. PubMed ID: 32687893

    Hertzler, J. I., Simonovitch, S. I., Albertson, R. M., Weiner, A. T., Nye, D. M. R. and Rolls, M. M. (2020). Kinetochore proteins suppress neuronal microtubule dynamics and promote dendrite regeneration. Mol Biol Cell: mbcE20040237T. PubMed ID: 32673176

    Weiner, A. T., Seebold, D. Y., Torres-Gutierrez, P., Folker, C., Swope, R. D., Kothe, G. O., Stoltz, J. G., Zalenski, M. K., Kozlowski, C., Barbera, D. J., Patel, M. A., Thyagarajan, P., Shorey, M., Nye, D. M. R., Keegan, M., Behari, K., Song, S., Axelrod, J. D. and Rolls, M. M. (2020). Endosomal Wnt signaling proteins control microtubule nucleation in dendrites. PLoS Biol 18(3): e3000647. PubMed ID: 32163403

    Nye, D. M. R., Albertson, R. M., Weiner, A. T., Hertzler, J. I., Shorey, M., Goberdhan, D. C. I., Wilson, C., Janes, K. A. and Rolls, M. M. (2020). The receptor tyrosine kinase Ror is required for dendrite regeneration in Drosophila neurons. PLoS Biol 18(3): e3000657. PubMed ID: 32163406

    Feng, C., Thyagarajan, P., Shorey, M., Seebold, D. Y., Weiner, A. T., Albertson, R. M., Rao, K. S., Sagasti, A., Goetschius, D. J. and Rolls, M. M. (2019). Patronin-mediated minus end growth is required for dendritic microtubule polarity. J Cell Biol. PubMed ID: 31076454

    Weiner, A. T., Seebold, D. Y., Michael, N. L., Guignet, M., Feng, C., Follick, B., Yusko, B. A., Wasilko, N. P., Torres-Gutierrez, P. and Rolls, M. M. (2018). Identification of proteins required for precise positioning of Apc2 in dendrites. G3 (Bethesda). PubMed ID: 29602811

    Jegla, T., Nguyen, M. M., Feng, C., Goetschius, D. J., Luna, E., van Rossum, D. B., Kamel, B., Pisupati, A., Milner, E. S. and Rolls, M. M. (2016). Bilaterian giant Ankyrins have a common evolutionary origin and play a conserved role in patterning the axon initial segment. PLoS Genet 12(12): e1006457. PubMed ID: 27911898

    Chen, L., Nye, D. M., Stone, M. C., Weiner, A. T., Gheres, K. W., Xiong, X., Collins, C. A. and Rolls, M. M. (2016). Mitochondria and caspases tune Nmnat-mediated stabilization to promote axon regeneration. PLoS Genet 12(12): e1006503. PubMed ID: 27923046

    Rao, K., Stone, M. C., Weiner, A. T., Gheres, K. W., Zhou, C., Deitcher, D. L., Levitan, E. S. and Rolls, M. M. (2016). Spastin, atlastin and ER relocalization are involved in axon, but not dendrite, regeneration. Mol Biol Cell. PubMed ID: 27605706

  • Gregg Roman Biology and Biochemistry, University of Mississippi
    Myers, J. L., Porter, M., Narwold, N., Bhat, K., Dauwalder, B. and Roman, G. (2021). Mutants of the white ABCG Transporter in Drosophila melanogaster Have Deficient Olfactory Learning and Cholesterol Homeostasis. Int J Mol Sci 22(23). PubMed ID: 34884779

    Kang, Y. Y., Wachi, Y., Engdorf, E., Fumagalli, E., Wang, Y., Myers, J., Massey, S., Greiss, A., Xu, S. and Roman, G. (2020). Normal Ethanol Sensitivity and Rapid Tolerance Require the G Protein Receptor Kinase 2 in Ellipsoid Body Neurons in Drosophila. Alcohol Clin Exp Res. PubMed ID: 32573992

    Xu, S., Pany, S., Benny, K., Tarique, K., Al-Hatem, O., Gajewski, K., Leasure, J. L., Das, J. and Roman, G. (2018). Ethanol regulates presynaptic activity and sedation through presynaptic Unc13 uroteins in Drosophila. eNeuro 5(3). PubMed ID: 29911175

    de la Flor, M., Chen, L., Manson-Bishop, C., Chu, T. C., Zamora, K., Robbins, D., Gunaratne, G. and Roman, G. (2017). Drosophila increase exploration after visually detecting predators. PLoS One 12(7): e0180749. PubMed ID: 28746346

    Zhang, S. and Roman, G. (2013). Presynaptic Inhibition of Gamma Lobe Neurons Is Required for Olfactory Learning in Drosophila. Curr Biol. PubMed ID: 24291093

    Das, J., Xu, S., Pany, S., Guillory, A., Shah, V. and Roman, G. W. (2013). The pre-synaptic Munc13-1 binds alcohol and modulates alcohol self-administration in Drosophila. J Neurochem 126: 715-726. PubMed ID: 23692447

    Soibam, B., Shah, S., Gunaratne, G. H. and Roman, G. W. (2013). Modeling novelty habituation during exploratory activity in Drosophila. Behav Processes 97: 63-75. PubMed ID: 23597866

    Bittner, E. R., Madalan, A., Czader, A. and Roman, G. (2012). Quantum origins of molecular recognition and olfaction in Drosophila. J Chem Phys 137: 22A551. PubMed ID: 23249088

    Soibam, B., Goldfeder, R. L., Manson-Bishop, C., Gamblin, R., Pletcher, S. D., Shah, S., Gunaratne, G. H. and Roman, G. W. (2012). Modeling Drosophila positional preferences in open field arenas with directional persistence and wall attraction. PLoS One 7: e46570. PubMed ID: 23071591

  • Yikang Rong School of Life Sciences, Sun Yat-sen University, Guangzhou, China
    Huang, W., Liu, Z. and Rong, Y. S. (2021). Dynamic localization of DNA topoisomerase I and its functional relevance during Drosophila development. G3 (Bethesda) 11(9). PubMed ID: 34544118

    Cheng, L., Zhang, Y., Zhang, Y., Chen, T., Xu, Y. Z. and Rong, Y. S. (2020). Loss of the RNA trimethylguanosine cap is compatible with nuclear accumulation of spliceosomal snRNAs but not pre-mRNA splicing or snRNA processing during animal development. PLoS Genet 16(10): e1009098. PubMed ID: 33085660

    Ji, J., Tang, X., Hu, W., Maggert, K. A. and Rong, Y. S. (2019). The processivity factor Pol32 mediates nuclear localization of DNA polymerase delta and prevents chromosomal fragile site formation in Drosophila development. PLoS Genet 15(5): e1008169. PubMed ID: 31100062

    Morciano, P., Di Giorgio, M. L., Porrazzo, A., Licursi, V., Negri, R., Rong, Y. and Cenci, G. (2019). Depletion of ATP-Citrate Lyase (ATPCL) affects chromosome integrity without altering histone acetylation in Drosophila mitotic cells. Front Physiol 10: 383. PubMed ID: 31019471

    Lu, D., Li, Z., Li, L., Yang, L., Chen, G., Yang, D., Zhang, Y., Singh, V., Smith, S., Xiao, Y., Wang, E., Ye, Y., Zhang, W., Zhou, L., Rong, Y. and Zhou, J. (2018). The Ubx Polycomb response element bypasses an unpaired Fab-8 insulator via cis transvection in Drosophila. PLoS One 13(6): e0199353. PubMed ID: 29928011

    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

    Kurzhals, R. L., Fanti, L., Gonzalez Ebsen, A. C., Rong, Y. S., Pimpinelli, S. and Golic, K. G. (2017). Chromosome healing is promoted by the telomere cap component Hiphop in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 28942425

    Tang, X., Cao, J., Zhang, L., Huang, Y., Zhang, Q. and Rong, Y. S. (2017). Maternal Haploid, a metalloprotease enriched at the largest satellite repeat and essential for genome integrity in Drosophila embryos. Genetics. PubMed ID: 28615282

    Zhang Y, Zhang L, Tang X, Bhardwaj SR, Ji J, Rong YS (2016). MTV, an ssDNA Protecting Complex Essential for Transposon-Based Telomere Maintenance in Drosophila. PLoS Genet. 2016 Nov 11;12(11):e1006435. PubMed ID: 27835648

    Zhang, L., Beaucher, M., Cheng, Y. and Rong, Y. S. (2014). Coordination of transposon expression with DNA replication in the targeting of telomeric retrotransposons in Drosophila. EMBO J [Epub ahead of print]. PubMed ID: 24733842

  • Matthew Ronshaugen University of Manchester
    Gallicchio, L., Griffiths-Jones, S. and Ronshaugen, M. (2022). miR-9a regulates levels of both rhomboid mRNA and protein in the early Drosophila melanogaster embryo. G3 (Bethesda) 12(4). PubMed ID: 35143618

    Gallicchio, L., Griffiths-Jones, S. and Ronshaugen, M. (2021). Single-cell visualization of mir-9a and Senseless co-expression during Drosophila melanogaster embryonic and larval peripheral nervous system development. G3 (Bethesda) 11(1). PubMed ID: 33561238

    Ninova, M., Griffiths-Jones, S. and Ronshaugen, M. (2017). Abundant expression of somatic transposon-derived piRNAs throughout Tribolium castaneum embryogenesis. Genome Biol 18(1): 184. PubMed ID: 28950880

    Kozomara, A., Hunt, S., Ninova, M., Griffiths-Jones, S. and Ronshaugen, M. (2014). Target Repression Induced by Endogenous microRNAs: Large Differences, Small Effects. PLoS One 9: e104286. PubMed ID: 25141277

    Ninova, M., Ronshaugen, M. and Griffiths-Jones, S. (2014). Fast-evolving microRNAs are highly expressed in the early embryo of Drosophila virilis. RNA. PubMed ID: 24448446

    Marco, A., Ninova, M., Ronshaugen, M. and Griffiths-Jones, S. (2013). Clusters of microRNAs emerge by new hairpins in existing transcripts. Nucleic Acids Res. PubMed ID: 23775791

    Hui, J. H., Marco, A., Hunt, S., Melling, J., Griffiths-Jones, S., Ronshaugen, M. (2013) Structure, evolution and function of the bi-directionally transcribed iab-4/iab-8 microRNA locus in arthropods. Nucleic Acids Res. PubMed ID: 23335784

    Griffiths-Jones, S., Hui, J. H., Marco, A. and Ronshaugen, M. (2011). MicroRNA evolution by arm switching. EMBO Rep 12: 172-177. PubMed ID: 21212805

    Marco, A., Hui, J. H., Ronshaugen, M. and Griffiths-Jones, S. (2010). Functional shifts in insect microRNA evolution. Genome Biol Evol 2: 686-696. PubMed ID: 20817720

  • Stéphane Ronsseray Institute of Biology, Université Pierre et Marie, Paris
    Asif-Laidin, A., Delmarre, V., Laurentie, J., Miller, W. J., Ronsseray, S. and Teysset, L. (2017). Short and long-term evolutionary dynamics of subtelomeric piRNA clusters in Drosophila. DNA Res [Epub ahead of print]. PubMed ID: 28459978

    Marie, P.P., Ronsseray, S. and Boivin, A. From embryo to adult: piRNA-mediated silencing throughout germline development in Drosophila. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27932388

    Hermant, C., Boivin, A., Teysset, L., Delmarre, V., Asif-Laidin, A., Beek, M. V., Antoniewski, C. and Ronsseray, S. (2015). Paramutation in Drosophila Requires Both Nuclear and Cytoplasmic Actors of the piRNA Pathway and Induces Cis-Spreading of piRNA Production. Genetics. PubMed ID: 26482790

    Basquin, D., Spierer, A., Begeot, F., Koryakov, D. E., Todeschini, A. L., Ronsseray, S., Vieira, C., Spierer, P. and Delattre, M. (2014). The Drosophila Su(var)3-7 gene is required for oogenesis and female fertility, genetically interacts with piwi and aubergine, but impacts only weakly transposon silencing. PLoS One 9: e96802. PubMed ID: 24820312

    Dufourt, J., Dennis, C., Boivin, A., Gueguen, N., Theron, E., Goriaux, C., Pouchin, P., Ronsseray, S., Brasset, E. and Vaury, C. (2014). Spatio-temporal requirements for transposable element piRNA-mediated silencing during Drosophila oogenesis. Nucleic Acids Res 42: 2512-2524. PubMed ID: 24288375

  • Katja Roper MRC Laboratory of Molecular Biology, Cambridge
    Sanchez-Corrales, Y. E., Blanchard, G. B. and Roper, K. (2018). Radially-patterned cell behaviours during tube budding from an epithelium. Elife 7. PubMed ID: 30015616

    Girdler, G. C., Applewhite, D. A., Perry, W. M., Rogers, S. L. and Roper, K. (2015). The Gas2 family protein Pigs is a microtubule +TIP that affects cytoskeleton organisation. J Cell Sci [Epub ahead of print]. PubMed ID: 26585311

    Roper, K. (2015). Integration of cell-cell adhesion and contractile actomyosin activity during morphogenesis. Curr Top Dev Biol 112: 103-127. PubMed ID: 25733139

    Booth, A. J., Blanchard, G. B., Adams, R. J. and Roper, K. (2014). A dynamic microtubule cytoskeleton directs medial actomyosin function during tube formation. Dev Cell 29: 562-576. PubMed ID: 24914560

    Girdler, G. C. and Roper, K. (2014). Controlling cell shape changes during salivary gland tube formation in Drosophila. Semin Cell Dev Biol. PubMed ID: 24685610

    Roper, K. (2013). Supracellular actomyosin assemblies during development. Bioarchitecture 3: 45-49. PubMed ID: 23760352

    Roper, K. (2012). Anisotropy of Crumbs and aPKC drives myosin cable assembly during tube formation. Dev Cell 23: 939-953. PubMed ID: 23153493

    Pines, M. K., Housden, B. E., Bernard, F., Bray, S. J. and Roper, K. (2010). The cytolinker Pigs is a direct target and a negative regulator of Notch signalling. Development 137: 913-922. PubMed ID: 20150280

  • Enzo Rosato Department of Genetics, University of Leicester
    Delfino, L., Mason, R. P., Kyriacou, C. P., Giorgini, F. and Rosato, E. (2020). Rab8 Promotes Mutant HTT Aggregation, Reduces Neurodegeneration, and Ameliorates Behavioural Alterations in a Drosophila Model of Huntington's Disease. J Huntingtons Dis 9(3): 253-263. PubMed ID: 33044189

    Pegoraro, M., Picot, E., Hansen, C. N., Kyriacou, C. P., Rosato, E. and Tauber, E. (2015). Gene Expression Associated with Early and Late Chronotypes in Drosophila melanogaster. Front Neurol 6: 100. PubMed ID: 26097463

    Dissel, S., Hansen, C. N., Ozkaya, O., Hemsley, M., Kyriacou, C. P. and Rosato, E. (2014). The Logic of Circadian Organization in Drosophila. Curr Biol. PubMed ID: 25220056

    Mallon, E. B., Alghamdi, A., Holdbrook, R. T. and Rosato, E. (2014). Immune stimulation reduces sleep and memory ability in Drosophila melanogaster. PeerJ 2: e434. PubMed ID: 24949247

    Damulewicz, M., Rosato, E. and Pyza, E. (2013). Circadian regulation of the Na+/K+-ATPase alpha subunit in the visual system is mediated by the pacemaker and by retina photoreceptors in Drosophila melanogaster. PLoS One 8: e73690. PubMed ID: 24040028

    Kijak, E., Rosato, E., Knapczyk, K. and Pyza, E. (2014). Drosophila melanogaster as a model system of aluminum toxicity and aging. Insect Sci 21: 189-202. PubMed ID: 23956142

  • Michael Rosbash Brandeis University
    Yang, N., Srivastav, S. P., Rahman, R., Ma, Q., Dayama, G., Li, S., Chinen, M., Lei, E. P., Rosbash, M. and Lau, N. C. (2022). Transposable element landscapes in aging Drosophila. PLoS Genet 18(3): e1010024. PubMed ID: 35239675

    Ma, D., Przybylski, D., Abruzzi, K. C., Schlichting, M., Li, Q., Long, X. and Rosbash, M. (2021). A transcriptomic taxonomy of Drosophila circadian neurons around the clock. Elife 10. PubMed ID: 33438579

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

    Jin, H., Xu, W., Rahman, R., Na, D., Fieldsend, A., Song, W., Liu, S., Li, C. and Rosbash, M. (2020). TRIBE editing reveals specific mRNA targets of eIF4E-BP in Drosophila and in mammals. Sci Adv 6(33): eabb8771. PubMed ID: 32851185

    Schlichting, M., Weidner, P., Diaz, M., Menegazzi, P., Dalla Benetta, E., Helfrich-Forster, C. and Rosbash, M. (2019). Light-mediated circuit switching in the Drosophila neuronal clock network. Curr Biol 29(19): 3266-3276. PubMed ID: 31564496

    Schlichting, M., Diaz, M. M., Xin, J. and Rosbash, M. (2019). Neuron-specific knockouts indicate the importance of network communication to Drosophila rhythmicity. Elife 8. PubMed ID: 31613223

    Schlichting, M., Menegazzi, P., Rosbash, M. and Helfrich-Forster, C. (2019). A distinct visual pathway mediates high light intensity adaptation of the circadian clock in Drosophila. J Neurosci. PubMed ID: 30606757

    Diaz, M. M., Schlichting, M., Abruzzi, K. C., Long, X. and Rosbash, M. (2019). Allatostatin-C/AstC-R2 is a novel pathway to modulate the circadian activity pattern in Drosophila. Curr Biol 29(1): 13-22.e13. PubMed ID: 30554904

    Guo, F., Holla, M., Diaz, M. M. and Rosbash, M. (2018). A Circadian Output Circuit Controls Sleep-Wake Arousal in Drosophila. Neuron. PubMed ID: 30269992

    Luo, W., Guo, F., McMahon, A., Couvertier, S., Jin, H., Diaz, M., Fieldsend, A., Weerapana, E. and Rosbash, M. (2018). NonA and CPX link the circadian clockwork to locomotor activity in Drosophila. Neuron 99(4): 768-780.e763. PubMed ID: 30057203

    Rahman, R., Xu, W., Jin, H. and Rosbash, M. (2018). Identification of RNA-binding protein targets with HyperTRIBE. Nat Protoc. PubMed ID: 30013039

  • Michael Rose University of California, Irvine
    Kezos, J. N., Phillips, M. A., Thomas, M. D., Ewunkem, A. J., Rutledge, G. A., Barter, T. T., Santos, M. A., Wong, B. D., Arnold, K. R., Humphrey, L. A., Yan, A., Nouzille, C., Sanchez, I., Cabral, L. G., Bradley, T. J., Mueller, L. D., Graves, J. L., Jr. and Rose, M. R. (2019). Genomics of early cardiac dysfunction and mortality in obese Drosophila melanogaster. Physiol Biochem Zool 92(6): 591-611. PubMed ID: 31603376

    Kezos, J. N., Cabral, L. G., Wong, B. D., Khou, B. K., Oh, A., Harb, J. F., Chiem, D., Bradley, T. J., Mueller, L. D. and Rose, M. R. (2017). Starvation but not locomotion enhances heart robustness in Drosophila. J Insect Physiol [Epub ahead of print]. PubMed ID: 28285040

    Graves, J. L., Jr., Hertweck, K. L., Phillips, M. A., Han, M. V., Cabral, L. G., Barter, T. T., Greer, L. F., Burke, M. K., Mueller, L. D. and Rose, M. R. (2017). Genomics of Parallel Experimental Evolution in Drosophila. Mol Biol Evol. PubMed ID: 28087779

    Phillips, M. A., Long, A. D., Greenspan, Z. S., Greer, L. F., Burke, M. K., Villeponteau, B., Matsagas, K. C., Rizza, C. L., Mueller, L. D. and Rose, M. R. (2016). Genome-wide analysis of long-term evolutionary domestication in Drosophila melanogaster. Sci Rep 6: 39281. PubMed ID: 28004838

    Shahrestani, P., Wilson, J. B., Mueller, L. D. and Rose, M. R. (2016). Patterns of physiological decline due to age and selection in Drosophila melanogaster. Evolution [Epub ahead of print]. PubMed ID: 27624548

    Burke, M.K., Barter, T.T., Cabral, L.G., Kezos, J.N., Phillips, M.A., Rutledge, G.A., Phung, K.H., Chen, R.H., Nguyen, H.D., Mueller, L.D. and Rose, M.R. (2016). Rapid divergence and convergence of life-history in experimentally evolved Drosophila melanogaster. Evolution [Epub ahead of print]. PubMed ID: 27431916

    Mueller, L. D., Joshi, A., Santos, M. and Rose, M. R. (2013). Effective population size and evolutionary dynamics in outbred laboratory populations of Drosophila. J Genet 92: 349-361. PubMed ID: 24371158

    Santos, J., Pascual, M., Simoes, P., Fragata, I., Lima, M., Kellen, B., Santos, M., Marques, A., Rose, M. R. and Matos, M. (2012). From nature to the laboratory: the impact of founder effects on adaptation. J Evol Biol 25: 2607-2622. PubMed ID: 23110657

    Shahrestani, P., Quach, J., Mueller, L. D. and Rose, M. R. (2012). Paradoxical physiological transitions from aging to late life in Drosophila. Rejuvenation Res 15: 49-58. PubMed ID: 22233126

  • Siegfried Roth Institute of Developmental Biology, University of Cologne
    Pechmann, M., Kenny, N. J., Pott, L., Heger, P., Chen, Y. T., Buchta, T., Ozuak, O., Lynch, J. and Roth, S. (2021). Striking parallels between dorsoventral patterning in Drosophila and Gryllus reveal a complex evolutionary history behind a model gene regulatory network. Elife 10. PubMed ID: 33783353

    Benton, M. A., Frey, N., Nunes da Fonseca, R., von Levetzow, C., Stappert, D., Hakeemi, M. S., Conrads, K. H., Pechmann, M., Panfilio, K. A., Lynch, J. A. and Roth, S. (2019). Fog signaling has diverse roles in epithelial morphogenesis in insects. Elife 8. PubMed ID: 31573513

    Benton, M. A., Pechmann, M., Frey, N., Stappert, D., Conrads, K. H., Chen, Y. T., Stamataki, E., Pavlopoulos, A. and Roth, S. (2016). Toll genes have an ancestral role in axis elongation. Curr Biol [Epub ahead of print]. PubMed ID: 27212406

    Sachs, L., Chen, Y. T., Drechsler, A., Lynch, J. A., Panfilio, K. A., Lassig, M., Berg, J. and Roth, S. (2015). Dynamic BMP signaling polarized by Toll patterns the dorsoventral axis in a hemimetabolous insect. Elife 4. PubMed ID: 25962855

    Panfilio, K. A., Oberhofer, G. and Roth, S. (2013). High plasticity in epithelial morphogenesis during insect dorsal closure. Biol Open 2: 1108-1118. PubMed ID: 24244847

    Baffet, A. D., Benoit, B., Januschke, J., Audo, J., Gourhand, V., Roth, S. and Guichet, A. (2012). Drosophila tubulin-binding cofactor B is required for microtubule network formation and for cell polarity. Mol Biol Cell 23: 3591-3601. PubMed ID: 22855530

    Technau, M., Knispel, M. and Roth, S. (2012). Molecular mechanisms of EGF signaling-dependent regulation of pipe, a gene crucial for dorsoventral axis formation in Drosophila. Dev Genes Evol 222: 1-17. PubMed ID: 22198544

  • Adrian Rothenfluh University of Utah, Salt Lake City
    Merrill, C. B., Pabon, M. A., Montgomery, A. B., Rodan, A. R. and Rothenfluh, A. (2022). Optimized assay for transposase-accessible chromatin by sequencing (ATAC-seq) library preparation from adult Drosophila melanogaster neurons. Sci Rep 12(1): 6043. PubMed ID: 35411004

    Schellinger, J. N., Sun, Q., Pleinis, J. M., An, S. W., Hu, J., Mercenne, G., Titos, I., Huang, C. L., Rothenfluh, A. and Rodan, A. R. (2022). Chloride oscillation in pacemaker neurons regulates circadian rhythms through a chloride-sensing WNK kinase signaling cascade. Curr Biol 32(6): 1429-1438.e1426. PubMed ID: 35303418

    Mishra, P., Yang, S. E., Montgomery, A. B., Reed, A. R., Rodan, A. R. and Rothenfluh, A. (2021). The fly liquid-food electroshock assay (FLEA) suggests opposite roles for neuropeptide F in avoidance of bitterness and shock. BMC Biol 19(1): 31. PubMed ID: 33593351

    Chvilicek, M. M., Titos, I. and Rothenfluh, A. (2020). The Neurotransmitters Involved in Drosophila Alcohol-Induced Behaviors. Front Behav Neurosci 14: 607700. PubMed ID: 33384590

    Butts, A. R., Ojelade, S. A., Pronovost, E. D., Seguin, A., Merrill, C. B., Rodan, A. R. and Rothenfluh, A. (2019). Altered actin filament dynamics in the Drosophila mushroom bodies lead to fast acquisition of alcohol consumption preference. J Neurosci. PubMed ID: 31558618

    Shalaby, N. A., Pinzon, J. H., Narayanan, A. S., Jin, E. J., Ritz, M. P., Dove, R. J., Wolfenberg, H., Rodan, A. R., Buszczak, M. and Rothenfluh, A. (2018). JmjC domain proteins modulate circadian behaviors and sleep in Drosophila. Sci Rep 8(1): 815. PubMed ID: 29339751

    Pinzon, J. H., Reed, A. R., Shalaby, N. A., Buszczak, M., Rodan, A. R. and Rothenfluh, A. (2017). Alcohol-Induced Behaviors Require a Subset of Drosophila JmjC-Domain Histone Demethylases in the Nervous System. Alcohol Clin Exp Res 41(12): 2015-2024. PubMed ID: 28940624

  • Daniela Rotin Biochemistry, University of Toronto
    Wasserman, S. S., Shteiman-Kotler, A., Harris, K., Iliadi, K. G., Persaud, A., Zhong, Y., Zhang, Y., Fang, X., Boulianne, G. L., Stewart, B. and Rotin, D. (2018). Regulation of SH3PX1 by dNedd4-long at the Drosophila Neuromuscular Junction. J Biol Chem. PubMed ID: 30518551

    Safi, F., Shteiman-Kotler, A., Zhong, Y., Iliadi, K. G., Boulianne, G. L. and Rotin, D. (2016). Drosophila Nedd4-long reduces Amphiphysin levels in muscles and leads to impaired T-tubule formation. Mol Biol Cell 27(6): 907-918. PubMed ID: 26823013

    Zhong, Y., Shtineman-Kotler, A., Nguyen, L., Iliadi, K. G., Boulianne, G. L. and Rotin, D. (2011). A splice isoform of DNedd4, DNedd4-long, negatively regulates neuromuscular synaptogenesis and viability in Drosophila. PLoS One 6(11): e27007. PubMed ID: 22110599

  • Alain Roussel Architecture et Fonction des Macromolécules Biologiques, CNRS, Marseille
    Colinet, H., Renault, D. and Roussel, D. (2016). Cold acclimation allows Drosophila flies to maintain mitochondrial functioning under cold stress. Insect Biochem Mol Biol 80: 52-60. PubMed ID: 27903433

    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

    Costechareyre, D., Capo, F., Fabre, A., Chaduli, D., Kellenberger, C., Roussel, A., Charroux, B. and Royet, J. (2015). Tissue-Specific Regulation of Drosophila NF-kB Pathway Activation by Peptidoglycan Recognition Protein SC. J Innate Immun. PubMed ID: 26513145

    Leone, P., Bebeacua, C., Opota, O., Kellenberger, C., Klaholz, B., Orlov, I., Cambillau, C., Lemaitre, B. and Roussel, A. (2015). X-ray and Cryo-electron Microscopy Structures of Monalysin Pore-forming Toxin Reveal Multimerization of the Pro-form. J Biol Chem 290: 13191-13201. PubMed ID: 25847242

  • Francois Rouyer Le Centre national de la recherche scientifique, Paris
    Grima, B., Papin, C., Martin, B., Chelot, E., Ponien, P., Jacquet, E. and Rouyer, F. (2019). PERIOD-controlled deadenylation of the timeless transcript in the Drosophila circadian clock. Proc Natl Acad Sci U S A 116(12): 5721-5726. PubMed ID: 30833404

    Alejevski, F., Saint-Charles, A., Michard-Vanhee, C., Martin, B., Galant, S., Vasiliauskas, D. and Rouyer, F. (2019). The HisCl1 histamine receptor acts in photoreceptors to synchronize Drosophila behavioral rhythms with light-dark cycles. Nat Commun 10(1): 252. PubMed ID: 30651542

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

    Szabo, A., Papin, C., Cornu, D., Chelot, E., Lipinszki, Z., Udvardy, A., Redeker, V., Mayor, U. and Rouyer, F. (2018). Ubiquitylation dynamics of the clock cell proteome and TIMELESS during a circadian sycle. Cell Rep 23(8): 2273-2282. Pubmed ID: 29791839

    Saint-Charles, A., Michard-Vanhée, C., Alejevski, F., Chélot, E., Boivin, A. and Rouyer, F. (2016). Four of the six Drosophila rhodopsin-expressing photoreceptors can mediate circadian entrainment in low light. J Comp Neurol [Epub ahead of print]. PubMed ID: 26972685

    Szabo, A., Papin, C., Zorn, D., Ponien, P., Weber, F., Raabe, T. and Rouyer, F. (2013). The CK2 Kinase Stabilizes CLOCK and Represses Its Activity in the Drosophila Circadian Oscillator. PLoS Biol 11: e1001645. PubMed ID: 24013921

    Grima, B., Dognon, A., Lamouroux, A., Chelot, E. and Rouyer, F. (2012). CULLIN-3 controls TIMELESS oscillations in the Drosophila circadian clock. PLoS Biol 10: e1001367. PubMed ID: 22879814

    Klarsfeld, A., Picot, M., Vias, C., Chelot, E. and Rouyer, F. (2011). Identifying specific light inputs for each subgroup of brain clock neurons in Drosophila larvae. J Neurosci 31: 17406-17415. PubMed ID: 22131402

    Lamaze, A., Lamouroux, A., Vias, C., Hung, H. C., Weber, F. and Rouyer, F. (2011). The E3 ubiquitin ligase CTRIP controls CLOCK levels and PERIOD oscillations in Drosophila. EMBO Rep 12: 549-557. PubMed ID: 21525955

  • Jagat Kumar Roy Department of Zoology, Banaras Hindu University, Varanas
    Kunar, R. and Roy, J. K. (2021). The mRNA decapping protein 2 (DCP2) is a major regulator of developmental events in Drosophila-insights from expression paradigms. Cell Tissue Res. PubMed ID: 34536141

    Nandy, N. and Roy, J. K. (2020). Rab11 is essential for lgl mediated JNK-Dpp signaling in dorsal closure and epithelial morphogenesis in Drosophila. Dev Biol 464(2): 188-201. PubMed ID: 32562757

    Mishra, R., Kunar, R., Mandal, L., Alone, D. P., Chandrasekharan, S., Tiwari, A. K., Tapadia, M. G., Mukherjee, A. and Roy, J. K. (2020). A Forward Genetic Approach to Mapping a P-Element Second Site Mutation Identifies DCP2 as a Novel Tumour Suppressor in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 32591349

    Nandy, N. and Roy, J. K. (2020). Rab11 is essential for lgl mediated JNK-Dpp signaling in dorsal closure and epithelial morphogenesis in Drosophila. Dev Biol 464(2): 188-201. PubMed ID: 32562757

    Choubey, P. K., Nandy, N., Pandey, A. and Roy, J. K. (2020). Rab11 plays a key role in stellate cell differentiation via non-canonical Notch pathway in Malpighian tubules of Drosophila melanogaster. Dev Biol. PubMed ID: 31911183

    Bhuin, T. and Roy, J. K. (2019). Developmental expression, co-localization and genetic interaction of exocyst component Sec15 with Rab11 during Drosophila development. Exp Cell Res. PubMed ID: 31071318

    Banerjee, A. and Roy, J. K. (2018). Bantam regulates the axonal geometry of Drosophila larval brain by modulating actin regulator enabled. Invert Neurosci 18(2): 7. PubMed ID: 29777401

    Choubey, P. K. and Roy, J. K. (2017). Rab11 is required for tubulogenesis of Malpighian tubules in Drosophila melanogaster. Genesis. PubMed ID: 28653473

    Banerjee, A. and Roy, J.K. (2017). Dicer-1 regulates proliferative potential of Drosophila larval neural stem cells through bantam miRNA based down-regulation of the G1/S inhibitor Dacapo. Dev Biol [Epub ahead of print]. PubMed ID: 28109717

    Choubey, P. K. and Roy, J. K. (2016). Rab11, a vesicular trafficking protein, affects endoreplication through Ras-mediated pathway in Drosophila melanogaster. Cell Tissue Res [Epub ahead of print]. PubMed ID: 27677270

  • Sougata Roy University of Maryland, College Park
    Patel, A., Wu, Y., Han, X., Su, Y., Maugel, T., Shroff, H. and Roy, S. (2022). Cytonemes coordinate asymmetric signaling and organization in the Drosophila muscle progenitor niche. Nat Commun 13(1): 1185. PubMed ID: 35246530

    Mandi, M., Khatun, S., Rajak, P., Mazumdar, A. and Roy, S. (2019). Potential risk of organophosphate exposure in male reproductive system of a non-target insect model Drosophila melanogaster. Environ Toxicol Pharmacol 74: 103308. PubMed ID: 31816565

    Sohr, A., Du, L., Wang, R., Lin, L. and Roy, S. (2019). Drosophila FGF cleavage is required for efficient intracellular sorting and intercellular dispersal. J Cell Biol. PubMed ID: 30808704

    Du, L., Sohr, A., Yan, G. and Roy, S. (2018). Feedback regulation of cytoneme-mediated transport shapes a tissue-specific FGF morphogen gradient. Elife 7. PubMed ID: 30328809

    Du, L., Zhou, A., Sohr, A. and Roy, S. (2018). An efficient strategy for generating tissue-specific binary transcription systems in Drosophila by genome editing. J Vis Exp(139). PubMed ID: 30295654

    Sarkar, S., Khatun, S., Dutta, M. and Roy, S. (2017). Trans-generational transmission of altered phenotype resulting from flubendiamide-induced changes in apoptosis in larval imaginal discs of Drosophila melanogaster. Environ Toxicol Pharmacol 56: 350-360. PubMed ID: 29121551

    Kashima, R., Redmond, P. L., Ghatpande, P., Roy, S., Kornberg, T. B., Hanke, T., Knapp, S., Lagna, G. and Hata, A. (2017). Hyperactive locomotion in a Drosophila model is a functional readout for the synaptic abnormalities underlying fragile X syndrome. Sci Signal 10(477). PubMed ID: 28465421

    Du, L., Zhou, A., Patel, A., Rao, M., Anderson, K. and Roy, S. (2017). Unique patterns of organization and migration of FGF-expressing cells during Drosophila morphogenesis. Dev Biol [Epub ahead of print]. PubMed ID: 28502613

    Kashima, R., Redmond, P. L., Ghatpande, P., Roy, S., Kornberg, T. B., Hanke, T., Knapp, S., Lagna, G. and Hata, A. (2017). Hyperactive locomotion in a Drosophila model is a functional readout for the synaptic abnormalities underlying fragile X syndrome. Sci Signal 10(477). PubMed ID: 28465421

    Rao, P. R., Lin, L., Huang, H., Guha, A., Roy, S. and Kornberg, T. B. (2015). Developmental compartments in the larval trachea of Drosophila. Elife 4. PubMed ID: 26491942

  • Julian Royet Aix-Marseille Université, CNRS, Institut de Biologie du Développement de Marseille-Luminy, Marseille
    Masuzzo, A., Manière, G., Grosjean, Y., Kurz, L. and Royet, J. (2022). Bacteria-Derived Peptidoglycan Triggers a Noncanonical Nuclear Factor-kappaB-Dependent Response in Drosophila Gustatory Neurons. J Neurosci 42(41): 7809-7823. PubMed ID: 36414007

    Charroux, B. and Royet, J. (2020). Protocol for a Binary Choice Feeding Assay Using Adult, Axenic Drosophila. STAR Protoc 1(3): 100117. PubMed ID: 33377013

    Zugasti, O., Tavignot, R. and Royet, J. (2020). Gut bacteria-derived peptidoglycan induces a metabolic syndrome-like phenotype via NF-κB-dependent insulin/PI3K signaling reduction in Drosophila renal system. Sci Rep 10(1): 14097. PubMed ID: 32839462

    Charroux, B., Daian, F. and Royet, J. (2020). Drosophila Aversive Behavior toward Erwinia carotovora carotovora Is Mediated by Bitter Neurons and Leukokinin. iScience 23(6): 101152. PubMed ID: 32450516

    Masuzzo, A., Maniere, G., Viallat-Lieutaud, A., Avazeri, E., Zugasti, O., Grosjean, Y., Kurz, C. L. and Royet, J. (2019). Peptidoglycan-dependent NF-kappaB activation in a small subset of brain octopaminergic neurons controls female oviposition. Elife 8. PubMed ID: 31661076

    El Fissi, N., Rojo, M., Aouane, A., Karatas, E., Poliacikova, G., David, C., Royet, J. and Rival, T. (2018). Mitofusin gain and loss of function drive pathogenesis in Drosophila models of CMT2A neuropathy. EMBO Rep. PubMed ID: 29898954

    Capo, F., Chaduli, D., Viallat-Lieutaud, A., Charroux, B. and Royet, J. (2017). Oligopeptide transporters of the SLC15 family are dispensable for peptidoglycan sensing and transport in Drosophila. J Innate Immun. PubMed ID: 28715804

    Kurz, C. L., Charroux, B., Chaduli, D., Viallat-Lieutaud, A. and Royet, J. (2017). Peptidoglycan sensing by octopaminergic neurons modulates Drosophila oviposition. Elife 6. PubMed ID: 28264763

    Tavignot, R., Chaduli, D., Djitte, F., Charroux, B. and Royet, J. (2017). Inhibition of a NF-κB/Diap1 pathway by PGRP-LF is required for proper apoptosis during Drosophila development.PLoS Genet [Epub ahead of print]. PubMed ID: 28085885

    Capo, F., Charroux, B. and Royet, J. (2016). Bacteria sensing mechanisms in Drosophila gut: Local and systemic consequences. Dev Comp Immunol [Epub ahead of print]. PubMed ID: 26778296

    Costechareyre, D., Capo, F., Fabre, A., Chaduli, D., Kellenberger, C., Roussel, A., Charroux, B. and Royet, J. (2015). Tissue-specific regulation of Drosophila NF-κB pathway activation by peptidoglycan recognition protein SC. J Innate Immun [Epub ahead of print]. PubMed ID: 26513145

  • Julio Rozas Departament de Genètica, Facultat de Biologia, Universitat de Barcelona
    Sanchez-Gracia, A., Guirao-Rico, S., Hinojosa-Alvarez, S. and Rozas, J. (2017). Computational prediction of the phenotypic effects of genetic variants: basic concepts and some application examples in Drosophila nervous system genes. J Neurogenet: 1-13. PubMed ID: 29168413

    Clifton, B. D., Librado, P., Yeh, S. D., Solares, E., Real, D., Jayasekera, S., Zhang, W., Shi, M., Park, R., Magie, R., Ma, H. C., Xia, X. Q., Marco, A., Rozas, J. and Ranz, J. M. (2016). Rapid functional and sequence differentiation of a tandemly-repeated species-specific multigene family in Drosophila. Mol Biol Evol [Epub ahead of print]. PubMed ID: 27702774

    Librado, P. and Rozas, J. (2016). Weak polygenic selection drives the rapid adaptation of the chemosensory system: lessons from the upstream regions of the major gene families. Genome Biol Evol [Epub ahead of print]. PubMed ID: 27503297

    Librado, P. and Rozas, J. (2013). Uncovering the functional constraints underlying the genomic organization of the odorant-binding protein genes. Genome Biol Evol 5: 2096-2108. PubMed ID: 24148943

    Almeida, F. C., Sanchez-Gracia, A., Campos, J. L. and Rozas, J. (2014). Family size evolution in Drosophila chemosensory gene families: a comparative analysis with a critical appraisal of methods. Genome Biol Evol 6: 1669-1682. PubMed ID: 24951565

    Yeh, S. D., Do, T., Chan, C., Cordova, A., Carranza, F., Yamamoto, E. A., Abbassi, M., Gandasetiawan, K. A., Librado, P., Damia, E., Dimitri, P., Rozas, J., Hartl, D. L., Roote, J. and Ranz, J. M. (2012). Functional evidence that a recently evolved Drosophila sperm-specific gene boosts sperm competition. Proc Natl Acad Sci U S A 109: 2043-2048. PubMed ID: 22308475

    Alvarez-Ponce, D., Guirao-Rico, S., Orengo, D. J., Segarra, C., Rozas, J. and Aguade, M. (2012). Molecular population genetics of the insulin/TOR signal transduction pathway: a network-level analysis in Drosophila melanogaster. Mol Biol Evol 29: 123-132. PubMed ID: 21680868

  • Gerry Rubin HHMI, Janelia Farm
    Kind, E., Longden, K. D., Nern, A., Zhao, A., Sancer, G., Flynn, M. A., Laughland, C. W., Gezahegn, B., Ludwig, H. D., Thomson, A. G., Obrusnik, T., Alarcon, P. G., Dionne, H., Bock, D. D., Rubin, G. M., Reiser, M. B. and Wernet, M. F. (2021). Synaptic targets of photoreceptors specialized to detect color and skylight polarization in Drosophila. Elife 10. PubMed ID: 34913436

    Hulse, B. K., Haberkern, H., Franconville, R., Turner-Evans, D. B., Takemura, S. Y., Wolff, T., Noorman, M., Dreher, M., Dan, C., Parekh, R., Hermundstad, A. M., Rubin, G. M. and Jayaraman, V. (2021). A connectome of the Drosophila central complex reveals network motifs suitable for flexible navigation and context-dependent action selection. Elife 10. PubMed ID: 34696823

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

    Schretter, C. E., Aso, Y., Robie, A. A., Dreher, M., Dolan, M. J., Chen, N., Ito, M., Yang, T., Parekh, R., Branson, K. M. and Rubin, G. M. (2020). Cell types and neuronal circuitry underlying female aggression in Drosophila. Elife 9. PubMed ID: 33141021

    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

    Wolff, T. and Rubin, G. M. (2018). Neuroarchitecture of the Drosophila central complex: A catalog of nodulus and asymmetrical body neurons and a revision of the protocerebral bridge catalog. J Comp Neurol. PubMed ID: 30084503

    Dionne, H., Hibbard, K. L., Cavallaro, A., Kao, J. C. and Rubin, G. M. (2018). Genetic Reagents for Making Split-GAL4 Lines in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 29535151

    Takemura, S. Y., Aso, Y., Hige, T., Wong, A., Lu, Z., Xu, C. S., Rivlin, P. K., Hess, H., Zhao, T., Parag, T., Berg, S., Huang, G., Katz, W., Olbris, D. J., Plaza, S., Umayam, L., Aniceto, R., Chang, L. A., Lauchie, S., Ogundeyi, O., Ordish, C., Shinomiya, A., Sigmund, C., Takemura, S., Tran, J., Turner, G. C., G. M. and Scheffer, L. K. (2017). A connectome of a learning and memory center in the adult Drosophila brain. Elife 6. PubMed ID: 28718765

    Robie, A. A., Hirokawa, J., Edwards, A. W., Umayam, L. A., Lee, A., Phillips, M. L., Card, G. M., Korff, W., Rubin, G. M., Simpson, J. H., Reiser, M. B. and Branson, K. (2017). Mapping the Neural Substrates of Behavior. Cell 170(2): 393-406.e328. PubMed ID: 28709004

    Takemura, S. Y., Nern, A., Chklovskii, D. B., Scheffer, L. K., Rubin, G. M. and Meinertzhagen, I. A. (2017). The comprehensive connectome of a neural substrate for 'ON' motion detection in Drosophila. Elife 6 [Epub ahead of print]. PubMed ID: 28432786

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

    Strother, J. A., Wu, S. T., Wong, A. M., Nern, A., Rogers, E. M., Le, J. Q., Rubin, G. M. and Reiser, M. B. (2017). The emergence of directional selectivity in the visual motion pathway of Drosophila. Neuron 94(1): 168-182.e110. PubMed ID: 28384470

    Sen, R., Wu, M., Branson, K., Robie, A., Rubin, G.M. and Dickson, B.J. (2017). Moonwalker descending neurons mediate visually evoked retreat in Drosophila. Curr Biol 27: 766-771. PubMed ID: 28238656

    Wu, M., Nern, A., Williamson, W. R., Morimoto, M. M., Reiser, M. B., Card, G. M. and Rubin, G. M. (2016). Visual projection neurons in the Drosophila lobula link feature detection to distinct behavioral programs. Elife 5. PubMed ID: 28029094

  • Douglas Ruden Department of Physiology, Wayne State University, Detroit, MI
    Shah, E. J., Huttemann, M., Sanderson, T. H., Gurdziel, K. and Ruden, D. M. (2021). Inhibiting Mitochondrial Cytochrome c Oxidase Downregulates Gene Transcription After Traumatic Brain Injury in Drosophila. Front Physiol 12: 628777. PubMed ID: 33790803

    Shah, E. J., Gurdziel, K. and Ruden, D. M. (2021). Sex-Differences in Traumatic Brain Injury in the Absence of Tau in Drosophila. Genes (Basel) 12(6). PubMed ID: 34198629

    >Shah, E. J., Gurdziel, K. and Ruden, D. M. (2020). Drosophila Exhibit Divergent Sex-Based Responses in Transcription and Motor Function After Traumatic Brain Injury. Front Neurol 11: 511. PubMed ID: 32636795

  • Alfredo Ruiz Universitat Autonoma de Barcelona
    Rius, N., Guillen, Y., Delprat, A., Kapusta, A., Feschotte, C. and Ruiz, A. (2016). Exploration of the Drosophila buzzatii transposable element content suggests underestimation of repeats in Drosophila genomes. BMC Genomics 17: 344. PubMed ID: 27164953

    Guillen, Y., et al. (2014). Genomics of ecological adaptation in cactophilic Drosophila. Genome Biol Evol [Epub ahead of print]. PubMed ID: 25552534

    Dias, G. B., Svartman, M., Delprat, A., Ruiz, A. and Kuhn, G. C. (2014). Tetris is a foldback transposon that provided the building blocks for an emerging satellite DNA of Drosophila virilis. Genome Biol Evol. PubMed ID: 24858539

    Rossato, D. O., Ludwig, A., Depra, M., Loreto, E. L., Ruiz, A. and Valente, V. L. (2014). BuT2 is a member of the third major group of hAT transposons and is involved in horizontal transfer events in the genus Drosophila. Genome Biol Evol. PubMed ID: 24459285

    Marzo, M., Bello, X., Puig, M., Maside, X., Ruiz, A. (2013) Striking structural dynamism and nucleotide sequence variation of the transposon Galileo in the genome of Drosophila mojavensis. Mob DNA 4: 6. PubMed ID: 23374229

    Calvete, O., Gonzalez, J., Betran, E. and Ruiz, A. (2012). Segmental duplication, microinversion, and gene loss associated with a complex inversion breakpoint region in Drosophila. Mol Biol Evol 29: 1875-1889. PubMed ID: 22328714

    Guillen, Y. and Ruiz, A. (2012). Gene alterations at Drosophila inversion breakpoints provide prima facie evidence for natural selection as an explanation for rapid chromosomal evolution. BMC Genomics 13: 53. PubMed ID: 22296923

  • Mar Ruiz-Gómez Centro de Biologia Molecular, Madrid
    Carrasco-Rando, M., Prieto-Sanchez, S., Culi, J., Tutor, A. S. and Ruiz-Gomez, M. (2019). A specific isoform of Pyd/ZO-1 mediates junctional remodeling and formation of slit diaphragms. J Cell Biol. PubMed ID: 31171632

    Tutor, A. S., Prieto-Sanchez, S. and Ruiz-Gomez, M. (2013). Src64B phosphorylates Dumbfounded and regulates slit diaphragm dynamics: Drosophila as a model to study nephropathies. Development. PubMed ID: 24335255

    Carrasco-Rando, M., Tutor, A. S., Prieto-Sanchez, S., Gonzalez-Perez, E., Barrios, N., Letizia, A., Martin, P., Campuzano, S. and Ruiz-Gomez, M. (2011). Drosophila araucan and caupolican integrate intrinsic and signalling inputs for the acquisition by muscle progenitors of the lateral transverse fate. PLoS Genet 7: e1002186. PubMed ID: 21811416

    Guruharsha, K. G., Ruiz-Gomez, M., Ranganath, H. A., Siddharthan, R. and Vijayraghavan, K. (2009). The complex spatio-temporal regulation of the Drosophila myoblast attractant gene duf/kirre. PLoS One 4: e6960. PubMed ID: 19742310

  • Eric Rulifson UCSF
    Makhijani, K., Alexander, B., Tanaka, T., Rulifson, E. and Bruckner, K. (2011). The peripheral nervous system supports blood cell homing and survival in the Drosophila larva. Development 138: 5379-5391. PubMed ID: 22071105

    Hwang, H. J. and Rulifson, E. (2011). Serial specification of diverse neuroblast identities from a neurogenic placode by Notch and Egfr signaling. Development 138: 2883-2893. PubMed ID: 21653613

  • Sebastian Rumpf Cells-in-Motion Cluster of Excellence, Münster, Germany
    Marzano, M., Herzmann, S., Elsbroek, L., Sanal, N., Tarbashevich, K., Raz, E., Krahn, M. P. and Rumpf, S. (2021). AMPK adapts metabolism to developmental energy requirement during dendrite pruning in Drosophila. Cell Rep 37(7): 110024. PubMed ID: 34788610

    Wolterhoff, N., Gigengack, U. and Rumpf, S. (2020). PP2A phosphatase is required for dendrite pruning via actin regulation in Drosophila. EMBO Rep: e48870. PubMed ID: 32207238

    Kramer, R., Rode, S. and Rumpf, S. (2019). Rab11 is required for neurite pruning and developmental membrane protein degradation in Drosophila sensory neurons. Dev Biol 451(1): 68-78. PubMed ID: 30871987

    Kramer, R., Rode, S. and Rumpf, S. (2019). Rab11 is required for neurite pruning and developmental membrane protein degradation in Drosophila sensory neurons. Dev Biol. PubMed ID: 30871987

    Zulbahar, S., Sieglitz, F., Kottmeier, R., Altenhein, B., Rumpf, S. and Klambt, C. (2018). Differential expression of Obek controls ploidy in the Drosophila blood-brain barrier. Development 145(15). PubMed ID: 30002129

    Rode, S., Ohm, H., Anhauser, L., Wagner, M., Rosing, M., Deng, X., Sin, O., Leidel, S. A., Storkebaum, E., Rentmeister, A., Zhu, S. and Rumpf, S. (2018). Differential Requirement for Translation Initiation Factor Pathways during Ecdysone-Dependent Neuronal Remodeling in Drosophila. Cell Rep 24(9): 2287-2299 e2284. PubMed ID: 30157424

    Herzmann, S., Gotzelmann, I., Reekers, L. F. and Rumpf, S. (2018). Spatial regulation of microtubule disruption during dendrite pruning in Drosophila. Development 145(9). PubMed ID: 29712642

  • Howard Rundle Department of Biology, University of Ottawa
    Videlier, M., Careau, V., Wilson, A. J. and Rundle, H. D. (2020). Quantifying selection on standard metabolic rate and body mass in Drosophila melanogaster. Evolution. PubMed ID: 33196104

    Yun, L., Chen, P. J., Singh, A., Agrawal, A. F. and Rundle, H. D. (2017). The physical environment mediates male harm and its effect on selection in females. Proc Biol Sci 284(1858). PubMed ID: 28679725

    Chenoweth, S. F., Appleton, N. C., Allen, S. L. and Rundle, H. D. (2015). Genomic evidence that sexual selection impedes adaptation to a novel environment. Curr Biol 25: 1860-1866. PubMed ID: 26119752

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

    Dyer, K. A., White, B. E., Sztepanacz, J. L., Bewick, E. R. and Rundle, H. D. (2013). Reproductive Character Displacement of Epicuticular Compounds and Their Contribution to Mate Choice in Drosophila Subquinaria and D. Recens. Evolution. PubMed ID: 24351014

    Arbuthnott, D., Dutton, E. M., Agrawal, A. F. and Rundle, H. D. (2013). The ecology of sexual conflict: ecologically dependent parallel evolution of male harm and female resistance in Drosophila melanogaster. Ecol Lett. PubMed ID: 24215269

    Punzalan, D., Delcourt, M. and Rundle, H. D. (2013). Comparing the intersex genetic correlation for fitness across novel environments in the fruit fly, Drosophila serrata. Heredity (Edinb). PubMed ID: 24045292

    Wong, A. and Rundle, H. (2013). Selection on the Drosophila seminal fluid protein Acp62F. Ecol Evol 3: 1942-1950. PubMed ID: 23919141

    Curtis, S., Sztepanacz, J. L., White, B. E., Dyer, K. A., Rundle, H. D. and Mayer, P. (2013). Epicuticular compounds of Drosophila subquinaria and D. recens: identification, quantification, and their role in female mate choice. J Chem Ecol 39: 579-590. PubMed ID: 23604703

    Sztepanacz, J. L. and Rundle, H. D. (2012). Reduced genetic variance among high fitness individuals: inferring stabilizing selection on male sexual displays in Drosophila serrata. Evolution 66: 3101-3110. PubMed ID: 23025601

  • Hannele Ruohola-Baker Department of Biochemistry, University of Washington, Seattle
    Ishibashi, J. R., Keshri, R., Taslim, T. H., Brewer, D. K., Chan, T. C., Lyons, S., McManamen, A. M., Chen, A., Del Castillo, D. and Ruohola-Baker, H. (2021). Chemical Genetic Screen in Drosophila Germline Uncovers Small Molecule Drugs That Sensitize Stem Cells to Insult-Induced Apoptosis. Cells 10(10). PubMed ID: 34685753

    Ishibashi, J. R., Keshri, R., Taslim, T. H., Brewer, D. K., Chan, T. C., Lyons, S., McManamen, A. M., Chen, A., Del Castillo, D. and Ruohola-Baker, H. (2021). Chemical Genetic Screen in Drosophila Germline Uncovers Small Molecule Drugs That Sensitize Stem Cells to Insult-Induced Apoptosis. Cells 10(10). PubMed ID: 34685753

    Artoni, F., Kreipke, R., Palmeira, O., Dixon, C., Goldberg, Z. and Ruohola-Baker, H. (2017). Loss of foxo rescues stem cell aging in Drosophila germ line. Elife 6. PubMed ID: 28925355

    Xing, Y., Su, T. T. and Ruohola-Baker, H. (2015). Tie-mediated signal from apoptotic cells protects stem cells in Drosophila melanogaster. Nat Commun 6: 7058. PubMed ID: 25959206

    Nguyen-Tran, D. H., Hait, N., Sperber, H., Qi, J., Fischer, K., Ieronimakis, N., Pantoja, M., Hays, A., Allegood, J., Reyes, M., Spiegel, S. and Ruohola-Baker, H. (2013). Molecular Mechanism of Sphingosine-1-Phosphate Action in Duchenne Muscular Dystrophy. Dis Model Mech. PubMed ID: 24077965

    Xing, Y., Kurtz, I., Thuparani, M., Legard, J. and Ruohola-Baker, H. (2012). Loss-of-Function Screen Reveals Novel Regulators Required for Drosophila Germline Stem Cell Self-Renewal. G3 (Bethesda) 2: 343-351. PubMed ID: 22413088

    Mosqueira, M., Willmann, G., Ruohola-Baker, H. and Khurana, T. S. (2010). Chronic hypoxia impairs muscle function in the Drosophila model of Duchenne's muscular dystrophy (DMD). PLoS One 5: e13450. PubMed ID: 20975992

    Chan, K. and Ruohola-Baker, H. (2010). Assessing in vivo microRNA function in the germline stem cells of the Drosophila ovary. Methods Mol Biol 650: 201-212. PubMed ID: 20686953

  • Nasser Rusan Laboratory of Molecular Machines and Tissue Architecture, NHLBI, NIH
    O'Neill, R. S. and Rusan, N. M. (2022). Traip controls mushroom body size by suppressing mitotic defects. Development 149(7). PubMed ID: 35297981

    O'Neill, R. S. and Rusan, N. M. (2022). Traip controls mushroom body size by suppressing mitotic defects. Development 149(7). PubMed ID: 35297981

    Galletta, B. J., Ortega, J. M., Smith, S. L., Fagerstrom, C. J., Fear, J. M., Mahadevaraju, S., Oliver, B. and Rusan, N. M. (2020). Sperm Head-Tail Linkage Requires Restriction of Pericentriolar Material to the Proximal Centriole End. Dev Cell. PubMed ID: 32169161

    Schoborg, T. A., Smith, S. L., Smith, L. N., Morris, H. D. and Rusan, N. M. (2019). Micro-computed tomography as a platform for exploring Drosophila development. Development. PubMed ID: 31722883

    Swider, Z. T., Ng, R. K., Varadarajan, R., Fagerstrom, C. J. and Rusan, N. M. (2019). Fascetto (PRC1) interacting protein (FIP) ensures proper cytokinesis and ploidy. Mol Biol Cell: mbcE18090573. PubMed ID: 30726162

    Boese, C. J., Nye, J., Buster, D. W., McLamarrah, T. A., Byrnes, A. E., Slep, K. C., Rusan, N. M. and Rogers, G. C. (2018). Asterless is a Polo-like kinase 4 substrate that both activates and inhibits kinase activity depending on its phosphorylation state. Mol Biol Cell: mbcE18070445. PubMed ID: 30256714

    Lerit, D.A., Shebelut, C.W., Lawlor, K.J., Rusan, N.M., Gavis, E.R., Schedl, P. and Deshpande, G. (2017). Germ Cell-less promotes centrosome segregation to induce germ cell formation. Cell Rep 18: 831-839. PubMed ID: 28122234

    Galletta, B. J., Fagerstrom, C. J., Schoborg, T. A., McLamarrah, T. A., Ryniawec, J. M., Buster, D. W., Slep, K. C., Rogers, G. C. and Rusan, N. M. (2016). A centrosome interactome provides insight into organelle assembly and reveals a non-duplication role for Plk4. Nat Commun 7: 12476. PubMed ID: 27558293

    Galletta, B.J., Jacobs, K.C., Fagerstrom, C.J. and Rusan, N.M. (2016). Asterless is required for centriole length control and sperm development. J Cell Biol [Epub ahead of print]. PubMed ID: 27185836

    Plevock, K.M., Galletta, B.J., Slep, K.C. and Rusan, N.M. (2015). Newly characterized region of CP190 associates with microtubules and mediates proper spindle morphology in Drosophila stem cells. PLoS One 10: e0144174. PubMed ID: 26649574

    Schoborg, T., Zajac, A.L., Fagerstrom, C.J., Guillen, R.X. and Rusan, N.M. (2015). An Asp-CaM complex is required for centrosome-pole cohesion and centrosome inheritance in neural stem cells. J Cell Biol [Epub ahead of print]. PubMed ID: 26620907

    Smyth, J. T., Schoborg, T. A., Bergman, Z. J., Riggs, B. and Rusan, N. M. (2015). Proper symmetric and asymmetric endoplasmic reticulum partitioning requires astral microtubules. Open Biol 5. PubMed ID: 26289801

    Lerit, D.A., Jordan, H.A., Poulton, J.S., Fagerstrom, C.J., Galletta, B.J., Peifer, M. and Rusan, N.M. (2015). Interphase centrosome organization by the PLP-Cnn scaffold is required for centrosome function. J Cell Biol 210: 79-97. PubMed ID: 26150390

    Lerit, D. A., Plevock, K. M. and Rusan, N. M. (2014). Live imaging of Drosophila larval neuroblasts. J Vis Exp. PubMed ID: 25046336

  • Christine Rushlow Department of Biology, New York University
    Atienza-Manuel, A., Castillo-Mancho, V., De Renzis, S., Culi, J. and Ruiz-Gomez, M. (2021). Endocytosis mediated by an atypical CUBAM complex modulates slit diaphragm dynamics in nephrocytes. Development. PubMed ID: 34738617

    Patel, A. L., Zhang, L., Keenan, S. E., Rushlow, C. A., Fradin, C. and Shvartsman, S. Y. (2021). Capicua is a fast-acting transcriptional brake. Curr Biol. PubMed ID: 34166605

    Yamada, S., Whitney, P. H., Huang, S. K., Eck, E. C., Garcia, H. G. and Rushlow, C. A. (2019). The Drosophila pioneer factor Zelda modulates the nuclear microenvironment of a Dorsal target enhancer to potentiate transcriptional output. Curr Biol 29(8): 1387-1393.e1385. PubMed ID: 30982648

    Djabrayan, N. J., Smits, C. M., Krajnc, M., Stern, T., Yamada, S., Lemon, W. C., Keller, P. J., Rushlow, C. A. and Shvartsman, S. Y. (2019). Metabolic Regulation of Developmental Cell Cycles and Zygotic Transcription. Curr Biol 29(7): 1193-1198.e1195. PubMed ID: 30880009

    Samee, M. A., Lim, B., Samper, N., Lu, H., Rushlow, C. A., Jimenez, G., Shvartsman, S. Y. and Sinha, S. (2015). A systematic ensemble approach to thermodynamic modeling of gene expression from sequence data. Cell Syst 1: 396-407. PubMed ID: 27136354

    Sun, Y., Nien, C.Y., Chen, K., Liu, H.Y., Johnston, J., Zeitlinger, J. and Rushlow, C. (2015). Zelda overcomes the high intrinsic nucleosome barrier at enhancers during Drosophila zygotic genome activation. Genome Res [Epub ahead of print]. PubMed ID: 26335633

    Foo, S. M., Sun, Y., Lim, B., Ziukaite, R., O'Brien, K., Nien, C. Y., Kirov, N., Shvartsman, S. Y. and Rushlow, C. A. (2014). Zelda Potentiates Morphogen Activity by Increasing Chromatin Accessibility. Curr Biol. PubMed ID: 24909324

    Fu, S., Nien, C. Y., Liang, H. L. and Rushlow, C. (2014). Co-activation of microRNAs by Zelda is essential for early Drosophila development. Development. PubMed ID: 24764079

    Lim, B., Samper, N., Lu, H., Rushlow, C., Jimenez, G. and Shvartsman, S. Y. (2013). Kinetics of gene derepression by ERK signaling. Proc Natl Acad Sci U S A. PubMed ID: 23733957

    Liang, H. L., Xu, M., Chuang, Y. C. and Rushlow, C. (2012). Response to the BMP gradient requires highly combinatorial inputs from multiple patterning systems in the Drosophila embryo. Development 139: 1956-1964. PubMed ID: 22513375

  • Steve Russell Department of Genetics, University of Cambridge
    Korona, D., Nightingale, D., Fabre, B., Nelson, M., Fischer, B., Johnson, G., Lees, J., Hubbard, S., Lilley, K. and Russell, S. (2020). Characterisation of protein isoforms encoded by the Drosophila Glycogen Synthase Kinase 3 gene shaggy. PLoS One 15(8): e0236679. PubMed ID: 32760087

    Chan, S. K. K., Cerda-Moya, G., Stojnic, R., Millen, K., Fischer, B., Fexova, S., Skalska, L., Gomez-Lamarca, M., Pillidge, Z., Russell, S. and Bray, S. J. (2017). Role of co-repressor genomic landscapes in shaping the Notch response. PLoS Genet 13(11): e1007096. PubMed ID: 29155828

    El-Sharnouby, S., Fischer, B., Magbanua, J. P., Umans, B., Flower, R., Choo, S. W., Russell, S. and White, R. (2017). Regions of very low H3K27me3 partition the Drosophila genome into topological domains. PLoS One 12(3): e0172725. PubMed ID: 28282436

    Niwa, H., Nakamura, A., Urata, M., Shirae-Kurabayashi, M., Kuraku, S., Russell, S. and Ohtsuka, S. (2016). The evolutionally-conserved function of group B1 Sox family members confers the unique role of Sox2 in mouse ES cells. BMC Evol Biol 16: 173. PubMed ID: 27582319

    Lee, H., Cho, D.Y., Whitworth, C., Eisman, R., Phelps, M., Roote, J., Kaufman, T., Cook, K., Russell, S., Przytycka, T. and Oliver, B. (2016). Effects of gene dose, chromatin, and network topology on expression in Drosophila melanogaster. PLoS Genet 12: e1006295. PubMed ID: 27599372

    Beh, C. Y., El-Sharnouby, S., Chatzipli, A., Russell, S., Choo, S. W. and White, R. (2016). Roles of cofactors and chromatin accessibility in Hox protein target specificity. Epigenetics Chromatin 9: 1. PubMed ID: 26753000
    Carl, S. H. and Russell, S. (2015). Common binding by redundant group B Sox proteins is evolutionarily conserved in Drosophila. BMC Genomics 16: 292. PubMed ID: 25887553

    Ferrero, E., Fischer, B. and Russell, S. (2014). SoxNeuro orchestrates central nervous system specification and differentiation in Drosophila and is only partially redundant with Dichaete. Genome Biol 15: R74. PubMed ID: 24886562

    Aleksic, J., Ferrero, E., Fischer, B., Shen, S. P. and Russell, S. (2013). The role of Dichaete in transcriptional regulation during Drosophila embryonic development. BMC Genomics 14: 861. PubMed ID: 24314314

    Chan, Y. S., Huen, D. S., Glauert, R., Whiteway, E., Russell, S. (2013) Optimising Homing Endonuclease Gene Drive Performance in a Semi-Refractory Species: The Drosophila melanogaster Experience. PLoS One 8: e54130. PubMed ID: 23349805

    Bossing, T., Barros, C. S., Fischer, B., Russell, S. and Shepherd, D. (2012). Disruption of microtubule integrity initiates mitosis during CNS repair. Dev Cell 23: 433-440. PubMed ID: 22841498

  • Tor Erik Rusten Oslo University Hospital
    Khezri, R., Holland, P., Schoborg, T. A., Abramovich, I., Takats, S., Dillard, C., Jain, A., O'Farrell, F., Schultz, S. W., Hagopian, W. M., Quintana, E. M., Ng, R., Katheder, N. S., Rahman, M. M., Teles Reis, J. G., Brech, A., Jasper, H., Rusan, N. M., Jahren, A. H., Gottlieb, E. and Rusten, T. E. (2021). Host autophagy mediates organ wasting and nutrient mobilization for tumor growth. Embo j: e107336. PubMed ID: 34309071

    O'Farrell, F., Lobert, V. H., Sneeggen, M., Jain, A., Katheder, N. S., Wenzel, E. M., Schultz, S. W., Tan, K. W., Brech, A., Stenmark, H. and Rusten, T. E. (2017). Class III phosphatidylinositol-3-OH kinase controls epithelial integrity through endosomal LKB1 regulation. Nat Cell Biol 19(12): 1412-1423. PubMed ID: 29084199

    Katheder, N. S., Khezri, R., O'Farrell, F., Schultz, S. W., Jain, A., Rahman, M. M., Schink, K. O., Theodossiou, T. A., Johansen, T., Juhasz, G., Bilder, D., Brech, A., Stenmark, H. and Rusten, T. E. (2017). Microenvironmental autophagy promotes tumour growth. Nature. PubMed ID: 28077876

    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(7): e0159473. PubMed ID: 27467079

    Jain, A., Rusten, T. E., Katheder, N., Elvenes, J., Bruun, J. A., Sjottem, E., Lamark, T. and Johansen, T. (2015). p62/Sequestosome-1, Autophagy-related Gene 8, and Autophagy in Drosophila Are Regulated by Nuclear Factor Erythroid 2-related Factor 2 (NRF2), Independent of Transcription Factor TFEB. J Biol Chem 290(24): 14945-14962. PubMed ID: 25931115

    Morelli, E., Ginefra, P., Mastrodonato, V., Beznoussenko, G. V., Rusten, T. E., Bilder, D., Stenmark, H., Mironov, A. A. and Vaccari, T. (2014). Multiple functions of the SNARE protein Snap29 in autophagy, endocytic, and exocytic trafficking during epithelial formation in Drosophila. Autophagy 10(12): 2251-2268. PubMed ID: 25551675

  • Vanessa Ruta Laboratory of Neurophysiology and Behavior, Rockefeller University
    Zolin, A., Cohn, R., Pang, R., Siliciano, A. F., Fairhall, A. L. and Ruta, V. (2021). Context-dependent representations of movement in Drosophila dopaminergic reinforcement pathways. Nat Neurosci 24(11): 1555-1566. PubMed ID: 34697455

    Seeholzer, L. F., Seppo, M., Stern, D. L. and Ruta, V. (2018). Evolution of a central neural circuit underlies Drosophila mate preferences. Nature 559(7715): 564-569. PubMed ID: 29995860

    Cohn, R., Morantte, I. and Ruta, V. (2015). Coordinated and compartmentalized neuromodulation shapes sensory processing in Drosophila. Cell 163: 1742-1755. PubMed ID: 26687359

    Clowney, E. J., Iguchi, S., Bussell, J. J., Scheer, E. and Ruta, V. (2015). Multimodal chemosensory circuits controlling male courtship in Drosophila. Neuron 87: 1036-1049. PubMed ID: 26279475

    Caron, S. J., Ruta, V., Abbott, L. F. and Axel, R. (2013). Random convergence of olfactory inputs in the Drosophila mushroom body. Nature 497: 113-117. PubMed ID: 23615618

    Ruta, V., Datta, S. R., Vasconcelos, M. L., Freeland, J., Looger, L. L. and Axel, R. (2010). A dimorphic pheromone circuit in Drosophila from sensory input to descending output. Nature 468(7324): 686-690. PubMed ID: 21124455

  • Stefanie Ryglewski Institue of Developmental Biology and Neurobiology, University of Mainz, Germany
    Heinrich, L. and Ryglewski, S. (2020). Different functions of two putative Drosophila alpha2delta subunits in the same identified motoneurons. Sci Rep 10(1): 13670. PubMed ID: 32792569

    Werner, J., Arian, J., Bernhardt, I., Ryglewski, S. and Duch, C. (2020). Differential localization of voltage-gated potassium channels during Drosophila metamorphosis. J Neurogenet 34(1): 133-150. PubMed ID: 31997675

    Schutzler, N., Girwert, C., Hugli, I., Mohana, G., Roignant, J. Y., Ryglewski, S. and Duch, C. (2019). Tyramine action on motoneuron excitability and adaptable tyramine/octopamine ratios adjust Drosophila locomotion to nutritional state. Proc Natl Acad Sci U S A 116(9): 3805-3810. PubMed ID: 30808766

  • Hyung Don Ryoo NYU Langone Medical Center
    Vasudevan, D., Katow, H., Huang, H. W., Tang, G. and Ryoo, H. D. (2021). A Protein-trap allele reveals roles for Drosophila ATF4 in photoreceptor degeneration, oogenesis and wing development. Dis Model Mech. PubMed ID: 34919148

    Huang, H. W. and Ryoo, H. D. (2021). Drosophila fabp is required for light-dependent Rhodopsin-1 clearance and photoreceptor survival. PLoS Genet 17(10): e1009551. PubMed ID: 34714826

    Brown, B., Mitra, S., Roach, F. D., Vasudevan, D. and Ryoo, H. D. (2021). The transcription factor Xrp1 is required for PERK-mediated antioxidant gene induction in Drosophila. Elife 10. PubMed ID: 34605405

    Mitra, S. and Ryoo, H. D. (2021). The role of Ire1 in Drosophila eye pigmentation revealed by an RNase dead allele. Dev Biol. PubMed ID: 34265355

    Vasudevan, D., Neuman, S. D., Yang, A., Lough, L., Brown, B., Bashirullah, A., Cardozo, T. and Ryoo, H. D. (2020). Translational induction of ATF4 during integrated stress response requires noncanonical initiation factors eIF2D and DENR. Nat Commun 11(1): 4677. PubMed ID: 32938929

    Huang, H. W., Brown, B., Chung, J., Domingos, P. M. and Ryoo, H. D. (2018). highroad is a carboxypetidase induced by retinoids to clear mutant Rhodopsin-1 in Drosophila retinitis pigmentosa models. Cell Rep 22(6): 1384-1391. PubMed ID: 29425495

    Vasudevan, D., Clark, N. K., Sam, J., Cotham, V. C., Ueberheide, B., Marr, M. T., and Ryoo, H. D. (2017). The GCN2-ATF4 signaling pathway induces 4E-BP to bias translation and boost antimicrobial peptide synthesis in response to bacterial infection. Cell Rep 21(8): 2039-2047. PubMed ID: 29166596

    Huang, H. W., Zeng, X., Rhim, T., Ron, D. and Ryoo, H. D. (2017). The requirement of IRE1 and XBP1 in resolving physiological stress during Drosophila development. J Cell Sci 130(18): 3040-3049. PubMed ID: 28775151

    Kang, M. J., Vasudevan, D., Kang, K., Kim, K., Park, J. E., Zhang, N., Zeng, X., Neubert, T. A., Marr, M. T., and Don Ryoo, H. (2016). 4E-BP is a target of the GCN2-ATF4 pathway during Drosophila development and aging. J Cell Biol. PubMed ID: 27979906

    Zhang, T., Liao, Y., Hsu, F. N., Zhang, R., Searle, J. S., Pei, X., Li, X., Ryoo, H. D., Ji, J. Y. and Du, W. (2014). Hyperactivated Wnt signaling induces synthetic lethal interaction with Rb inactivation by elevating TORC1 activities. PLoS Genet 10: e1004357. PubMed ID: 24809668



  • This listing of Fly Labs and References is a work in progress. Please contact Tom Brody ([email protected]) with your suggestions or corrections.

    Return to The Interactive Fly