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


  • Tetsuya Tabata Institute of Molecular and Cellular Biosciences, University of Tokyo
    Yamazaki, D., Hiroi, M., Abe, T., Shimizu, K., Minami-Ohtsubo, M., Maeyama, Y., Horiuchi, J. and Tabata, T. (2018). Two Parallel Pathways Assign Opposing Odor Valences during Drosophila Memory Formation. Cell Rep 22(9): 2346-2358. PubMed ID: 29490271

    Ueoka, Y., Hiroi, M., Abe, T. and Tabata, T. (2017). Suppression of a single pair of mushroom body output neurons in Drosophila triggers aversive associations. FEBS Open Bio 7(4): 562-576. PubMed ID: 28396840

    Murakami, S., Minami-Ohtsubo, M., Nakato, R., Shirahige, K. and Tabata, T. (2017). Two components of aversive memory in Drosophila, anesthesia-sensitive and anesthesia-resistant memory, require distinct domains within the Rgk1 small GTPase. J Neurosci. PubMed ID: 28416593

    Nitta, Y., Yamazaki, D., Sugie, A., Hiroi, M. and Tabata, T. (2016). DISCO Interacting Protein 2 regulates axonal bifurcation and guidance of Drosophila mushroom body neurons. Dev Biol. PubMed ID: 27908785

    Abe, T., Yamazaki, D., Murakami, S., Hiroi, M., Nitta, Y., Maeyama, Y. and Tabata, T. (2014). The NAV2 homolog Sickie regulates F-actin-mediated axonal growth in Drosophila mushroom body neurons via the non-canonical Rac-Cofilin pathway. Development. PubMed ID: 25411210

    Hiroi, M., Ohkura, M., Nakai, J., Masuda, N., Hashimoto, K., Inoue, K., Fiala, A. and Tabata, T. (2013). Principal component analysis of odor coding at the level of third-order olfactory neurons in Drosophila. Genes Cells. PubMed ID: 24118654

    Kawamori, H., Tai, M., Sato, M., Yasugi, T. and Tabata, T. (2011). Fat/Hippo pathway regulates the progress of neural differentiation signaling in the Drosophila optic lobe. Dev. Growth Differ. 53(5): 653-67. PubMed Citation: 21671914

    Ogiso, Y., Tsuneizumi, K., Masuda, N., Sato, M. and Tabata, T. (2011). Robustness of the Dpp morphogen activity gradient depends on negative feedback regulation by the inhibitory Smad, Dad. Dev Growth Differ 53: 668-678. PubMed ID: 21671915

    Shimizu, K., Sato, M. and Tabata, T. (2011). The Wnt5/planar cell polarity pathway regulates axonal development of the Drosophila mushroom body neuron. J Neurosci 31: 4944-4954. PubMed ID: 21451033

  • Paul Taghert
    Yang, S., McAdow, J., Du, Y., Trigg, J., Taghert, P. H. and Johnson, A. N. (2022). Spatiotemporal expression of regulatory kinases directs the transition from mitosis to cellular morphogenesis in Drosophila. Nat Commun 13(1): 772. PubMed ID: 35140224

    Liang, X., Ho, M. C. W., Zhang, Y., Li, Y., Wu, M. N., Holy, T. E. and Taghert, P. H. (2019). Morning and evening circadian pacemakers independently drive premotor centers via a specific dopamine Relay. Neuron. PubMed ID: 30981533

    Liang, X., Holy, T. E. and Taghert, P. H. (2017). A series of suppressive signals within the Drosophila circadian neural circuit generates sequential daily Outputs. Neuron 94(6): 1173-1189.e1174. PubMed ID: 28552314

    Klose, M., Duvall, L. B., Li, W., Liang, X., Ren, C., Steinbach, J. H. and Taghert, P. H. (2016). Functional PDF signaling in the Drosophila circadian neural circuit is gated by Ral A-dependent modulation. Neuron [Epub ahead of print]. PubMed ID: 27161526

    Liang, X., Holy, T.E. and Taghert, P.H. (2016). Synchronous Drosophila circadian pacemakers display nonsynchronous Ca²⁺ rhythms in vivo. Science 351: 976-981. PubMed ID: 26917772

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

    Beebe, K., Park, D., Taghert, P. H. and Micchelli, C. A. (2015). The Drosophila pro-secretory transcription factor dimmed is dynamically regulated in adult enteroendocrine cells and protects against gram-negative infection. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 25999585

    Hadzic, T., Park, D., Abruzzi, K. C., Yang, L., Trigg, J. S., Rohs, R., Rosbash, M. and Taghert, P. H. (2015). Genome-wide features of neuroendocrine regulation in Drosophila by the basic helix-loop-helix transcription factor DIMMED. Nucleic Acids Res. PubMed ID: 25634895

    Park, D., Li, P., Dani, A. and Taghert, P. H. (2014). Peptidergic Cell-Specific Synaptotagmins in Drosophila: Localization to Dense-Core Granules and Regulation by the bHLH Protein DIMMED. J Neurosci 34: 13195-13207. PubMed ID: 25253864

    Duvall, L. B. and Taghert, P. H. (2013). E and M Circadian Pacemaker Neurons Use Different PDF Receptor Signalosome Components in Drosophila. J Biol Rhythms 28: 239-248. PubMed ID: 23929551

    Taghert, P. H. and Nitabach, M. N. (2012). Peptide neuromodulation in invertebrate model systems. Neuron 76: 82-97. PubMed ID: 23040808

  • Jussi Taipale Karolinska Institute
    Nitta, K.R., Jolma, A., Yin, Y., Morgunova, E., Kivioja, T., Akhtar, J., Hens, K., Toivonen, J., Deplancke, B., Furlong, E.E. and Taipale, J. (2015). Conservation of transcription factor binding specificities across 600 million years of bilateria evolution. Elife 4. PubMed ID: 25779349

    Bonke, M., Turunen, M., Sokolova, M., Vaharautio, A., Kivioja, T., Taipale, M., Bjorklund, M., Taipale, J. (2013) Transcriptional networks controlling the cell cycle. G3 (Bethesda) 3: 75-90. PubMed ID: 23316440

  • Aya Takahashi Evolutionary Genetics Laboratory, Department of Biological Sciences, School of Science and Engineering, Tokyo Metropolitan University
    Akiyama, N., Sato, S., Tanaka, K. M., Sakai, T. and Takahashi, A. (2022). The role of the epidermis enhancer element in positive and negative transcriptional regulation of ebony in Drosophila melanogaster. G3 (Bethesda) 12(3). PubMed ID: 35100378

    Massey, J. H., Akiyama, N., Bien, T., Dreisewerd, K., Wittkopp, P. J., Yew, J. Y. and Takahashi, A. (2019). Pleiotropic effects of ebony and tan on pigmentation and cuticular hydrocarbon composition in Drosophila melanogaster. Front Physiol 10: 518. PubMed ID: 31118901

    Tanaka, K. M., Kamimura, Y. and Takahashi, A. (2018). Mechanical incompatibility caused by modifications of multiple male genital structures using genomic introgression in Drosophila. Evolution. PubMed ID: 30198555

    Muto, L., Kamimura, Y., Tanaka, K. M. and Takahashi, A. (2018). An innovative ovipositor for niche exploitation impacts genital coevolution between sexes in a fruit-damaging Drosophila. Proc Biol Sci 285(1887). PubMed ID: 30257912

    Osada, N., Miyagi, R. and Takahashi, A. (2017). Cis- and trans-regulatory effects on gene expression in a natural population of Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 28615283

    Sunaga, S., Akiyama, N., Miyagi, R. and Takahashi, A. (2016). Factors underlying natural variation in body pigmentation of Drosophila melanogaster. Genes Genet Syst. PubMed ID: 27021917

    Miyagi, R., Akiyama, N., Osada, N. and Takahashi, A. (2015). Complex patterns of cis-regulatory polymorphisms in ebony underlie standing pigmentation variation in Drosophila melanogaster. Mol Ecol. PubMed ID: 26503353

    Tanaka, K. M., Takahashi, A., Fuse, N. and Takano-Shimizu-Kouno, T. (2014). A novel cell death gene acts to repair patterning defects in Drosophila melanogaster. Genetics 197: 739-742. PubMed ID: 24671768

    Takahashi, A. (2013). Pigmentation and behavior: potential association through pleiotropic genes in Drosophila. Genes Genet Syst 88: 165-174. PubMed ID: 24025245

    Isobe, K., Takahashi, A. and Tamura, K. (2013). Cold tolerance and metabolic rate increased by cold acclimation in Drosophila albomicans from natural populations. Genes Genet Syst 88: 289-300. PubMed ID: 24694392

  • Kazuo Takahashi Graduate School of Environmental and Life Science, Okayama University, Japan
    Kidera, H., Hatabu, T. and Takahashi, K. H. (2020). Apoptosis inhibition mitigates aging effects in Drosophila melanogaster. Genetica 148(2): 69-76. PubMed ID: 32219590

    Takahashi, K. H., Ishimori, M. and Iwata, H. (2018). HSP90 as a global genetic modifier for male genital morphology in Drosophila melanogaster. Evolution 72(11): 2419-2434. PubMed ID: 30221481

    Yamashita, S., Takigahira, T. and Takahashi, K. H. (2018). Genome-wide association analysis of host genotype and plastic wing morphological variation of an endoparasitoid wasp Asobara japonica (Hymenoptera: Braconidae). Genetica 146(3): 313-321. PubMed ID: 29748763

    Takahashi, K. H. (2017). Little effect of HSP90 inhibition on the quantitative wing traits variation in Drosophila melanogaster. Genetica 145(1): 9-18. PubMed ID: 27909948

  • John Tamkun Molecular, Cell and Developmental Biology, U. C. at Santa Cruz
    Kingston, R. E. and Tamkun, J. W. (2014). Transcriptional regulation by trithorax-group proteins. Cold Spring Harb Perspect Biol 6: a019349. PubMed ID: 25274705

    Siriaco, G., Deuring, R., Mawla, G. D. and Tamkun, J. W. (2015). A novel approach for studying Histone H1 function in vivo. Genetics [Epub ahead of print]. PubMed ID: 25805849

    Siriaco, G. and Tamkun, J. W. (2013). A Histone Timer for Zygotic Genome Activation. Dev Cell 26: 558-559. PubMed ID: 24091009

    Dorighi, K. M. and Tamkun, J. W. (2013). The trithorax group proteins Kismet and ASH1 promote H3K36 dimethylation to counteract Polycomb group repression in Drosophila. Development. PubMed ID: 24004944

    Fasulo, B., Deuring, R., Murawska, M., Gause, M., Dorighi, K. M., Schaaf, C. A., Dorsett, D., Brehm, A. and Tamkun, J. W. (2012). The Drosophila MI-2 chromatin-remodeling factor regulates higher-order chromatin structure and cohesin dynamics in vivo. PLoS Genet 8: e1002878. PubMed ID: 22912596

    Siriaco, G., Deuring, R., Chioda, M., Becker, P. B. and Tamkun, J. W. (2009). Drosophila ISWI regulates the association of histone H1 with interphase chromosomes in vivo. Genetics 182: 661-669. PubMed ID: 19380479

    Srinivasan, S., Dorighi, K. M. and Tamkun, J. W. (2008). Drosophila Kismet regulates histone H3 lysine 27 methylation and early elongation by RNA polymerase II. PLoS Genet 4: e1000217. PubMed ID: 18846226

  • Eng King Tan Duke-NUS Medical School, Singapore
    Liu, W., Wang, J., Zhang, H. Y., Yang, Y. C., Kang, R. X., Bai, P., Fu, H., Chen, L. R., Gao, Y. P. and Tan, E. K. (2020). Symbiotic bacteria attenuate Drosophila oviposition repellence to alkaline through acidification. Insect Sci. PubMed ID: 32725723

    Tio, M., Wen, R., Lim, Y. L., Wang, H., Ling, S. C., Zhao, Y. and Tan, E. K. (2016). FUS-linked essential tremor associated with motor dysfunction in Drosophila. Hum Genet. PubMed ID: 27395408

    Wang, H. S., Toh, J., Ho, P., Tio, M., Zhao, Y. and Tan, E. K. (2014). In vivo evidence of pathogenicity of VPS35 mutations in the Drosophila. Mol Brain 7: 73. PubMed ID: 25288323

    Angeles, D. C., Ho, P., Chua, L. L., Wang, C., Yap, Y. W., Ng, C., Zhou, Z., Lim, K. L., Wszolek, Z. K., Wang, H. Y. and Tan, E. K. (2014). Thiol peroxidases ameliorate LRRK2 mutant-induced mitochondrial and dopaminergic neuronal degeneration in Drosophila. Hum Mol Genet 23: 3157-3165. PubMed ID: 24459295

  • Nobuaki Tanaka Hokkaido University
    Sugime, Y., Watanabe, D., Yasuno, Y., Shinada, T., Miura, T. and Tanaka, N. K. (2017). Upregulation of juvenile hormone titers in female Drosophila melanogaster through mating experiences and host food occupied by eggs and larvae. Zoolog Sci 34(1): 52-57. PubMed ID: 28148219
    Yagi, R., Mabuchi, Y., Mizunami, M. and Tanaka, N. K. (2016). Convergence of multimodal sensory pathways to the mushroom body calyx in Drosophila melanogaster. Sci Rep 6: 29481. PubMed ID: 27404960

    Tanaka, N. K., Endo, K. and Ito, K. (2012). Organization of antennal lobe-associated neurons in adult Drosophila melanogaster brain. J Comp Neurol 520: 4067-4130. PubMed ID: 22592945

    Tanaka, N. K., Suzuki, E., Dye, L., Ejima, A. and Stopfer, M. (2012). Dye fills reveal additional olfactory tracts in the protocerebrum of wild-type Drosophila. J Comp Neurol 520: 4131-4140. PubMed ID: 22592823

  • Guy Tanentzapf Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver
    Ho, K. Y. L., Khadilkar, R. J., Carr, R. L. and Tanentzapf, G. (2021). A gap-junction-mediated, calcium-signaling network controls blood progenitor fate decisions in hematopoiesis. Curr Biol. PubMed ID: 34480855

    Khadilkar, R. J., Ho, K. Y. L., Venkatesh, B. and Tanentzapf, G. (2020). Integrins Modulate Extracellular Matrix Organization to Control Cell Signaling during Hematopoiesis. Curr Biol. PubMed ID: 32649911

    Khadilkar, R. J. and Tanentzapf, G. (2019). Septate junction components control Drosophila hematopoiesis through the Hippo pathway. Development. PubMed ID: 30890573

    Camp, D., Haage, A., Solianova, V., Castle, W. M., Xu, Q. A., Lostchuck, E., Goult, B. T. and Tanentzapf, G. (2018). Direct binding of Talin to Rap1 is required for cell-ECM adhesion in Drosophila. J Cell Sci. PubMed ID: 30446511

    Fairchild, M. J., Islam, F. and Tanentzapf, G. (2017). Identification of genetic networks that act in the somatic cells of the testis to mediate the developmental program of spermatogenesis. PLoS Genet 13(9): e1007026. PubMed ID: 28957323

    Goodwin, K., Lostchuck, E. E., Cramb, K. M., Zulueta-Coarasa, T., Fernandez-Gonzalez, R. and Tanentzapf, G. (2017). Cell-cell and cell-ECM adhesions cooperate to organize actomyosin networks and maintain force transmission during Dorsal Closure. Mol Biol Cell [Epub ahead of print]. PubMed ID: 28331071
    Goodwin, K., Ellis, S. J., Lostchuck, E., Zulueta-Coarasa, T., Fernandez-Gonzalez, R. and Tanentzapf, G. (2016). Basal cell-extracellular matrix adhesion regulates force transmission during tissue morphogenesis. Dev Cell 39(5): 611-625. PubMed ID: 27923121

    Hakonardottir, G. K., Lopez-Ceballos, P., Herrera-Reyes, A. D., Das, R., Coombs, D. and Tanentzapf, G. (2015). In vivo quantitative analysis of Talin turnover in response to force. Mol Biol Cell 26: 4149-4162. PubMed ID: 26446844

    Fairchild, M. J., Yang, L., Goodwin, K. and Tanentzapf, G. (2016). Occluding junctions maintain stem cell niche homeostasis in the fly testes. Curr Biol [Epub ahead of print]. PubMed ID: 27546574

    Smendziuk, C. M., Messenberg, A., Vogl, W. and Tanentzapf, G. (2015). Bi-directional gap junction-mediated Soma-Germline communication is essential for spermatogenesis. Development. PubMed ID: 26116660

  • Amy Tang Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk
    Tang, A. H. (2011). Are you my friends or are you my enemies? Self Nonself 2: 142-146. PubMed ID: 22496931

    Schmidt, R. L., Rinaldo, F. M., Hesse, S. E., Hamada, M., Ortiz, Z., Beleford, D. T., Page-McCaw, A., Platt, J. L. and Tang, A. H. (2011). Cleavage of PGRP-LC receptor in the Drosophila IMD pathway in response to live bacterial infection in S2 cells. Self Nonself 2: 125-141. PubMed ID: 22496930

    Ahmed, A. U., Schmidt, R. L., Park, C. H., Reed, N. R., Hesse, S. E., Thomas, C. F., Molina, J. R., Deschamps, C., Yang, P., Aubry, M. C. and Tang, A. H. (2008). Effect of disrupting seven-in-absentia homolog 2 function on lung cancer cell growth. J Natl Cancer Inst 100: 1606-1629. PubMed ID: 19001609

  • Tian Tang State Key Laboratory of Biocontrol and Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
    Huang, Y., Shang, R., Lu, G. A., Zeng, W., Huang, C., Zou, C. and Tang, T. (2022). Spatiotemporal Regulation of a Single Adaptively Evolving Trans-Regulatory Element Contributes to Spermatogenetic Expression Divergence in Drosophila. Mol Biol Evol 39(7). PubMed ID: 35687719

    >Lyu, Y., Liufu, Z., Xiao, J. and Tang, T. (2021). A Rapid Evolving microRNA Cluster Rewires Its Target Regulatory Networks in Drosophila. Front Genet 12: 760530. PubMed ID: 34777478

    Dai, A., Wang, Y., Greenberg, A., Liufu, Z. and Tang, T. (2021). Rapid Evolution of Autosomal Binding Sites of the Dosage Compensation Complex in Drosophila melanogaster and Its Association With Transcription Divergence. Front Genet 12: 675027. PubMed ID: 34194473

    Zhao, Y., Lu, G. A., Yang, H., Lin, P., Liufu, Z., Tang, T. and Xu, J. (2021). Run or Die in the Evolution of New MicroRNAs-Testing the Red Queen Hypothesis on De Novo New Genes. Mol Biol Evol 38(4): 1544-1553. PubMed ID: 33306129

  • Robert Tanguay Cell and Developmental Genetics Laboratory, Pavillon C-E. Marchand Université Laval, Sainte-Foy, Québec
    Dabbaghizadeh, A., Morrow, G., Amer, Y. O., Chatelain, E. H., Pichaud, N. and Tanguay, R. M. (2018). Identification of proteins interacting with the mitochondrial small heat shock protein Hsp22 of Drosophila melanogaster: Implication in mitochondrial homeostasis. PLoS One 13(3): e0193771. PubMed ID: 29509794

    Moutaoufik, M. T., Morrow, G., Finet, S. and Tanguay, R. M. (2017). Effect of N-terminal region of nuclear Drosophila melanogaster small heat shock protein DmHsp27 on function and quaternary structure. PLoS One 12(5): e0177821. PubMed ID: 28520783

    Morrow, G., Kim, H. J., Pellerito, O., Bourrelle-Langlois, M., Le Pecheur, M., Groebe, K. and Tanguay, R. M. (2016). Changes in Drosophila mitochondrial proteins following chaperone-mediated lifespan extension confirm a role of Hsp22 in mitochondrial UPR and reveal a mitochondrial localization for cathepsin D. Mech Ageing Dev 155: 36-47. PubMed ID: 26930296

    Morrow, G., Le Pecheur, M. and Tanguay, R. M. (2015). Drosophila melanogaster mitochondrial Hsp22: a role in resistance to oxidative stress, aging and the mitochondrial unfolding protein response. Biogerontology [Epub ahead of print]. PubMed ID: 26155908

    Morrow, G. and Tanguay, R. M. (2015). Drosophila melanogaster Hsp22: a mitochondrial small heat shock protein influencing the aging process. Front Genet 6: 1026. PubMed ID: 25852752

    Kim, A. Y., Seo, J. B., Kim, W. T., Choi, H. J., Kim, S. Y., Morrow, G., Tanguay, R. M., Steller, H. and Koh, Y. H. (2015). The pathogenic human Torsin A in Drosophila activates the unfolded protein response and increases susceptibility to oxidative stress. BMC Genomics 16: 338. PubMed ID: 25903460

    Zhang, D., Ke, L., Mackovicova, K., Van Der Want, J. J., Sibon, O. C., Tanguay, R. M., Morrow, G., Henning, R. H., Kampinga, H. H. and Brundel, B. J. (2011). Effects of different small HSPB members on contractile dysfunction and structural changes in a Drosophila melanogaster model for Atrial Fibrillation. J Mol Cell Cardiol 51: 381-389. PubMed ID: 21745477

  • Hiromu Tanimoto Laboratory of Neuroethology Graduate School of Life Sciences, Tohoku University, Sendai
    Tanizawa, F. and Takemoto, H. (2021). Sleep contributes to preference for novel food odours in Drosophila melanogaster. Sci Rep 11(1): 9395. PubMed ID: 33931708

    Ikarashi, M. and Tanimoto, H. (2021). Drosophila acquires seconds-scale rhythmic behavior. J Exp Biol. PubMed ID: 33795422

    Kanno, M., Hiramatsu, S., Kondo, S., Tanimoto, H. and Ichinose, T. (2021). Voluntary intake of psychoactive substances is regulated by the dopamine receptor Dop1R1 in Drosophila. Sci Rep 11(1): 3432. PubMed ID: 33564023

    Ichinose, T., Kanno, M., Wu, H., Yamagata, N., Sun, H., Abe, A. and Tanimoto, H. (2021). Mushroom body output differentiates memory processes and distinct memory-guided behaviors. Curr Biol. PubMed ID: 33476556

    Sun, H., Nishioka, T., Hiramatsu, S., Kondo, S., Amano, M., Kaibuchi, K., Ichinose, T. and Tanimoto, H. (2020). Dopamine receptor Dop1R2 stabilizes appetitive olfactory memory through the Raf/MAPK pathway in Drosophila. J Neurosci. PubMed ID: 32102921

    Kondo, S., Takahashi, T., Yamagata, N., Imanishi, Y., Katow, H., Hiramatsu, S., Lynn, K., Abe, A., Kumaraswamy, A. and Tanimoto, H. (2020). Neurochemical Organization of the Drosophila Brain Visualized by Endogenously Tagged Neurotransmitter Receptors. Cell Rep 30(1): 284-297.e285. PubMed ID: 31914394

    Katow, H., Takahashi, T., Saito, K., Tanimoto, H. and Kondo, S. (2019). Tango knock-ins visualize endogenous activity of G protein-coupled receptors in Drosophila. J Neurogenet: 1-8. PubMed ID: 31084242

    Thoma, V., Kobayashi, K. and Tanimoto, H. (2017). The role of the gustatory system in the coordination of feeding. eNeuro 4(6). PubMed ID: 29159281

    Yamagata, N., Hiroi, M., Kondo, S., Abe, A. and Tanimoto, H. (2016). Suppression of dopamine neurons mediates reward. PLoS Biol 14: e1002586. PubMed ID: 27997541

    Ichinose, T. and Tanimoto, H. (2016). Dynamics of memory-guided choice behavior in Drosophila. Proc Jpn Acad Ser B Phys Biol Sci 92: 346-357. PubMed ID: 27725473

    Vogt, K., Aso, Y., Hige, T., Knapek, S., Ichinose, T., Friedrich, A. B., Turner, G. C., Rubin, G. M. and Tanimoto, H. (2016). Direct neural pathways convey distinct visual information to mushroom bodies. Elife 5. PubMed ID: 27083044

  • Teiichi Tanimura Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka
    Kudow, N., Kamikouchi, A. and Tanimura, T. (2019). Softness sensing and learning in Drosophila larvae. J Exp Biol. PubMed ID: 30833462

    Brockmann, A., Basu, P., Shakeel, M., Murata, S., Murashima, N., Boyapati, R. K., Prabhu, N. G., Herman, J. J. and Tanimura, T. (2018). Sugar intake elicits intelligent searching behavior in flies and honey bees. Front Behav Neurosci 12: 280. PubMed ID: 30546299

    Uchizono, S., Itoh, T. Q., Kim, H., Hamada, N., Kwon, J. Y. and Tanimura, T. (2017). Deciphering the genes for taste receptors for fructose in Drosophila. Mol Cells 40(10): 731-736. PubMed ID: 29047261

    Murata, S., Brockmann, A. and Tanimura, T. (2017). Pharyngeal stimulation with sugar triggers local searching behavior in Drosophila. J Exp Biol [Epub ahead of print]. PubMed ID: 28684466

    Seki, Y. and Tanimura, T. (2014). Ultradian rhythm unmasked in the Pdf clock mutant of Drosophila. J Biosci 39: 585-594. PubMed ID: 25116613

    Toshima, N., Hara, C., Scholz, C. J. and Tanimura, T. (2014). Genetic variation in food choice behaviour of amino acid-deprived Drosophila. J Insect Physiol. PubMed ID: 25010547

    Itoh, T. Q., Matsumoto, A. and Tanimura, T. (2013). C-Terminal Binding Protein (CtBP) Activates the Expression of E-Box Clock Genes with CLOCK/CYCLE in Drosophila. PLoS One 8: e63113. PubMed ID: 23646183

    Gruber, F., Knapek, S., Fujita, M., Matsuo, K., Bracker, L., Shinzato, N., Siwanowicz, I., Tanimura, T. and Tanimoto, H. (2013). Suppression of conditioned odor approach by feeding is independent of taste and nutritional value in Drosophila. Curr Biol 23: 507-514. PubMed ID: 23477724

    Matsuda, T., Kanki, T., Tanimura, T., Kang, D. and Matsuura, E. T. (2013). Effects of overexpression of mitochondrial transcription factor A on lifespan and oxidative stress response in Drosophila melanogaster. Biochem Biophys Res Commun 430: 717-721. PubMed ID: 23206694

    Toshima, N. and Tanimura, T. (2012). Taste preference for amino acids is dependent on internal nutritional state in Drosophila melanogaster. J Exp Biol 215: 2827-2832. PubMed ID: 22837455

  • Mark Tanouye Department of Environmental Science, Policy and Management, University of California, Berkeley
    Saras, A., Wu, V. V., Brawer, H. J. and Tanouye, M. A. (2017). Investigation of seizure-susceptibility in a Drosophila model of human epilepsy with optogenetic stimulation. Genetics. PubMed ID: 28630111

    Saras, A. and Tanouye, M. A. (2016). Seizure suppression by high Temperature via cAMP modulation in Drosophila. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27558668

    Saras, A. and Tanouye, M. A. (2016). Mutations of the calcium channel gene cacophony suppress seizures in Drosophila. PLoS Genet 12: e1005784. PubMed ID: 26771829

    Kroll, J.R., Wong, K.G., Siddiqui, F.M. and Tanouye, M.A. (2015). Disruption of endocytosis with the dynamin mutant shibirets1 suppresses seizures in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 26341658

    Kroll, J. R., Saras, A. and Tanouye, M. A. (2015). Drosophila sodium channel mutations: Contributions to seizure-susceptibility. Exp Neurol. PubMed ID: 26093037

    Howlett, I. C. and Tanouye, M. A. (2013). Seizure-Sensitivity in Drosophila Is Ameliorated by Dorsal Vessel Injection of the Antiepileptic Drug Valproate. J Neurogenet. PubMed ID: 23941042

    Howlett, I. C., Rusan, Z. M., Parker, L. and Tanouye, M. A. (2013). Drosophila as a Model for Intractable Epilepsy: gilgamesh Suppresses Seizures in parabss1 Heterozygote Flies. G3 (Bethesda). PubMed ID: 23797108

    Kroll, J. R., Tanouye, M. A. (2013) Rescue of easily shocked mutant seizure-sensitivity in Drosophila adults. J Comp Neurol. PubMed ID: 23682034

    Parker, L., Howlett, I. C., Rusan, Z. M. and Tanouye, M. A. (2011). Seizure and epilepsy: studies of seizure disorders in Drosophila. Int Rev Neurobiol 99: 1-21. PubMed ID: 21906534

    Parker, L., Padilla, M., Du, Y., Dong, K. and Tanouye, M. A. (2011). Drosophila as a model for epilepsy: bss is a gain-of-function mutation in the para sodium channel gene that leads to seizures. Genetics 187: 523-534. PubMed ID: 21115970

  • Madhu Gwaldas Tapadia Banaras Hindu University
    Ojha, S. and Tapadia, M. G. (2021). Non-apoptotic role of Caspase-3 in regulating Rho1GTPase mediated morphogenesis of epithelial tubes of Drosophila renal system. Dev Dyn. PubMed ID: 34773432

    Ojha, S. and Tapadia, M. G. (2020). Transcriptome profiling identifies multistep regulation through E93, Forkhead and Ecdysone Oxidase in survival of Malpighian tubules during metamorphosis in Drosophila. Int J Dev Biol 64(4-5-6): 341-351. PubMed ID: 32658993

    Dubey, S.K. and Tapadia, M.G. (2017). Yorkie regulates neurodegeneration through canonical pathway and innate immune response. Mol Neurobiol [Epub ahead of print]. PubMed ID: 28102471

    Yadav, S. and Tapadia, M.G. (2015). Expression of polyQ aggregates in Malpighian tubules leads to degeneration in Drosophila melanogaster. Dev Biol [Epub ahead of print]. PubMed ID: 26517966

    Verma, P. and Tapadia, M. G. (2015). Early gene Broad complex plays a key role in regulating the immune response triggered by ecdysone in the Malpighian tubules of Drosophila melanogaster. Mol Immunol 66: 325-339. PubMed ID: 25931442

    Gautam, N. K., Verma, P. and Tapadia, M. G. (2014). Ecdysone regulates morphogenesis and function of malpighian tubules in Drosophila melanogaster through EcR-B2 isoform. Dev Biol. PubMed ID: 25476260

    Verma, P. and Tapadia, M. G. (2013). Epithelial Immune Response in Drosophila Malpighian Tubules: Interplay between Diap2 and Ion Channels. J Cell Physiol. PubMed ID: 24374974

    Yadav, S. and Tapadia, M. G. (2013). Neurodegeneration caused by polyglutamine expansion is regulated by P-glycoprotein in Drosophila melanogaster. Genetics 195: 857-870. PubMed ID: 24037265

    Verma, P. and Tapadia, M. G. (2012). Immune response and anti-microbial peptides expression in Malpighian tubules of Drosophila melanogaster is under developmental regulation. PLoS One 7: e40714. PubMed ID: 22808242

  • Nic Tapon Francis Crick Institute, London
    Chan, E. H. Y., Zhou, Y., Aerne, B. L., Holder, M. V., Weston, A., Barry, D. J., Collinson, L. and Tapon, N. (2021). RASSF8-mediated transport of Echinoid via the exocyst promotes Drosophila wing elongation and epithelial ordering. Development 148(20). PubMed ID: 34532737

    Fulford, A. D., Holder, M. V., Frith, D., Snijders, A. P., Tapon, N. and Ribeiro, P. S. (2019). Casein kinase 1 family proteins promote Slimb-dependent Expanded degradation. Elife 8. PubMed ID: 31567070

    Bertran, M. T., Mouilleron, S., Zhou, Y., Bajaj, R., Uliana, F., Kumar, G. S., van Drogen, A., Lee, R., Banerjee, J. J., Hauri, S., O'Reilly, N., Gstaiger, M., Page, R., Peti, W. and Tapon, N. (2019). ASPP proteins discriminate between PP1 catalytic subunits through their SH3 domain and the PP1 C-tail. Nat Commun 10(1): 771. PubMed ID: 30770806

    Kulaberoglu, Y., Lin, K., Holder, M., Gai, Z., Gomez, M., Assefa Shifa, B., Mavis, M., Hoa, L., Sharif, A. A. D., Lujan, C., Smith, E. S. J., Bjedov, I., Tapon, N., Wu, G. and Hergovich, A. (2017). Stable MOB1 interaction with Hippo/MST is not essential for development and tissue growth control. Nat Commun 8(1): 695. PubMed ID: 28947795

    Banerjee, J. J., Aerne, B. L., Holder, M. V., Hauri, S., Gstaiger, M. and Tapon, N. (2017). Meru couples planar cell polarity with apical-basal polarity during asymmetric cell division. Elife 6. PubMed ID: 28665270

    Aerne, B. L., Gailite, I., Sims, D. and Tapon, N. (2015). Hippo stabilises its adaptor Salvador by antagonising the HECT ubiquitin ligase Herc4. PLoS One 10: e0131113. PubMed ID: 26125558

    Heller, D., Hoppe, A., Restrepo, S., Gatti, L., Tournier, A. L., Tapon, N., Basler, K. and Mao, Y. (2016). EpiTools: An open-source image analysis toolkit for quantifying epithelial growth dynamics. Dev Cell 36: 103-116. PubMed ID: 26766446

    Keder, A., Rives-Quinto, N., Aerne, B. L., Franco, M., Tapon, N. and Carmena, A. (2015). The Hippo Pathway Core Cassette Regulates Asymmetric Cell Division. Curr Biol 25: 2739-2750. PubMed ID: 26592338

    Gailite, I., Aerne, B. L. and Tapon, N. (2015). Differential control of Yorkie activity by LKB1/AMPK and the Hippo/Warts cascade in the central nervous system. Proc Natl Acad Sci U S A. [Epub ahead of print] PubMed ID: 26324895

    Di Cara, F., Maile, T. M., Parsons, B. D., Magico, A., Basu, S., Tapon, N. and King-Jones, K. (2015). The Hippo pathway promotes cell survival in response to chemical stress. Cell Death Differ [Epub ahead of print]. PubMed ID: 26021298

  • Aaron Tarone Department of Entomology, Texas A & M University, College Station
    Ellis, L. L., Huang, W., Quinn, A. M., Ahuja, A., Alfrejd, B., Gomez, F. E., Hjelmen, C. E., Moore, K. L., Mackay, T. F., Johnston, J. S. and Tarone, A. M. (2014). Intrapopulation Genome Size Variation in D. melanogaster Reflects Life History Variation and Plasticity. PLoS Genet 10: e1004522. PubMed ID: 25057905

    Huang, W., et al. (2014). Natural variation in genome architecture among 205 Drosophila melanogaster Genetic Reference Panel lines. Genome Res 24: 1193-1208. PubMed ID: 24714809

    Scott, M. J., Pimsler, M. L. and Tarone, A. M. (2014). Sex determination mechanisms in the Calliphoridae (blow flies). Sex Dev 8: 29-37. PubMed ID: 24401179

  • Marc Tatar Ecology & Evolutionary Biology, Brown University
    Yamamoto, R., Palmer, M., Koski, H., Curtis-Joseph, N. and Tatar, M. (2021). Aging modulated by the Drosophila insulin receptor through distinct structure-defined mechanisms. Genetics 217(2). PubMed ID: 33724413

    Tatar, M. (2021). Aging Regulated Through a Stability Model of Insulin/Insulin Growth Factor Receptor Function. Front Endocrinol (Lausanne) 12: 649880. PubMed ID: 33776941

    Zheng, W., Ocorr, K. and Tatar, M. (2020). Extra-cellular matrix induced by steroids and aging through a G-protein coupled receptor in a Drosophila model of renal fibrosis. Dis Model Mech. PubMed ID: 32461236

    Post, S., Liao, S., Yamamoto, R., Veenstra, J. A., Nassel, D. R. and Tatar, M. (2018). Drosophila insulin-like peptide dilp1 increases lifespan and glucagon-like Akh expression epistatic to dilp2. Aging Cell: e12863. PubMed ID: 30511458

    Zheng, W., Rus, F., Hernandez, A., Kang, P., Goldman, W., Silverman, N. and Tatar, M. (2018). Dehydration triggers ecdysone-mediated recognition-protein priming and elevated anti-bacterial immune responses in Drosophila Malpighian tubule renal cells. BMC Biol 16(1): 60. PubMed ID: 29855367

    Post, S., Karashchuk, G., Wade, J. D., Sajid, W., De Meyts, P. and Tatar, M. (2018). Drosophila Insulin-Like Peptides DILP2 and DILP5 Differentially Stimulate Cell Signaling and Glycogen Phosphorylase to Regulate Longevity. Front Endocrinol (Lausanne) 9: 245. PubMed ID: 29892262

    Zheng, W., Rus, F., Hernandez, A., Kang, P., Goldman, W., Silverman, N. and Tatar, M. (2018). Dehydration triggers ecdysone-mediated recognition-protein priming and elevated anti-bacterial immune responses in Drosophila Malpighian tubule renal cells. BMC Biol 16(1): 60. Pubmed ID: 29855367

    Post, S. and Tatar, M. (2016). Nutritional geometric profiles of insulin/IGF expression in Drosophila melanogaster. PLoS One 11: e0155628. PubMed ID: 27171400

    Bai, H., Post, S. Kang, P. and Tatar, M. (2015). Drosophila longevity assurance conferred by reduced insulin receptor substrate Chico partially requires d4eBP. PLoS One 10: e0134415. PubMed ID: 26252766

    Bai, H., Kang, P., Hernandez, A. M. and Tatar, M. (2013). Activin Signaling Targeted by Insulin/dFOXO Regulates Aging and Muscle Proteostasis in Drosophila. PLoS Genet 9: e1003941. PubMed ID: 24244197

  • Eran Tauber Department of Genetics, University of Leichester
    Pegoraro, M., Flavell, L. M. M., Menegazzi, P., Colombi, P., Dao, P., Helfrich-Forster, C. and Tauber, E. (2020). The genetic basis of diurnal preference in Drosophila melanogaster. BMC Genomics 21(1): 596. PubMed ID: 32862827

    Noreen, S., Pegoraro, M., Nouroz, F., Tauber, E. and Kyriacou, C. P. (2018). Interspecific studies of circadian genes period and timeless in Drosophila. Gene [Epub ahead of print]. PubMed ID: 29353056

    Zonato, V., Vanin, S., Costa, R., Tauber, E. and Kyriacou, C. P. (2017). Inverse European Latitudinal Cline at the timeless Locus of Drosophila melanogaster Reveals Selection on a Clock Gene: Population Genetics of ls-tim. J Biol Rhythms: 748730417742309. PubMed ID: 29183263

    Adewoye, A. B., Nuzhdin, S. V. and Tauber, E. (2017). Mapping quantitative trait loci underlying circadian light sensitivity in Drosophila. J Biol Rhythms 32(5): 394-405. PubMed ID: 28990443

    Pegoraro, M., Zonato, V., Tyler, E. R., Fedele, G., Kyriacou, C. P. and Tauber, E. (2017). Geographical analysis of diapause inducibility in European Drosophila melanogaster populations. J Insect Physiol 98: 238-244. PubMed ID: 28131702

    Khericha, M., Kolenchery, J. B. and Tauber, E. (2016). Neural and non-neural contributions to sexual dimorphism of mid-day sleep in Drosophila melanogaster: a pilot study. Physiol Entomol 41: 327-334. PubMed ID: 27840547

    Adewoye, A. B., Kyriacou, C. P. and Tauber, E. (2015). Identification and functional analysis of early gene expression induced by circadian light-resetting in Drosophila. BMC Genomics 16: 570. PubMed ID: 26231660

    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

    Pegoraro, M., Noreen, S., Bhutani, S., Tsolou, A., Schmid, R., Kyriacou, C. P. and Tauber, E. (2014). Molecular evolution of a pervasive natural amino-acid substitution in Drosophila cryptochrome. PLoS One 9: e86483. PubMed ID: 24475129

  • Diethard Tautz Department Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, Plön, Germany
    Zhang, W., Reeves, G. R. and Tautz, D. (2021). Testing Implications of the Omnigenic Model for the Genetic Analysis of Loci Identified through Genome-wide Association. Curr Biol. PubMed ID: 33417882

    Zhang, W., Reeves, R. G. and Tautz, D. (2019). Identification of a genetic network for an ecologically relevant behavioral phenotype in Drosophila melanogaster. Mol Ecol. PubMed ID: 31867742

    Reeves, R. G. and Tautz, D. (2017). Automated Phenotyping Indicates Pupal Size in Drosophila Is a Highly Heritable Trait with an Apparent Polygenic Basis. G3 (Bethesda) 7(4): 1277-1286. PubMed ID: 28258111

    Domazet-Loso, T., Carvunis, A. R., Alba, M. M., Sestak, M. S., Bakaric, R., Neme, R. and Tautz, D. (2017). No Evidence for Phylostratigraphic Bias Impacting Inferences on Patterns of Gene Emergence and Evolution. Mol Biol Evol 34(4): 843-856. PubMed ID: 28087778

    Aranda, M., Marques-Souza, H., Bayer, T. and Tautz, D. (2008). The role of the segmentation gene hairy in Tribolium. Dev Genes Evol 218(9): 465-477. PubMed ID: 18679713

  • Gaia Tavosanis German Center for Neurodegenerative Diseases, Bonn
    Richard, M., Doubkova, K., Nitta, Y., Kawai, H., Sugie, A. and Tavosanis, G. (2022). A Quantitative Model of Sporadic Axonal Degeneration in the Drosophila Visual System. J Neurosci 42(24): 4937-4952. PubMed ID: 35534228

    Prisco, L., Deimel, S. H., Yeliseyeva, H., Fiala, A. and Tavosanis, G. (2021). The anterior paired lateral neuron normalizes odour-evoked activity in the Drosophila mushroom body calyx. Elife 10. PubMed ID: 34964714

    Baltruschat, L., Prisco, L., Ranft, P., Lauritzen, J. S., Fiala, A., Bock, D. D. and Tavosanis, G. (2021). Circuit reorganization in the Drosophila mushroom body calyx accompanies memory consolidation. Cell Rep 34(11): 108871. PubMed ID: 33730583

    Ferreira Castro, A., Baltruschat, L., Sturner, T., Bahrami, A., Jedlicka, P., Tavosanis, G. and Cuntz, H. (2020). Achieving functional neuronal dendrite structure through sequential stochastic growth and retraction. Elife 9. PubMed ID: 33241995

    Marchetti, G. and Tavosanis, G. (2019). Modulators of hormonal response regulate temporal fate specification in the Drosophila brain. PLoS Genet 15(12): e1008491. PubMed ID: 31809495

    Ziegler, A. B., Thiele, C., Tenedini, F., Richard, M., Leyendecker, P., Hoermann, A., Soba, P. and Tavosanis, G. (2017). Cell-autonomous control of neuronal dendrite expansion via the fatty acid synthesis regulator SREBP. Cell Rep 21(12): 3346-3353. PubMed ID: 29262315

    Marchetti, G. and Tavosanis, G. (2017). Steroid Hormone Ecdysone Signaling Specifies Mushroom Body Neuron Sequential Fate via Chinmo. Curr Biol 27(19): 3017-3024.e3014. PubMed ID: 28966087

    Sugie, A., Mohl, C., Hakeda-Suzuki, S., Matsui, H., Suzuki, T. and Tavosanis, G. (2017). Analyzing Synaptic Modulation of Drosophila melanogaster Photoreceptors after Exposure to Prolonged Light. J Vis Exp(120) [Epub ahead of print]. PubMed ID: 28287587

    Sugie, A., Hakeda-Suzuki, S., Suzuki, E., Silies, M., Shimozono, M., Mohl, C., Suzuki, T. and Tavosanis, G. (2015). Molecular remodeling of the presynaptic active zone of Drosophila photoreceptors via activity-dependent feedback [Epub ahead of print]. Neuron. PubMed ID: 25892303

    Nagel, J., et al. (2012). Fascin controls neuronal class-specific dendrite arbor morphology. Development 139(16): 2999-3009. PubMed Citation: 22764047

  • J. Paul Taylor St. Jude Children's Research Hospital, Memphis
    Lee, K. H., et al. (2016). C9orf72 dipeptide repeats impair the assembly, dynamics, and function of membrane-less organelles. Cell 167: 774-788 e717. PubMed ID: 27768896

    Li, S., Freibaum, B. D., Zhang, P., Kim, N. C., Kolaitis, R. M., Molliex, A., Kanagaraj, A. P., Yabe, I., Tanino, M., Tanaka, S., Sasaki, H., Ross, E. D., Taylor, J. P. and Kim, H. J. (2016). Genetic Interaction of hnRNPA2B1 and DNAJB6 in a Drosophila Model of Multisystem Proteinopathy. Hum Mol Genet. PubMed ID: 26744327

    Freibaum, B.D., Lu, Y., Lopez-Gonzalez, R., Kim, N.C., Almeida, S., Lee, K.H., Badders, N., Valentine, M., Miller, B.L., Wong, P.C., Petrucelli, L., Kim, H.J., Gao, F.B. and Taylor, J.P. (2015). GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature [Epub ahead of print]. PubMed ID: 26308899

    He, F., Krans, A., Freibaum, B. D., Taylor, J. P. and Todd, P. K. (2014). TDP-43 suppresses CGG repeat-induced neurotoxicity through interactions with HnRNP A2/B1. Hum Mol Genet. PubMed ID: 24920338

    Alami, N. H., Smith, R. B., Carrasco, M. A., Williams, L. A., Winborn, C. S., Han, S. S., Kiskinis, E., Winborn, B., Freibaum, B. D., Kanagaraj, A., Clare, A. J., Badders, N. M., Bilican, B., Chaum, E., Chandran, S., Shaw, C. E., Eggan, K. C., Maniatis, T. and Taylor, J. P. (2014). Axonal transport of TDP-43 mRNA granules is impaired by ALS-causing mutations. Neuron 81: 536-543. PubMed ID: 24507191

    Kim, N. C., Tresse, E., Kolaitis, R. M., Molliex, A., Thomas, R. E., Alami, N. H., Wang, B., Joshi, A., Smith, R. B., Ritson, G. P., Winborn, B. J., Moore, J., Lee, J. Y., Yao, T. P., Pallanck, L., Kundu M, Taylor JP. (2013) VCP Is Essential for Mitochondrial Quality Control by PINK1/Parkin and this Function Is Impaired by VCP Mutations. Neuron. PubMed ID: 23498974

    Smith, R. and Taylor, J. P. (2011). Dissection and imaging of active zones in the Drosophila neuromuscular junction. J Vis Exp. PubMed ID: 21559003

    Lanson, N. A., Jr., Maltare, A., King, H., Smith, R., Kim, J. H., Taylor, J. P., Lloyd, T. E. and Pandey, U. B. (2011). A Drosophila model of FUS-related neurodegeneration reveals genetic interaction between FUS and TDP-43. Hum Mol Genet 20: 2510-2523. PubMed ID: 21487023

    Todd, P. K., Oh, S. Y., Krans, A., Pandey, U. B., Di Prospero, N. A., Min, K. T., Taylor, J. P. and Paulson, H. L. (2010). Histone deacetylases suppress CGG repeat-induced neurodegeneration via transcriptional silencing in models of fragile X tremor ataxia syndrome. PLoS Genet 6: e1001240. PubMed ID: 21170301

  • Guy Tear School of Biomedical Sciences, King's College, London
    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

    Povellato, G., Tuxworth, R. I., Hanger, D. P. and Tear, G. (2014). Modification of the Drosophila model of in vivo Tau toxicity reveals protective phosphorylation by GSK3beta. Biol Open 3: 1-11. PubMed ID: 24429107

    van den Brink, D. M., Banerji, O., Tear, G. (2013) Commissureless Regulation of Axon Outgrowth across the Midline Is Independent of Rab Function. PLoS One 8: e64427. PubMed ID: 23696892

  • Gerhard Technau Institut fur Genetik, University of Mainz
    Pukhlyakova, E., Aman, A. J., Elsayad, K. and Technau, U. (2018). beta-Catenin-dependent mechanotransduction dates back to the common ancestor of Cnidaria and Bilateria. Proc Natl Acad Sci U S A. Pubmed ID: 29784822

    Rickert, C., Luer, K., Vef, O. and Technau, G. M. (2018). Progressive derivation of serially homologous neuroblast lineages in the gnathal CNS of Drosophila. PLoS One 13(2): e0191453. PubMed ID: 29415052

    Hessinger, C., Technau, G.M. and Rogulja-Ortmann, A. (2016). The Drosophila Hox gene Ultrabithorax acts both in muscles and motoneurons to orchestrate formation of specific neuromuscular connections. Development [Epub ahead of print]. PubMed ID: 27913640

    Urbach, R., Jussen, D. and Technau, G.M. (2016). Gene expression profiles uncover individual identities of gnathal neuroblasts and serial homologies in the embryonic CNS of Drosophila. Development 143: 1290-1301. PubMed ID: 27095493

    Becker, H., Renner, S., Technau, G.M. and Berger, C. (2016). Cell-autonomous and non-cell-autonomous function of Hox genes specify segmental neuroblast identity in the gnathal region of the embryonic CNS in Drosophila. PLoS Genet 12: e1005961. PubMed ID: 27015425

    Birkholz, O., Rickert, C., Nowak, J., Coban, I.C. and Technau, G.M. (2015). Bridging the gap between postembryonic cell lineages and identified embryonic neuroblasts in the ventral nerve cord of Drosophila melanogaster. Biol Open [Epub ahead of print]. PubMed ID: 25819843

    Technau, G. M., Rogulja-Ortmann, A., Berger, C., Birkholz, O. and Rickert, C. (2014). Composition of a neuromere and its segmental diversification under the control of Hox genes in the embryonic CNS of Drosophila. J Neurogenet: 1-23. PubMed ID: 24913688

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

    Birkholz, O., Vef, O., Rogulja-Ortmann, A., Berger, C. and Technau, G. M. (2013). Abdominal-B and caudal inhibit the formation of specific neuroblasts in the Drosophila tail region. Development. PubMed ID: 23903193

    Birkholz, O., Rickert, C., Berger, C., Urbach, R., Technau, G. M. (2013) Neuroblast pattern and identity in the Drosophila tail region and role of doublesex in the survival of sex-specific precursors. Development 140: 1830-1842. PubMed ID: 23533181

  • Luis Teixeira Instituto Gulbenkian de Ciencia, Oeiras, Portugal
    Chrostek, E., Martins, N., Marialva, M. S. and Teixeira, L. (2021). Wolbachia-Conferred Antiviral Protection Is Determined by Developmental Temperature. mBio: e0292320. PubMed ID: 34488458

    Pais, I. S., Valente, R. S., Sporniak, M. and Teixeira, L. (2018). Drosophila melanogaster establishes a species-specific mutualistic interaction with stable gut-colonizing bacteria. PLoS Biol 16(7): e2005710. PubMed ID: 29975680

    Chrostek, E., Martins, N., Marialva, M. S. and Teixeira, L. (2021). Wolbachia-Conferred Antiviral Protection Is Determined by Developmental Temperature. mBio: e0292320. PubMed ID: 34488458

    Faria, V.G., Martins, N.E., Magalhães, S., Paulo, T.F., Nolte, V., Schlötterer, C., Sucena, É and Teixeira, L. (2016). Drosophila adaptation to viral infection through defensive symbiont evolution. PLoS Genet 12: e1006297. PubMed ID: 27684942

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

    Chrostek, E. and Teixeira, L. (2015). Mutualism breakdown by amplification of wolbachia genes. PLoS Biol 13: e1002065. PubMed ID: 25668031

    Ferreira, A. G., Naylor, H., Esteves, S. S., Pais, I. S., Martins, N. E. and Teixeira, L. (2014). The toll-dorsal pathway is required for resistance to viral oral infection in Drosophila. PLoS Pathog 10: e1004507. PubMed ID: 25473839

    Atallah, J., Teixeira, L., Salazar, R., Zaragoza, G. and Kopp, A. (2014). The making of a pest: the evolution of a fruit-penetrating ovipositor in Drosophila suzukii and related species. Proc Biol Sci 281: 20132840. PubMed ID: 24573846

    Chrostek, E., Marialva, M. S., Esteves, S. S., Weinert, L. A., Martinez, J., Jiggins, F. M. and Teixeira, L. (2013). Wolbachia variants induce differential protection to viruses in Drosophila melanogaster: a phenotypic and phylogenomic analysis. PLoS Genet 9: e1003896. PubMed ID: 24348259

    Teixeira, L. (2012). Whole-genome expression profile analysis of Drosophila melanogaster immune responses. Brief Funct Genomics 11: 375-386. PubMed ID: 23023664

  • Francisco Tejedor Instituto de Neurociencias, CSIC-UMH, Alicante, Spain
    Shaikh, M. N. and Tejedor, F. J. (2018). Mnb/Dyrk1A orchestrates a transcriptional network at the transition from self-renewing neurogenic progenitors to postmitotic neuronal precursors. J Neurogenet 32(1): 37-50. PubMed ID: 29495936

    Shaikh, M. N., Gutierrez-Avino, F., Colonques, J., Ceron, J., Hammerle, B. and Tejedor, F. J. (2016). Minibrain drives the Dacapo dependent cell cycle exit of neurons in the Drosophila brain by promoting asense and prospero expression. Development. PubMed ID: 27510975

    Hammerle, B., Ulin, E., Guimera, J., Becker, W., Guillemot, F. and Tejedor, F. J. (2011). Transient expression of Mnb/Dyrk1a couples cell cycle exit and differentiation of neuronal precursors by inducing p27KIP1 expression and suppressing NOTCH signaling. Development 138: 2543-2554. PubMed ID: 21610031

    Colonques, J., Ceron, J., Reichert, H. and Tejedor, F. J. (2011). A transient expression of Prospero promotes cell cycle exit of Drosophila postembryonic neurons through the regulation of Dacapo. PLoS One 6: e19342. PubMed ID: 21552484

  • Aurelio Teleman National Center for Tumor Diseases, Heidelberg
    Strassburger, K., Lutz, M., Muller, S. and Teleman, A. A. (2021). Ecdysone regulates Drosophila wing disc size via a TORC1 dependent mechanism. Nat Commun 12(1): 6684. PubMed ID: 34795214
    <>Bageritz, J., Willnow, P., Valentini, E., Leible, S., Boutros, M. and Teleman, A. A. (2019). Gene expression atlas of a developing tissue by single cell expression correlation analysis. Nat Methods 16(8): 750-756. PubMed ID: 31363221

    Strassburger, K., Kang, E. and Teleman, A. A. (2019). Drosophila ZDHHC8 palmitoylates scribble and Ras64B and controls growth and viability. PLoS One 14(2): e0198149. PubMed ID: 30735487

    Kwiatkowski, S., Seliga, A. K., Vertommen, D., Terreri, M., Ishikawa, T., Grabowska, I., Tiebe, M., Teleman, A. A., Jagielski, A. K., Veiga-da-Cunha, M. and Drozak, J. (2018). SETD3 protein is the actin-specific histidine N-methyltransferase. Elife 7. PubMed ID: 30526847

    Tiebe, M., Lutz, M., Levy, D. and Teleman, A. A. (2018). Phenotypic characterization of SETD3 knockout Drosophila. PLoS One 13(8): e0201609. PubMed ID: 30067821

    Moraru, A., Wiederstein, J., Pfaff, D., Fleming, T., Miller, A. K., Nawroth, P. and Teleman, A. A. (2018). Elevated Levels of the Reactive Metabolite Methylglyoxal Recapitulate Progression of Type 2 Diabetes. Cell Metab. PubMed ID: 29551588

    Drager, N. M., Nachman, E., Winterhoff, M., Bruhmann, S., Shah, P., Katsinelos, T., Boulant, S., Teleman, A. A., Faix, J. and Jahn, T. R. (2017). Bin1 directly remodels actin dynamics through its BAR domain. EMBO Rep. PubMed ID: 28893863

    Strassburger, K., Lorbeer, F. K., Lutz, M., Graf, F., Boutros, M. and Teleman, A. A. (2017). Oxygenation and adenosine deaminase support growth and proliferation of ex vivo cultured Drosophila wing imaginal discs. Development. PubMed ID: 28526754

    Moraru, A., Cakan-Akdogan, G., Strassburger, K., Males, M., Mueller, S., Jabs, M., Muelleder, M., Frejno, M., Braeckman, B. P., Ralser, M. and Teleman, A. A. (2017). THADA regulates the organismal balance between energy storage and heat production. Dev Cell 41(1): 72-81.e76. PubMed ID: 28399403

    Tsokanos, F. F., Albert, M. A., Demetriades, C., Spirohn, K., Boutros, M. and Teleman, A. A. (2016). eIF4A inactivates TORC1 in response to amino acid starvation. EMBO J [Epub ahead of print]. PubMed ID: 26988032

    Senyilmaz, D., Virtue, S., Xu, X., Tan, C.Y., Griffin, J.L., Miller, A.K., Vidal-Puig, A. and Teleman, A.A. (2015). Regulation of mitochondrial morphology and function by stearoylation of TFR1. Nature [Epub ahead of print]. PubMed ID: 26214738

    Tiebe, M., Lutz, M., De La Garza, A., Buechling, T., Boutros, M. and Teleman, A. A. (2015). REPTOR and REPTOR-BP regulate organismal metabolism and transcription downstream of TORC1. Dev Cell 33: 272-284. PubMed ID: 25920570

  • Kelly Ten Hagen Developmental Glycobiology Section, NIDCR, NIH, Bethesda
    May, C., Ji, S., Syed, Z. A., Revoredo, L., Daniel, E. J. P., Gerken, T. A., Tabak, L. A., Samara, N. L. and Ten Hagen, K. G. (2020). Differential splicing of the lectin domain of an O-glycosyltransferase modulates both peptide and glycopeptide preferences. J Biol Chem. PubMed ID: 32669364

    Reynolds, H. M., Zhang, L., Tran, D. T. and Ten Hagen, K. G. (2019). Tango1 coordinates the formation of ER/Golgi docking sites to mediate secretory granule formation. J Biol Chem. PubMed ID: 31690624

    Ji, S., Samara, N. L., Revoredo, L., Zhang, L., Tran, D. T., Muirhead, K., Tabak, L. A. and Ten Hagen, K. G. (2018). A molecular switch orchestrates enzyme specificity and secretory granule morphology. Nat Commun 9(1): 3508. PubMed ID: 30158631

    Zhang, L., Ribbeck, K., Turner, B. and Ten Hagen, K. G. (2017). Loss of the mucosal barrier alters the progenitor cell niche via JAK/STAT signaling. J Biol Chem 292(52):21231-21242. PubMed ID: 29127201

    Tran, D. T., Masedunskas, A., Weigert, R. and Ten Hagen, K. G. (2015). Arp2/3-mediated F-actin formation controls regulated exocytosis in vivo. Nat Commun 6: 10098. PubMed ID: 26639106

    Zhang, L., Syed, Z. A., van Dijk Hard, I., Lim, J. M., Wells, L. and Ten Hagen, K. G. (2014). O-Glycosylation regulates polarized secretion by modulating Tango1 stability. Proc Natl Acad Sci U S A. PubMed ID: 24799692

    Tian, E., Zhang, L. and Ten Hagen, K. G. (2013). Fluorescent lectin staining of Drosophila embryos and tissues to detect the spatial distribution of glycans during development. Methods Mol Biol 1022: 99-105. PubMed ID: 23765656

    Tran, D. T., Zhang, L., Zhang, Y., Tian, E., Earl, L. A. and Ten Hagen, K. G. (2012). Multiple members of the UDP-GalNAc: polypeptide N-acetylgalactosaminyltransferase family are essential for viability in Drosophila. J Biol Chem 287: 5243-5252. PubMed ID: 22157008

    Zhang, L. and Ten Hagen, K. G. (2010). Dissecting the biological role of mucin-type O-glycosylation using RNA interference in Drosophila cell culture. J Biol Chem 285: 34477-34484. PubMed ID: 20807760

    Sun, Q., Tian, E., Turner, R. J. and Ten Hagen, K. G. (2010). Developmental and functional studies of the SLC12 gene family members from Drosophila melanogaster. Am J Physiol Cell Physiol 298: C26-37. PubMed ID: 19828839

  • Jason Tennessen Department of Biology, Indiana University, Bloomington
    Li, H., Rai, M., Buddika, K., Sterrett, M. C., Luhur, A., Mahmoudzadeh, N. H., Julick, C. R., Pletcher, R. C., Chawla, G., Gosney, C. J., Burton, A. K., Karty, J. A., Montooth, K. L., Sokol, N. S. and Tennessen, J. M. (2019). Lactate dehydrogenase and glycerol-3-phosphate dehydrogenase cooperatively regulate growth and carbohydrate metabolism during Drosophila melanogaster larval development. Development. PubMed ID: 31399469

    St Clair, S. L., Li, H., Ashraf, U., Karty, J. A. and Tennessen, J. M. (2017). Metabolomic analysis reveals that the Drosophila gene lysine influences diverse aspects of metabolism. Genetics. PubMed ID: 28986444

    Li, H., Chawla, G., Hurlburt, A. J., Sterrett, M. C., Zaslaver, O., Cox, J., Karty, J. A., Rosebrock, A. P., Caudy, A. A. and Tennessen, J. M. (2017). Drosophila larvae synthesize the putative oncometabolite L-2-hydroxyglutarate during normal developmental growth. Proc Natl Acad Sci U S A 114(6): 1353-1358. PubMed ID: 28115720

    Tennessen, J. M., Barry, W. E., Cox, J. and Thummel, C. S. (2014). Methods for studying metabolism in Drosophila. Methods 68(1): 105-115. PubMed ID: 24631891

    Tennessen, J. M., Bertagnolli, N. M., Evans, J., Sieber, M. H., Cox, J. and Thummel, C. S. (2014). Coordinated metabolic transitions during Drosophila embryogenesis and the onset of aerobic glycolysis. G3 (Bethesda) 4(5): 839-850. PubMed ID: 24622332

  • Ulrich Tepass Department of Cell and Systems Biology, University of Toronto
    Simoes, S., Lerchbaumer, G., Pellikka, M., Giannatou, P., Lam, T., Kim, D., Yu, J., Ter Stal, D., Al Kakouni, K., Fernandez-Gonzalez, R. and Tepass, U. (2022). Crumbs complex-directed apical membrane dynamics in epithelial cell ingression. J Cell Biol 221(7). PubMed ID: 35588693

    Sarpal, R., Yan, V., Kazakova, L., Sheppard, L., Yu, J. C., Fernandez-Gonzalez, R. and Tepass, U. (2019). Role of alpha-Catenin and its mechanosensing properties in regulating Hippo/YAP-dependent tissue growth. PLoS Genet 15(11): e1008454. PubMed ID: 31697683

    Laffafian, A. and Tepass, U. (2019). Identification of genes required for apical protein trafficking in Drosophila photoreceptor cells. G3 (Bethesda). PubMed ID: 31649044

    Silver, J. T., Wirtz-Peitz, F., Simoes, S., Pellikka, M., Yan, D., Binari, R., Nishimura, T., Li, Y., Harris, T. J. C., Perrimon, N. and Tepass, U. (2019). Apical polarity proteins recruit the RhoGEF Cysts to promote junctional myosin assembly. J Cell Biol. PubMed ID: 31409654

    Pellikka, M. and Tepass, U. (2017). Unique cell biological profiles of retinal disease-causing missense mutations in the polarity protein crumbs. J Cell Sci [Epub ahead of print]. PubMed ID: 28515229

    Simoes, S., Oh, Y., Wang, M. F., Fernandez-Gonzalez, R. and Tepass, U. (2017). Myosin II promotes the anisotropic loss of the apical domain during Drosophila neuroblast ingression. J Cell Biol [Epub ahead of print]. PubMed ID: 28363972

    Houssin, E., Tepass, U. and Laprise, P. (2015). Girdin-mediated interactions between cadherin and the actin cytoskeleton are required for epithelial morphogenesis in Drosophila. Development 142: 1777-1784. PubMed ID: 25968313

    Escobar, D. J., Desai, R., Ishiyama, N., Folmsbee, S. S., Novak, M. N., Flozak, A. S., Daugherty, R. L., Mo, R., Nanavati, D., Sarpal, R., Leckband, D., Ikura, M., Tepass, U. and Gottardi, C. J. (2015). alpha-catenin phosphorylation promotes intercellular adhesion through a dual-kinase mechanism. J Cell Sci [Epub ahead of print]. PubMed ID: 25653389

    Escobar, D. J., Desai, R., Ishiyama, N., Folmsbee, S. S., Novak, M. N., Flozak, A. S., Daugherty, R. L., Mo, R., Nanavati, D., Sarpal, R., Leckband, D., Ikura, M., Tepass, U. and Gottardi, C. J. (2015). alpha-catenin phosphorylation promotes intercellular adhesion through a dual-kinase mechanism. J Cell Sci. PubMed ID: 25653389

    Desai, R., Sarpal, R., Ishiyama, N., Pellikka, M., Ikura, M., Tepass, U. (2013) Monomeric alpha-catenin links cadherin to the actin cytoskeleton. Nat Cell Biol. PubMed ID: 23417122

  • John Terblanche Department of Conservation Ecology and Entomology, Stellenbosch University, Matieland, South Africa​
    Huisamen, E. J., Colinet, H., Karsten, M. and Terblanche, J. S. (2022). Dietary salt supplementation adversely affects thermal acclimation responses of flight ability in Drosophila melanogaster. J Insect Physiol 140: 104403. PubMed ID: 35667397

    Willot, Q., Loos, B. and Terblanche, J. S. (2021). Interactions between developmental and adult acclimation have distinct consequences for heat tolerance and heat stress recovery. J Exp Biol 224(16). PubMed ID: 34308995

    De Araujo, L. I., Karsten, M. and Terblanche, J. S. (2019). Exploring thermal flight responses as predictors of flight ability and geographic range size in Drosophila. Comp Biochem Physiol A Mol Integr Physiol 236: 110532. PubMed ID: 31351148

  • Jonathan Terman University of Texas Southwestern Medical Center, Dallas
    Grintsevich, E. E., Ahmed, G., Ginosyan, A. A., Wu, H., Rich, S. K., Reisler, E. and Terman, J. R. (2021). Profilin and Mical combine to impair F-actin assembly and promote disassembly and remodeling. Nat Commun 12(1): 5542. PubMed ID: 34545088

    Yoon, J., Wu, H., Hung, R. J. and Terman, J. R. (2021). Enhanced Production of the Mical Redox Domain for Enzymology and F-actin Disassembly Assays. Int J Mol Sci 22(4). PubMed ID: 33671465

    Grintsevich, E. E., Yesilyurt, H. G., Rich, S. K., Hung, R. J., Terman, J. R. and Reisler, E. (2016). F-actin dismantling through a redox-driven synergy between Mical and cofilin. Nat Cell Biol 18: 876-885. PubMed ID: 27454820

    Yang, T. and Terman, J. R. (2013). Regulating small G protein signaling to coordinate axon adhesion and repulsion. Small GTPases 4: 34-41. PubMed ID: 23247636

    Yang, T. and Terman, J. R. (2012). 14-3-3epsilon couples protein kinase A to semaphorin signaling and silences plexin RasGAP-mediated axonal repulsion. Neuron 74: 108-121. PubMed ID: 22500634

    Hung, R. J., Pak, C. W. and Terman, J. R. (2011). Direct redox regulation of F-actin assembly and disassembly by Mical. Science 334: 1710-1713. PubMed ID: 22116028

    Williamson, W. R., Yang, T., Terman, J. R. and Hiesinger, P. R. (2010). Guidance receptor degradation is required for neuronal connectivity in the Drosophila nervous system. PLoS Biol 8: e1000553. PubMed ID: 21151882

  • Christophe Terzian Ecole Pratique des Hautes Etudes, Paris
    Touret, F., Guiguen, F., Greenland, T. and Terzian, C. (2014). In between: Gypsy in Drosophila melanogaster Reveals New Insights into Endogenous Retrovirus Evolution. Viruses 6: 4914-4925. PubMed ID: 25502325

    Touret, F., Guiguen, F. and Terzian, C. (2014). Wolbachia Influences the Maternal Transmission of the gypsy Endogenous Retrovirus in Drosophila melanogaster. MBio 5. PubMed ID: 25182324

    Shaw, A. E., Veronesi, E., Maurin, G., Ftaich, N., Guiguen, F., Rixon, F., Ratinier, M., Mertens, P., Carpenter, S., Palmarini, M., Terzian, C. and Arnaud, F. (2012). Drosophila melanogaster as a model organism for bluetongue virus replication and tropism. J Virol 86: 9015-9024. PubMed ID: 22674991

    Akkouche, A., Rebollo, R., Burlet, N., Esnault, C., Martinez, S., Viginier, B., Terzian, C., Vieira, C. and Fablet, M. (2012). tirant, a newly discovered active endogenous retrovirus in Drosophila simulans. J Virol 86: 3675-3681. PubMed ID: 22278247

  • Charles Tessier Indiana University School of Medicine, South Bend
    Rudisill, S. S., Martin, B. R., Mankowski, K. M. and Tessier, C. R. (2019). Iron Deficiency Reduces Synapse Formation in the Drosophila Clock Circuit. Biol Trace Elem Res 189(1): 241-250. PubMed ID: 30022428

    Hutson, R. L., Thompson, R. L., Bantel, A. P. and Tessier, C. R. (2018). Acamprosate rescues neuronal defects in the Drosophila model of Fragile X Syndrome. Life Sci 195: 65-70. PubMed ID: 29317220

    Bantel, A. P. and Tessier, C. R. (2016). Taste Preference Assay for Adult Drosophila. J Vis Exp(115). PubMed ID: 27684591

    Hagel, K. R., Beriont, J. and Tessier, C. R. (2015). Drosophila Cbp53E Regulates Axon Growth at the Neuromuscular Junction. PLoS One 10(7): e0132636. PubMed ID: 26167908

  • Ulrich Theopold Department of Molecular Biology and Functional Genomics, Stockholm University
    Dziedziech, A. and Theopold, U. (2021). Proto-pyroptosis: An ancestral origin for mammalian inflammatory cell death mechanism in Drosophila melanogaster. J Mol Biol: 167333. PubMed ID: 34756921

    Krautz, R., Khalili, D. and Theopold, U. (2020). Tissue-autonomous immune response regulates stress signalling during hypertrophy. Elife 9. PubMed ID: 33377870

    Arefin, B., Kunc, M., Krautz, R. and Theopold, U. (2017). The immune phenotype of three Drosophila leukemia models. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 28476910

    Kučerová, L., Kubrak, O.I., Bengtsson, J.M., Strnad, H., Nylin, S., Theopold, U. and Nässel, D.R. (2016). Slowed aging during reproductive dormancy is reflected in genome-wide transcriptome changes in Drosophila melanogaster. BMC Genomics 17: 50. PubMed ID: 26758761

    Arefin, B., Kucerova, L., Krautz, R., Kranenburg, H., Parvin, F. and Theopold, U. (2015). Apoptosis in hemocytes induces a shift in effector mechanisms in the Drosophila immune system and leads to a pro-inflammatory state. PLoS One 10: e0136593. PubMed ID: 26322507

    Hauling, T., Krautz, R., Markus, R., Volkenhoff, A., Kucerova, L. and Theopold, U. (2014). A Drosophila immune response against Ras-induced overgrowth. Biol Open. PubMed ID: 24659248

    Arefin, B., Kucerova, L., Dobes, P., Markus, R., Strnad, H., Wang, Z., Hyrsl, P., Zurovec, M. and Theopold, U. (2014). Genome-wide transcriptional analysis of Drosophila larvae infected by entomopathogenic nematodes shows involvement of complement, recognition and extracellular matrix proteins. J Innate Immun 6: 192-204. PubMed ID: 23988573

    Theopold, U., Krautz, R. and Dushay, M. S. (2014). The Drosophila clotting system and its messages for mammals. Dev Comp Immunol 42: 42-46. PubMed ID: 23545286

  • Pascal Thérond Institute of Biology Valrose, Nice
    Hurbain, I., Mace, A. S., Romao, M., Prince, E., Sengmanivong, L., Ruel, L., Basto, R., Therond, P. P., Raposo, G. and D'Angelo, G. (2022). Microvilli-derived extracellular vesicles carry Hedgehog morphogenic signals for Drosophila wing imaginal disc development. Curr Biol 32(2): 361-373. PubMed ID: 34890558

    Marcetteau, J., Matusek, T., Luton, F. and Therond, P. P. (2021). Arf6 is necessary for senseless expression in response to Wingless signalling during Drosophila wing development. Biol Open. PubMed ID: 34779478

    Bonche, R., Chessel, A., Boisivon, S., Smolen, P., Therond, P. and Pizette, S. (2020). Two different sources of Perlecan cooperate for its function in the basement membrane of the Drosophila wing imaginal disc. Dev Dyn. PubMed ID: 33269518
    Rallis, A., Navarro, J. A., Rass, M., Hu, A., Birman, S., Schneuwly, S. and Therond, P. P. (2020). Hedgehog Signaling Modulates Glial Proteostasis and Lifespan. Cell Rep 30(8): 2627-2643.e2625. PubMed ID: 32101741

    Matusek, T., Therond, P. and Furthauer, M. (2019). Functional analysis of ESCRT-positive extracellular vesicles in the Drosophila wing imaginal disc. Methods Mol Biol 1998: 31-47. PubMed ID: 31250292

    Giordano, C., Ruel, L., Poux, C. and Therond, P. (2018). Protein association changes in the Hedgehog signaling complex mediate differential signaling strength. Development 145(24). PubMed ID: 30541874

    D'Angelo, G., Matusek, T., Pizette, S. and Therond, P. P. (2015). Endocytosis of Hedgehog through Dispatched regulates long-range signaling. Dev Cell [Epub ahead of print]. PubMed ID: 25619925

    Matusek, T., Wendler, F., Poles, S., Pizette, S., D'Angelo, G., Furthauer, M. and Therond, P. P. (2014). The ESCRT machinery regulates the secretion and long-range activity of Hedgehog. Nature 516: 99-103. PubMed ID: 25471885

    Ranieri, N., Therond, P. P. and Ruel, L. (2014). Switch of PKA substrates from Cubitus interruptus to Smoothened in the Hedgehog signalosome complex. Nat Commun 5: 5034. PubMed ID: 25289679

    Briscoe, J., Therond, P. P. (2013) The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol. PubMed ID: 23719536

  • Marc Therrien IRIC, Université de Montréal
    Baril, C., Gavory, G., Bidla, G., Knaevelsrud, H., Sauvageau, G. and Therrien, M. (2016). Human NUP98-HOXA9 promotes hyperplastic growth of hematopoietic tissues in Drosophila. Dev Biol. PubMed ID: 27838340

    Ashton-Beaucage, D., Udell, C. M., Gendron, P., Sahmi, M., Lefrancois, M., Baril, C., Guenier, A. S., Duchaine, J., Lamarre, D., Lemieux, S. and Therrien, M. (2014). A Functional Screen Reveals an Extensive Layer of Transcriptional and Splicing Control Underlying RAS/MAPK Signaling in Drosophila. PLoS Biol 12: e1001809. PubMed ID: 24643257

    Stochmanski, S. J., Therrien, M., Laganiere, J., Rochefort, D., Laurent, S., Karemera, L., Gaudet, R., Vyboh, K., Van Meyel, D. J., Di Cristo, G., Dion, P. A., Gaspar, C. and Rouleau, G. A. (2012). Expanded ATXN3 frameshifting events are toxic in Drosophila and mammalian neuron models. Hum Mol Genet 21: 2211-2218. PubMed ID: 22337953

    Ashton-Beaucage, D., Udell, C. M., Lavoie, H., Baril, C., Lefrancois, M., Chagnon, P., Gendron, P., Caron-Lizotte, O., Bonneil, E., Thibault, P. and Therrien, M. (2010). The exon junction complex controls the splicing of MAPK and other long intron-containing transcripts in Drosophila. Cell 143: 251-262. PubMed ID: 20946983

  • William Theurkauf Program in Molecular Medicine, University of Massachusetts Medical School, Worchester
    Zhang, G., Yu, T., Parhad, S. S., Ho, S., Weng, Z. and Theurkauf, W. E. (2021). piRNA-independent transposon silencing by the Drosophila THO complex. Dev Cell 56(18): 2623-2635.e2625. PubMed ID: 34547226

    Parhad, S. S., Yu, T., Zhang, G., Rice, N. P., Weng, Z. and Theurkauf, W. E. (2020). Adaptive Evolution Targets a piRNA Precursor Transcription Network. Cell Rep 30(8): 2672-2685.e2675. PubMed ID: 32101744

    Zhang, G., Tu, S., Yu, T., Zhang, X. O., Parhad, S. S., Weng, Z. and Theurkauf, W. E. (2018). Co-dependent assembly of Drosophila piRNA precursor complexes and piRNA cluster heterochromatin. Cell Rep 24(13): 3413-3422.e3414. PubMed ID: 30257203

    Yu, B., Lin, Y. A., Parhad, S. S., Jin, Z., Ma, J., Theurkauf, W. E., Zhang, Z. Z. and Huang, Y. (2018). Structural insights into Rhino-Deadlock complex for germline piRNA cluster specification. EMBO Rep. PubMed ID: 29858487

    Bozler, J., Kacsoh, B. Z., Chen, H., Theurkauf, W. E., Weng, Z. and Bosco, G. (2017). A systems level approach to temporal expression dynamics in Drosophila reveals clusters of long term memory genes. PLoS Genet 13(10): e1007054. PubMed ID: 29084214

    Zhang, Z., Wang, J., Schultz, N., Zhang, F., Parhad, S. S., Tu, S., Vreven, T., Zamore, P. D., Weng, Z. and Theurkauf, W. E. (2014). The HP1 Homolog Rhino Anchors a Nuclear Complex that Suppresses piRNA Precursor Splicing. Cell 157: 1353-1363. PubMed ID: 24906152

    Zhang, Z., Koppetsch, B. S., Wang, J., Tipping, C., Weng, Z., Theurkauf, W. E. and Zamore, P. D. (2014). Antisense piRNA amplification, but not piRNA production or nuage assembly, requires the Tudor-domain protein Qin. EMBO J 33: 536-539. PubMed ID: 24652836

    Simkin, A., Wong, A., Poh, Y. P., Theurkauf, W. E. and Jensen, J. D. (2013). Recurrent and recent selective sweeps in the piRNA pathway. Evolution 67: 1081-1090. PubMed ID: 23550757

    Zhang, F., Wang, J., Xu, J., Zhang, Z., Koppetsch, B. S., Schultz, N., Vreven, T., Meignin, C., Davis, I., Zamore, P. D., Weng, Z. and Theurkauf, W. E. (2012). UAP56 couples piRNA clusters to the perinuclear transposon silencing machinery. Cell 151: 871-884. PubMed ID: 23141543

  • Claire Thomas Biology and Biochemistry and Molecular Biology, University Park, PA
    Saito, M., Watanabe-Nakayama, T., Machida, S., Osada, T., Afrin, R. and Ikai, A. (2015). Spectrin-ankyrin interaction mechanics: A key force balance factor in the red blood cell membrane skeleton. Biophys Chem 200-201: 1-8. PubMed ID: 25866912

    Wu, J., Bakerink, K. J., Evangelista, M. E. and Thomas, G. H. (2014). Cytoplasmic capes are nuclear envelope intrusions that are enriched in endosomal proteins and depend upon betaH-spectrin and Annexin B9. PLoS One 9: e93680. PubMed ID: 24705398

    Khanna, M. R., Mattie, F. J., Browder, K. C., Radyk, M. D., Crilly, S. E., Bakerink, K. J., Harper, S. L., Speicher, D. W. and Thomas, G. H. (2014). Spectrin tetramer formation is not required for viable development in Drosophila. J Biol Chem. PubMed ID: 25381248

    Lin, J. I., Mitchell, N. C., Kalcina, M., Tchoubrieva, E., Stewart, M. J., Marygold, S. J., Walker, C. D., Thomas, G., Leevers, S. J., Pearson, R. B., Quinn, L. M. and Hannan, R. D. (2011). Drosophila ribosomal protein mutants control tissue growth non-autonomously via effects on the prothoracic gland and ecdysone. PLoS Genet 7: e1002408. PubMed ID: 22194697

    Tjota, M., Lee, S. K., Wu, J., Williams, J. A., Khanna, M. R. and Thomas, G. H. (2011). Annexin B9 binds to beta(H)-spectrin and is required for multivesicular body function in Drosophila. J Cell Sci 124: 2914-2926. PubMed ID: 21878499

    Lee, S. K. and Thomas, G. H. (2011). Rac1 modulation of the apical domain is negatively regulated by beta (Heavy)-spectrin. Mech Dev 128: 116-128. PubMed ID: 21111816

  • Jeffrey H. Thomas Texas Tech University Health Sciences Center, Lubbock
    Holcomb, M. C., Gao, G. J., Servati, M., Schneider, D., McNeely, P. K., Thomas, J. H. and Blawzdziewicz, J. (2021). Mechanical feedback and robustness of apical constrictions in Drosophila embryo ventral furrow formation. PLoS Comput Biol 17(7): e1009173. PubMed ID: 34228708

    Carter, T. Y., Gadwala, S., Chougule, A. B., Bui, A. P. N., Sanders, A. C., Chaerkady, R., Cormier, N., Cole, R. N. and Thomas, J. H. (2019). Actomyosin contraction during cellularization is regulated in part by Src64 control of Actin 5C protein levels. Genesis: e23297. PubMed ID: 30974046

    Chougule, A.B., Hastert, M.C. and Thomas, J.H. (2016). Drak is required for actomyosin organization during Drosophila cellularization. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26818071

    Spencer, A.K., Siddiqui, B.A. and Thomas, J.H. (2015). Cell shape change and invagination of the cephalic furrow involves reorganization of F-actin. Dev Biol [Epub ahead of print]. PubMed ID: 25929228


  • John Thomas Salk Institute
    Wang, T., Wiater, E., Zhang, X., Thomas, J. B. and Montminy, M. (2021). Crtc modulates fasting programs associated with 1-C metabolism and inhibition of insulin signaling. Proc Natl Acad Sci U S A 118(12). PubMed ID: 33723074

    Yoshikawa, S., Long, H. and Thomas, J. B. (2016). A subset of interneurons required for Drosophila larval locomotion. Mol Cell Neurosci 70: 22-29. PubMed ID: 26621406

    Long, H., Yoshikawa, S. and Thomas, J. B. (2016). Equivalent activities of repulsive axon guidance receptors. J Neurosci 36: 1140-1150. PubMed ID: 26818503

    Read, R. D., Fenton, T. R., Gomez, G. G., Wykosky, J., Vandenberg, S. R., Babic, I., Iwanami, A., Yang, H., Cavenee, W. K., Mischel, P. S., Furnari, F. B., Thomas, J. B. (2013) A Kinome-Wide RNAi Screen in Drosophila Glia Reveals That the RIO Kinases Mediate Cell Proliferation and Survival through TORC2-Akt Signaling in Glioblastoma. PLoS Genet 9: e1003253. Pubmed ID: 23459592

    Wang, B., Moya, N., Niessen, S., Hoover, H., Mihaylova, M. M., Shaw, R. J., Yates, J. R., 3rd, Fischer, W. H., Thomas, J. B. and Montminy, M. (2011). A hormone-dependent module regulating energy balance. Cell 145: 596-606. PubMed ID: 21565616

  • Barry Thompson Lincoln’s Inn Fields Laboratories, Epithelial Biology Lab, The Crick Institute
    Bonello, T., Aguilar-Aragon, M., Tournier, A. and Thompson, B. J. (2021). A picket fence function for adherens junctions in epithelial cell polarity. Cells Dev: 203719. PubMed ID: 34242843

    Diaz-de-la-Loza, M. D., Loker, R., Mann, R. S. and Thompson, B. J. (2020). Control of tissue morphogenesis by the HOX gene Ultrabithorax. Development 147(5). PubMed ID: 32122911

    Aguilar-Aragon, M., Bonello, T. T., Bell, G. P., Fletcher, G. C. and Thompson, B. J. (2020). Adherens junction remodelling during mitotic rounding of pseudostratified epithelial cells. EMBO Rep: e49700. PubMed ID: 32030856

    Aguilar-Aragon, M., Fletcher, G. and Thompson, B. J. (2019). The cytoskeletal motor proteins Dynein and MyoV direct apical transport of Crumbs. Dev Biol. PubMed ID: 31881198

    Sidor, C., Borreguero-Munoz, N., Fletcher, G. C., Elbediwy, A., Guillermin, O. and Thompson, B. J. (2019). Mask family proteins ANKHD1 and ANKRD17 regulate YAP nuclear import and stability. Elife 8. PubMed ID: 31661072

    Diaz-de-la-Loza, M. D., Ray, R. P., Ganguly, P. S., Alt, S., Davis, J. R., Hoppe, A., Tapon, N., Salbreux, G. and Thompson, B. J. (2018). Apical and masal matrix remodeling control epithelial morphogenesis. Dev Cell 46(1): 23-39.e25. PubMed ID: 29974861

    Fletcher, G. C., Diaz-de-la-Loza, M. D., Borreguero-Munoz, N., Holder, M., Aguilar-Aragon, M. and Thompson, B. J. (2018). Mechanical strain regulates the Hippo pathway in Drosophila. Development 145(5). PubMed ID: 29440303

    Aguilar-Aragon, M., Elbediwy, A., Foglizzo, V., Fletcher, G. C., Li, V. S. W. and Thompson, B. J. (2018). Pak1 kinase maintains apical membrane identity in epithelia. Cell Rep 22(7): 1639-1646. PubMed ID: 29444419

    Soriano, E. V., Ivanova, M. E., Fletcher, G., Riou, P., Knowles, P. P., Barnouin, K., Purkiss, A., Kostelecky, B., Saiu, P., Linch, M., Elbediwy, A., Kjaer, S., O'Reilly, N., Snijders, A. P., Parker, P. J., Thompson, B. J. and McDonald, N. Q. (2016). aPKC Inhibition by Par3 CR3 Flanking Regions Controls Substrate Access and Underpins Apical-Junctional Polarization. Dev Cell 38: 384-398. PubMed ID: 27554858

    Khanal, I., Elbediwy, A., Diaz de la Loza, M.D., Fletcher, G.C. and Thompson, B.J. (2016). Shot and Patronin polarise microtubules to direct membrane traffic and biogenesis of microvilli in epithelia. J Cell Sci [Epub ahead of print]. PubMed ID: 27231092

    Ray, R. P., Matamoro-Vidal, A., Ribeiro, P. S., Tapon, N., Houle, D., Salazar-Ciudad, I. and Thompson, B. J. (2015). Patterned anchorage to the apical extracellular matrix defines tissue shape in the developing appendages of Drosophila. Dev Cell [Epub ahead of print]. PubMed ID: 26190146

  • Stephan Thor Developmental Biology, Linkoping University, Sweden
    Rubio-Ferrera, I., Baladron-de-Juan, P., Clarembaux-Badell, L., Truchado-Garcia, M., Jordan-Alvarez, S., Thor, S., Benito-Sipos, J. and Monedero Cobeta, I. (2022). Selective role of the DNA helicase Mcm5 in BMP retrograde signaling during Drosophila neuronal differentiation. PLoS Genet 18(6): e1010255. PubMed ID: 35737938

    Arefin, B., Parvin, F., Bahrampour, S., Stadler, C. B. and Thor, S. (2019). Drosophila Neuroblast Selection Is Gated by Notch, Snail, SoxB, and EMT Gene Interplay. Cell Rep 29(11): 3636-3651.e3633. PubMed ID: 31825841

    Curt, J. R., Yaghmaeian Salmani, B. and Thor, S. (2019). Anterior CNS expansion driven by brain transcription factors. Elife 8. PubMed ID: 31271353

    Bivik Stadler, C., Arefin, B., Ekman, H. and Thor, S. (2019). PIP degron-stabilized Dacapo/p21(Cip1) and mutations in ago act in an anti- versus pro-proliferative manner, yet both trigger an increase in Cyclin E levels. Development 146(13). PubMed ID: 31289041

    Stratmann, J., Ekman, H. and Thor, S. (2019). A branching gene regulatory network dictating different aspects of a neuronal cell identity. Development 146(6). PubMed ID: 30837222

    Bahrampour, S., Jonsson, C. and Thor, S. (2019). Brain expansion promoted by polycomb-mediated anterior enhancement of a neural stem cell proliferation program. PLoS Biol 17(2): e3000163. PubMed ID: 30807568

    Monedero Cobeta, I., Stadler, C. B., Li, J., Yu, P., Thor, S. and Benito-Sipos, J. (2018). Specification of Drosophila neuropeptidergic neurons by the splicing component brr2. PLoS Genet 14(8): e1007496. PubMed ID: 30133436

    Jonson, M., Nystrom, S., Sandberg, A., Carlback, M., Michno, W., Hanrieder, J., Starkenberg, A., Nilsson, K. P. R., Thor, S. and Hammarstrom, P. (2018). Aggregated Abeta1-42 is selectively toxic for neurons, whereas glial cells produce mature fibrils with low toxicity in Drosophila. Cell Chem Biol. PubMed ID: 29657084

    Yaghmaeian Salmani, B., Monedero Cobeta, I., Rakar, J., Bauer, S., Curt, J. R., Starkenberg, A. and Thor, S. (2018). Evolutionarily conserved anterior expansion of the central nervous system promoted by a common PcG-Hox program. Development. PubMed ID: 29530878

    Bahrampour, S., Gunnar, E., Jonsson, C., Ekman, H. and Thor, S. (2017). Neural Lineage Progression Controlled by a Temporal Proliferation Program. Dev Cell 43(3): 332-348.e334. PubMed ID: 29112852

    Fernius, J., Starkenberg, A. and Thor, S. (2017). Bar-coding neurodegeneration: Identifying sub-cellular effects of human neurodegenerative disease proteins using Drosophila leg neurons. Dis Model Mech [Epub ahead of print]. PubMed ID: 28615189

    Stratmann, J. and Thor, S. (2017). Neuronal cell fate specification by the molecular convergence of different spatio-temporal cues on a common initiator terminal selector gene. PLoS Genet 13(4): e1006729. PubMed ID: 28414802

  • Kevin Thornton Department of Ecology and Evolutionary Biology at the University of California, Irvine.
    Fabian, D. K., Melike Donertas, H., Fuentealba, M., Partridge, L. and Thornton, J. M. (2021). Transposable element landscape in Drosophila populations selected for longevity. Genome Biol Evol. PubMed ID: 33595657

    Bolukbasi, E., Woodling, N. S., Ivanov, D. K., Adcott, J., Foley, A., Rajasingam, A., Gittings, L. M., Aleyakpo, B., Niccoli, T., Thornton, J. M. and Partridge, L. (2021). Cell type-specific modulation of healthspan by Forkhead family transcription factors in the nervous system. Proc Natl Acad Sci U S A 118(8). PubMed ID: 33593901

    Ivanov, D. K., Bostelmann, G., Lan-Leung, B., Williams, J., Partridge, L., Escott-Price, V. and Thornton, J. M. (2020). A novel computational approach for predicting complex phenotypes in Drosophila (starvation-sensitive and sterile) by deriving their gene expression signatures from public data. PLoS One 15(10): e0240824. PubMed ID: 33104720

    Bolukbasi, E., Khericha, M., Regan, J. C., Ivanov, D. K., Adcott, J., Dyson, M. C., Nespital, T., Thornton, J. M., Alic, N. and Partridge, L. (2017). Intestinal Fork Head Regulates Nutrient Absorption and Promotes Longevity. Cell Rep 21(3): 641-653. PubMed ID: 29045833

  • Andreas Thum Neurobiology, Konstanz University
    Schumann, I., Berger, M., Nowag, N., Schafer, Y., Saumweber, J., Scholz, H. and Thum, A. S. (2021). Ethanol-guided behavior in Drosophila larvae. Sci Rep 11(1): 12307. PubMed ID: 34112872

    Brunner, B., Saumweber, J., Samur, M., Weber, D., Schumann, I., Mahishi, D., Rohwedder, A. and Thum, A. S. (2020). Food restriction reconfigures naive and learned choice behavior in Drosophila larvae. J Neurogenet: 1-10. PubMed ID: 31975653

    Mancini, N., Hranova, S., Weber, J., Weiglein, A., Schleyer, M., Weber, D., Thum, A. S. and Gerber, B. (2019). Reversal learning in Drosophila larvae. Learn Mem 26(11): 424-435. PubMed ID: 31615854

    Lyutova, R., Selcho, M., Pfeuffer, M., Segebarth, D., Habenstein, J., Rohwedder, A., Frantzmann, F., Wegener, C., Thum, A. S. and Pauls, D. (2019). Reward signaling in a recurrent circuit of dopaminergic neurons and peptidergic Kenyon cells. Nat Commun 10(1): 3097. PubMed ID: 31308381

    Saumweber, T., Rohwedder, A., Schleyer, M., Eichler, K., Chen, Y. C., Aso, Y., Cardona, A., Eschbach, C., Kobler, O., Voigt, A., Durairaja, A., Mancini, N., Zlatic, M., Truman, J. W., Thum, A. S. and Gerber, B. (2018). Functional architecture of reward learning in mushroom body extrinsic neurons of larval Drosophila. Nat Commun 9(1): 1104. PubMed ID: 29549237

    Huser, A., Eschment, M., Gullu, N., Collins, K. A. N., Bopple, K., Pankevych, L., Rolsing, E. and Thum, A. S. (2017). Anatomy and behavioral function of serotonin receptors in Drosophila melanogaster larvae. PLoS One 12(8): e0181865. PubMed ID: 28777821

    Eichler, K., Li, F., Litwin-Kumar, A., Park, Y., Andrade, I., Schneider-Mizell, C. M., Saumweber, T., Huser, A., Eschbach, C., Gerber, B., Fetter, R. D., Truman, J. W., Priebe, C. E., Abbott, L. F., Thum, A. S., Zlatic, M. and Cardona, A. (2017). The complete connectome of a learning and memory centre in an insect brain. Nature 548(7666): 175-182. PubMed ID: 28796202

    Widmann, A., Artinger, M., Biesinger, L., Boepple, K., Peters, C., Schlechter, J., Selcho, M. and Thum, A. S. (2016). Genetic dissection of aversive associative olfactory learning and memory in Drosophila larvae. PLoS Genet 12: e1006378. PubMed ID: 27768692

    Apostolopoulou, A. A., Kohn, S., Stehle, B., Lutz, M., Wust, A., Mazija, L., Rist, A., Galizia, C. G., Ludke, A. and Thum, A. S. (2016). Caffeine taste signaling in Drosophila larvae. Front Cell Neurosci 10: 193. PubMed ID: 27555807

    Rohwedder, A., Wenz, N. L., Stehle, B., Huser, A., Yamagata, N., Zlatic, M., Truman, J. W., Tanimoto, H., Saumweber, T., Gerber, B. and Thum, A. S. (2016). Four individually identified paired dopamine neurons signal reward in larval Drosophila. Curr Biol. PubMed ID: 26877086

    Rohwedder, A., Selcho, M., Chassot, B. and Thum, A. S. (2015). Neuropeptide F neurons modulate sugar reward during associative olfactory learning of Drosophila larvae. J Comp Neurol 523: Spc1. PubMed ID: 26463443

  • Carl Thummel Department of Human Genetics, University of Utah
    Lam, G., Beebe, K. and Thummel, C. S. (2022). A direct-drive GFP reporter for studies of tracheal development in Drosophila. Fly (Austin) 16(1): 105-110. PubMed ID: 35094652

    Lam, G., Nam, H. J., Velentzas, P. D., Baehrecke, E. H. and Thummel, C. S. (2021). Drosophila E93 promotes adult development and suppresses larval responses to ecdysone during metamorphosis. Dev Biol. PubMed ID: 34648816

    Praggastis, S. A., Nam, H. J., Lam, G., Child Vi, M. B., Castillo, D. M. and Thummel, C. S. (2021). Regulation of male fertility and accessory gland gene expression by the Drosophila HR39 nuclear receptor. Dev Biol 479: 51-60. PubMed ID: 34331899

    Praggastis, S. A., Lam, G., Horner, M. A., Nam, H. J. and Thummel, C. S. (2020). The Drosophila E78 nuclear receptor regulates dietary triglyceride uptake and systemic lipid levels. Dev Dyn. PubMed ID: 33368768

    Beebe, K., Robins, M. M., Hernandez, E. J., Lam, G., Horner, M. A. and Thummel, C. S. (2020). Drosophila estrogen-related receptor directs a transcriptional switch that supports adult glycolysis a

    Bensard, C. L., Wisidagama, D. R., Olson, K. A., Berg, J. A., Krah, N. M., Schell, J. C., Nowinski, S. M., Fogarty, S., Bott, A. J., Wei, P., Dove, K. K., Tanner, J. M., Panic, V., Cluntun, A., Lettlova, S., Earl, C. S., Namnath, D. F., Vazquez-Arreguin, K., Villanueva, C. J., Tantin, D., Murtaugh, L. C., Evason, K. J., Ducker, G. S., Thummel, C. S. and Rutter, J. (2019). Regulation of tumor initiation by the mitochondrial pyruvate carrier. Cell Metab. PubMed ID: 31813825

    Wisidagama, D. R. and Thummel, C. S. (2019). Regulation of Drosophila intestinal stem cell proliferation by enterocyte mitochondrial pyruvate metabolism. G3 (Bethesda). PubMed ID: 31488514

    Wisidagama, D. R., Thomas, S. M., Lam, G. and Thummel, C. S. (2019). Functional analysis of Aarf domain-containing kinase 1 in Drosophila melanogaster. Dev Dyn. PubMed ID: 31175694

    Storelli, G., Nam, H. J., Simcox, J., Villanueva, C. J. and Thummel, C. S. (2018). Drosophila HNF4 directs a switch in lipid metabolism that supports the transition to adulthood. Dev Cell. PubMed ID: 30554999

    Marxreiter, S. and Thummel, C. S. (2017). Adult functions for the Drosophila DHR78 nuclear receptor. Dev Dyn [Epub ahead of print]. PubMed ID: 29171103

  • Tony Tiganis Monash University, Australia
    Buszard, B. J., Johnson, T. K., Meng, T. C., Burke, R., Warr, C. G., Tiganis, T. (2013) The nuclear and ER-targeted forms of the protein tyrosine phosphatase PTP61F regulate Drosophila growth, lifespan and fecundity. Mol Cell Biol. PubMed ID: 23339871

    Wu, C. L., Buszard, B., Teng, C. H., Chen, W. L., Warr, C. G., Tiganis, T. and Meng, T. C. (2011). Dock/Nck facilitates PTP61F/PTP1B regulation of insulin signalling. Biochem J 439: 151-159. PubMed ID: 21707536

  • Chao-Ti Ting Institute of Ecology and Evolutionary Biology
    Li, J., Jiang, L., Wu, C. I., Lu, X., Fang, S. and Ting, C. T. (2019). Small Segmental Duplications in Drosophila-High Rate of Emergence and Elimination. Genome Biol Evol 11(2): 486-496. PubMed ID: 30689862

    >Chang, C. C., Ting, C. T., Chang, C. H., Fang, S. and Chang, H. Y. (2014). The persistence of facultative parthenogenesis in Drosophila albomicans. PLoS One 9(11): e113275. PubMed ID: 25415200

    Yang, H., He, B. Z., Ma, H., Tsaur, S. C., Ma, C., Wu, Y., Ting, C. T. and Zhang, Y. E. (2015). Expression profile and gene age jointly shaped the genome-wide distribution of premature termination codons in a Drosophila melanogaster population. Mol Biol Evol 32(1): 216-228. PubMed ID: 25371429

  • Robert Tjian Molecular and Cell Biology, Berkeley
    Zhang, Z., Boskovic, Z., Hussain, M. M., Hu, W., Inouye, C., Kim, H. J., Abole, A. K., Doud, M. K., Lewis, T. A., Koehler, A. N., Schreiber, S. L. and Tjian, R. (2015). Chemical perturbation of an intrinsically disordered region of TFIID distinguishes two modes of transcription initiation. Elife 4. PubMed ID: 26314865

    Guglielmi, B., La Rochelle, N. and Tjian, R. (2013). Gene-specific transcriptional mechanisms at the histone gene cluster revealed by single-cell imaging. Mol Cell 51: 480-492. PubMed ID: 23973376

  • Sokol Todi Department of Pharmacology & Department of Neurology, Wayne State University School of Medicine, Detroit
    Johnson, S. L., Prifti, M. V., Sujkowski, A., Libohova, K., Blount, J. R., Hong, L., Tsou, W. L. and Todi, S. V. (2022). Drosophila as a Model of Unconventional Translation in Spinocerebellar Ataxia Type 3. Cells 11(7). PubMed ID: 35406787

    Sujkowski, A., Richardson, K., Prifti, M. V., Wessells, R. J. and Todi, S. V. (2022). Endurance exercise ameliorates phenotypes in Drosophila models of spinocerebellar ataxias. Elife 11. PubMed ID: 35170431

    Johnson, S. L., Libohova, K., Blount, J. R., Sujkowski, A. L., Prifti, M. V., Tsou, W. L. and Todi, S. V. (2021). Targeting the VCP-binding motif of ataxin-3 improves phenotypes in Drosophila models of Spinocerebellar Ataxia Type 3. Neurobiol Dis 160: 105516. PubMed ID: 34563642

    Johnson, S. L., Libohova, K., Blount, J. R., Sujkowski, A. L., Prifti, M. V., Tsou, W. L. and Todi, S. V. (2021). Targeting the VCP-binding motif of ataxin-3 improves phenotypes in Drosophila models of Spinocerebellar Ataxia Type 3. Neurobiol Dis 160: 105516. PubMed ID: 34563642

    Johnson, S. L., Ranxhi, B., Libohova, K., Tsou, W. L. and Todi, S. V. (2020). Ubiquitin-interacting motifs of ataxin-3 regulate its polyglutamine toxicity through Hsc70-4-dependent aggregation. Elife 9. PubMed ID: 32955441

    Blount, J. R., Libohova, K., Silva, G. M. and Todi, S. V. (2020). Isoleucine 44 Hydrophobic Patch Controls Toxicity of Unanchored, Linear Ubiquitin Chains through NF-kappaB Signaling. Cells 9(6). PubMed ID: 32580388

    Johnson, S. L., Blount, J. R., Libohova, K., Ranxhi, B., Paulson, H. L., Tsou, W. L. and Todi, S. V. (2019). Differential toxicity of ataxin-3 isoforms in Drosophila models of Spinocerebellar Ataxia Type 3. Neurobiol Dis: 104535. PubMed ID: 31310802

    Blount, J. R., Meyer, D. N., Akemann, C., Johnson, S. L., Gurdziel, K., Baker, T. R. and Todi, S. V. (2019). Unanchored ubiquitin chains do not lead to marked alterations in gene expression in Drosophila melanogaster. Biol Open. PubMed ID: 31097444

    Blount, J. R., Libohova, K., Marsh, G. B., Sutton, J. R. and Todi, S. V. (2018). Expression and regulation of deubiquitinase-resistant, unanchored ubiquitin chains in Drosophila. Sci Rep 8(1): 8513. PubMed ID: 29855490

    Ristic, G., Sutton, J. R., Libohova, K. and Todi, S. V. (2018). Toxicity and aggregation of the polyglutamine disease protein, ataxin-3 is regulated by its binding to VCP/p97 in Drosophila melanogaster. Neurobiol Dis. PubMed ID: 29704548

    Tsou, W. L., Qiblawi, S. H., Hosking, R. R., Gomez, C. M. and Todi, S. V. (2016). Polyglutamine length-dependent toxicity from alpha1ACT in Drosophila models of spinocerebellar ataxia type 6. Biol Open 5(12): 1770-1775. PubMed ID: 27979829

  • Nicholas Tolwinski Life Sciences, Yale-NUS College, Singapore
    Kaur, P., Chua, E. H. Z., Lim, W. K., Liu, J., Harmston, N. and Tolwinski, N. S. (2022). Wnt Signaling Rescues Amyloid Beta-Induced Gut Stem Cell Loss. Cells 11(2). PubMed ID: 35053396

    Kaur, P., Kibat, C., Teo, E., Gruber, J., Mathuru, A. and Tolwinski, A. N. S. (2020). Use of Optogenetic Amyloid-beta to Monitor Protein Aggregation in Drosophila melanogaster, Danio rerio and Caenorhabditis elegans. Bio Protoc 10(23): e3856. PubMed ID: 33659494

    Lim, C. H., Kaur, P., Teo, E., Lam, V. Y. M., Zhu, F., Kibat, C., Gruber, J., Mathuru, A. S. and Tolwinski, N. S. (2020). Application of optogenetic Amyloid-beta distinguishes between metabolic and physical damages in neurodegeneration. Elife 9. PubMed ID: 32228858

    Bunnag, N., Tan, Q. H., Kaur, P., Ramamoorthy, A., Sung, I. C. H., Lusk, J. and Tolwinski, N. S. (2020). An Optogenetic Method to Study Signal Transduction in Intestinal Stem Cell Homeostasis. J Mol Biol 432(10): 3159-3176. PubMed ID: 32201167

    Admasu, T. D., Chaithanya Batchu, K., Barardo, D., Ng, L. F., Lam, V. Y. M., Xiao, L., Cazenave-Gassiot, A., Wenk, M. R., Tolwinski, N. S. and Gruber, J. (2018). Drug Synergy Slows Aging and Improves Healthspan through IGF and SREBP Lipid Signaling. Dev Cell 47(1): 67-79. PubMed ID: 30269951

    Kaur, P., Jin, H. J., Lusk, J. B. and Tolwinski, N. S. (2018). Modeling the Role of Wnt Signaling in Human and Drosophila Stem Cells. Genes (Basel) 9(2). PubMed ID: 29462894

  • Pavel Tomancack Max Planck Institute of Molecular Cell Biology and Genetics, Dresden
    Skouloudaki, K., Christodoulou, I., Khalili, D., Tsarouhas, V., Samakovlis, C., Tomancak, P., Knust, E. and Papadopoulos, D. K. (2019). Yorkie controls tube length and apical barrier integrity during airway development. J Cell Biol. PubMed ID: 31315941

    Viktorinova, I., Haase, R., Pietzsch, T., Henry, I. and Tomancak, P. (2019). Analysis of actomyosin dynamics at local cellular and tissue scales using time-lapse movies of cultured Drosophila egg chambers. J Vis Exp(148). PubMed ID: 31205315

    Skouloudaki, K., Papadopoulos, D. K., Tomancak, P. and Knust, E. (2019). The apical protein Apnoia interacts with Crumbs to regulate tracheal growth and inflation. PLoS Genet 15(1): e1007852. PubMed ID: 30645584

    Papadopoulos, D. K., Skouloudaki, K., Engstrom, Y., Terenius, L., Rigler, R., Zechner, C., Vukojevic, V. and Tomancak, P. (2019). Control of Hox transcription factor concentration and cell-to-cell variability by an auto-regulatory switch. Development. PubMed ID: 30642837

    Viktorinova, I., Henry, I. and Tomancak, P. (2017). Epithelial rotation is preceded by planar symmetry breaking of actomyosin and protects epithelial tissue from cell deformations. PLoS Genet 13(11): e1007107. PubMed ID: 29176774

    Börner, K., Jain, D., Vazquez-Pianzola, P., Vengadasalam, S., Steffen, N., Fyodorov, D.V., Tomancak, P., Konev, A., Suter, B. and Becker, P.B. (2016). A role for tuned levels of nucleosome remodeler subunit ACF1 during Drosophila oogenesis. Dev Biol [Epub ahead of print]. PubMed ID: 26851213

    Dunst, S., Kazimiers, T., von Zadow, F., Jambor, H., Sagner, A., Brankatschk, B., Mahmoud, A., Spannl, S., Tomancak, P., Eaton, S. and Brankatschk, M. (2015). Endogenously tagged rab proteins: a resource to study membrane trafficking in Drosophila. Dev Cell 33: 351-365. PubMed ID: 25942626

    Jambor, H., Surendranath, V., Kalinka, A. T., Mejstrik, P., Saalfeld, S. and Tomancak, P. (2015). Systematic imaging reveals features and changing localization of mRNAs in Drosophila development. Elife 4. PubMed ID: 25838129

    Jug, F., Pietzsch, T., Preibisch, S. and Tomancak, P. (2014). Bioimage Informatics in the context of Drosophila research. Methods. PubMed ID: 24732429

    Kayserili, M. A., Gerrard, D. T., Tomancak, P. and Kalinka, A. T. (2012). An excess of gene expression divergence on the X chromosome in Drosophila embryos: implications for the faster-X hypothesis. PLoS Genet 8: e1003200. PubMed ID: 23300473

    Cardona, A., Saalfeld, S., Preibisch, S., Schmid, B., Cheng, A., Pulokas, J., Tomancak, P. and Hartenstein, V. (2010). An integrated micro- and macroarchitectural analysis of the Drosophila brain by computer-assisted serial section electron microscopy. PLoS Biol 8. PubMed ID: 20957184

  • Yukihide Tomari Institute of Molecular and Cellular Biosciences, The University of Tokyo
    Naganuma, M., Tadakuma, H. and Tomari, Y. (2021). Single-molecule analysis of processive double-stranded RNA cleavage by Drosophila Dicer-2. Nat Commun 12(1): 4268. PubMed ID: 34257295

    Tsuboyama, K., Osaki, T., Matsuura-Suzuki, E., Kozuka-Hata, H., Okada, Y., Oyama, M., Ikeuchi, Y., Iwasaki, S. and Tomari, Y. (2020). A widespread family of heat-resistant obscure (Hero) proteins protect against protein instability and aggregation. PLoS Biol 18(3): e3000632. PubMed ID: 32163402

    Kobayashi, H., Shoji, K., Kiyokawa, K., Negishi, L. and Tomari, Y. (2019). VCP machinery mediates autophagic degradation of empty Argonaute. Cell Rep 28(5): 1144-1153.e1144. PubMed ID: 31365860

    Kobayashi, H., Shoji, K., Kiyokawa, K., Negishi, L. and Tomari, Y. (2018). Iruka eliminates dysfunctional Argonaute by selective ubiquitination of its empty state. Mol Cell. PubMed ID: 30503771

    Tsuboyama, K., Tadakuma, H. and Tomari, Y. (2018). Conformational activation of Argonaute by distinct yet coordinated actions of the Hsp70 and Hsp90 chaperone systems. Mol Cell 70(4): 722-729.e724. Pubmed ID: 29775584

    Niinuma, S. and Tomari, Y. (2017). ATP is dispensable for both miRNA- and Smaug-mediated deadenylation reactions. RNA [Epub ahead of print]. PubMed ID: 28250202

    Niinuma, S., Fukaya, T. and Tomari, Y. (2016). CCR4 and CAF1 deadenylases have an intrinsic activity to remove the post-poly(A) sequence. RNA [Epub ahead of print]. PubMed ID: 27484313

    Yao, C., Sasaki, H. M., Ueda, T., Tomari, Y. and Tadakuma, H. (2015). Single-molecule analysis of the target cleavage reaction by the Drosophila RNAi enzyme complex. Mol Cell 59: 125-132. PubMed ID: 26140368

    Iwasaki, S., Sasaki, H. M., Sakaguchi, Y., Suzuki, T., Tadakuma, H. and Tomari, Y. (2015). Defining fundamental steps in the assembly of the Drosophila RNAi enzyme complex. Nature [Epub ahead of print]. PubMed ID: 25822791

    Fukaya, T., Iwakawa, H. O. and Tomari, Y. (2014). MicroRNAs block assembly of eIF4F translation initiation complex in Drosophila. Mol Cell 56: 67-78. PubMed ID: 25280104

  • Seth Tomchik Department of Neuroscience, Scripps Florida
    Stahl, A., Noyes, N. C., Boto, T., Botero, V., Broyles, C. N., Jing, M., Zeng, J., King, L. B., Li, Y., Davis, R. L. and Tomchik, S. M. (2022). Associative learning drives longitudinally graded presynaptic plasticity of neurotransmitter release along axonal compartments. Elife 11. PubMed ID: 35285796

    Botero, V., Stanhope, B. A., Brown, E. B., Grenci, E. C., Boto, T., Park, S. J., King, L. B., Murphy, K. R., Colodner, K. J., Walker, J. A., Keene, A. C., Ja, W. W. and Tomchik, S. M. (2021). Neurofibromin regulates metabolic rate via neuronal mechanisms in Drosophila. Nat Commun 12(1): 4285. PubMed ID: 34257279

    King, L. B., Boto, T., Botero, V., Aviles, A. M., Jomsky, B. M., Joseph, C., Walker, J. A. and Tomchik, S. M. (2020). Developmental loss of neurofibromin across distributed neuronal circuits drives excessive grooming in Drosophila. PLoS Genet 16(7): e1008920. PubMed ID: 32697780

    Boto, T., Stahl, A. and Tomchik, S. M. (2020). Cellular and circuit mechanisms of olfactory associative learning in Drosophila. J Neurogenet: 1-11. PubMed ID: 32043414

    Boto, T., Stahl, A., Zhang, X., Louis, T. and Tomchik, S. M. (2019). Independent contributions of discrete dopaminergic circuits to cellular plasticity, memory strength, and valence in Drosophila. Cell Rep 27(7): 2014-2021.e2012. PubMed ID: 31091441

    Louis, T., Stahl, A., Boto, T. and Tomchik, S. M. (2018). Cyclic AMP-dependent plasticity underlies rapid changes in odor coding associated with reward learning. Proc Natl Acad Sci U S A 115(3): E448-e457. PubMed ID: 29284750

    Murphy, K.R., Deshpande, S.A., Yurgel, M.E., Quinn, J.P., Weissbach, J.L., Keene, A.C., Dawson-Scully, K., Huber, R., Tomchik, S.M. and Ja, W.W. (2016). Postprandial sleep mechanics in Drosophila. Elife 5. PubMed ID: 27873574

    King, L. B., Koch, M., Murphy, K., Velazquez, Y., Ja, W. W. and Tomchik, S. M. (2016). Neurofibromin Loss of Function Drives Excessive Grooming in Drosophila. G3 (Bethesda). PubMed ID: 26896440

    Boto, T., Louis, T., Jindachomthong, K., Jalink, K. and Tomchik, S. M. (2014). Dopaminergic Modulation of cAMP Drives Nonlinear Plasticity across the Drosophila Mushroom Body Lobes. Curr Biol. PubMed ID: 24684937

    Tomchik, S. M. (2013) Dopaminergic neurons encode a distributed, asymmetric representation of temperature in Drosophila. J Neurosci 33: 2166-2176. PubMed ID: 23365252

  • Jun Tomita Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
    Nakagawa, H., Maehara, S., Kume, K., Ohta, H. and Tomita, J. (2022). Biological functions of α2-adrenergic-like octopamine receptor in Drosophila melanogaster. Genes Brain Behav: e12807. PubMed ID: 35411674

    Yamaguchi, S. T., Kobayashi, R., Tomita, J. and Kume, K. (2022). The regulation of circadian rhythm by insulin signaling in Drosophila. Neurosci Res. PubMed ID: 35872183

    Nakagawa, H., Maehara, S., Kume, K., Ohta, H. and Tomita, J. (2022). Biological functions of alpha2-adrenergic-like octopamine receptor in Drosophila melanogaster. Genes Brain Behav 21(6): e12807. PubMed ID: 35411674

    Yamaguchi, S. T., Tomita, J. and Kume, K. (2022). Insulin signaling in clock neurons regulates sleep in Drosophila. Biochem Biophys Res Commun 591: 44-49. PubMed ID: 34998032

  • Andrew Tomlinson Department of Genetics and Development, Columbia University Medical Center, New York
    Katanaev, V. L., Egger-Adam, D. and Tomlinson, A. (2018). Antagonistic PCP signaling pathways in the developing Drosophila eye. Sci Rep 8(1): 5741. PubMed ID: 29636485

    Duan, H., de Navas, L. F., Hu, F., Sun, K., Mavromatakis, Y. E., Viets, K., Zhou, C., Kavaler, J., Johnston, R. J., Jr., Tomlinson, A. and Lai, E. C. (2018). The mir-279/996 cluster represses receptor tyrosine kinase signaling to determine cell fates in the Drosophila eye. Development. PubMed ID: 29540498

    Mavromatakis, Y.E. and Tomlinson, A. (2016). R7 photoreceptor specification in the developing Drosophila eye: The role of the transcription factor Deadpan. PLoS Genet 12: e1006159. PubMed ID: 27427987

    Kumar, S. R., Patel, H. and Tomlinson, A. (2015). Wingless mediated apoptosis: How cone cells direct the death of peripheral ommatidia in the developing Drosophila eye. Dev Biol. PubMed ID: 26428511

    Mavromatakis, Y. E. and Tomlinson, A. (2013). Switching cell fates in the developing Drosophila eye. Development. PubMed ID: 24067351

    Mavromatakis, Y. E. and Tomlinson, A. (2012). Stop and go: antagonistic signals in the specification of the Drosophila R7 photoreceptor viewed from an evolutionary perspective. Fly (Austin) 6: 228-233. PubMed ID: 22878552

    Tomlinson, A. (2012). The origin of the Drosophila subretinal pigment layer. J Comp Neurol 520: 2676-2682. PubMed ID: 22684937

    Mavromatakis, Y. E. and Tomlinson, A. (2012). The role of the small GTPase Rap in Drosophila R7 photoreceptor specification. Proc Natl Acad Sci U S A 109: 3844-3849. PubMed ID: 22355117

  • Chou Tong Life Sciences Institue, Zhejiang University
    Duan, X., Xu, L., Li, Y., Jia, L., Liu, W., Shao, W., Bayat, V., Shang, W., Wang, L., Liu, J. P. and Tong, C. (2021). Regulation of lipid homeostasis by the TBC protein dTBC1D22 via modulation of the small GTPase Rab40 to facilitate lipophagy. Cell Rep 36(9): 109541. PubMed ID: 34469730

    Zhou, J., Xu, L., Duan, X., Liu, W., Zhao, X., Wang, X., Shang, W., Fang, X., Yang, H., Jia, L., Bai, J., Zhao, J., Wang, L. and Tong, C. (2019). Large-scale RNAi screen identified Dhpr as a regulator of mitochondrial morphology and tissue homeostasis. Sci Adv 5(9): eaax0365. PubMed ID: 31555733

    Liu, W., Duan, X., Fang, X., Shang, W. and Tong, C. (2018). Mitochondrial protein import regulates cytosolic protein homeostasis and neuronal integrity. Autophagy. PubMed ID: 29909722

    Yu, J., Lan, X., Chen, X., Yu, C., Xu, Y., Liu, Y., Xu, L., Fan, H. Y. and Tong, C. (2016). Protein synthesis and degradation are critical to regulate germline stem cell homeostasis in Drosophila testes. Development. PubMed ID: 27471256

    Yu, J., Liu, Y., Lan, X., Wu, H., Wen, Y., Zhou, Z., Hu, Z., Sha, J., Guo, X. and Tong, C. (2016). CHES-1-like, the ortholog of a non-obstructive azoospermia-associated gene, blocks germline stem cell differentiation by upregulating Dpp expression in Drosophila testis. Oncotarget [Epub ahead of print]. PubMed ID: 27281616

    Zhao, X., Yang, H., Liu, W., Duan, X., Shang, W., Xia, D. and Tong, C. (2015). Sec22 regulates endoplasmic reticulum morphology but not autophagy and is required for eye development in Drosophila. J Biol Chem 290: 7943-7951. PubMed ID: 25670863

    Yu, J., Wu, H., Wen, Y., Liu, Y., Zhou, T., Ni, B., Lin, Y., Dong, J., Zhou, Z., Hu, Z., Guo, X., Sha, J. and Tong, C. (2015). Identification of seven genes essential for male fertility through a genome-wide association study of non-obstructive azoospermia and RNA interference-mediated large-scale functional screening in Drosophila. Hum Mol Genet 24: 1493-1503. PubMed ID: 25361961

  • Tina Tootle Molecular & Cellular Biology Program, University of Iowa
    Lamb, M. C., Kaluarachchi, C. P., Lansakara, T. I., Mellentine, S. Q., Lan, Y., Tivanski, A. V. and Tootle, T. L. (2021). Fascin limits Myosin activity within Drosophila border cells to control substrate stiffness and promote migration. Elife 10. PubMed ID: 34698017

    Fox, E. F., Lamb, M. C., Mellentine, S. Q. and Tootle, T. L. (2020). Prostaglandins regulate invasive, collective border cell migration. Mol Biol Cell: mbcE19100578. PubMed ID: 32432969

    Lamb, M. C., Anliker, K. K. and Tootle, T. L. (2020). Fascin regulates protrusions and delamination to mediate invasive, collective cell migration in vivo. Dev Dyn. PubMed ID: 32352613

    Spracklen, A. J., Lamb, M. C., Groen, C. M. and Tootle, T. L. (2019). Pharmaco-genetic screen To uncover actin regulators targeted by prostaglandins during Drosophila oogenesis. G3 (Bethesda). PubMed ID: 31506320

    Wineland, D. M., Kelpsch, D. J. and Tootle, T. L. (2018). Multiple pools of nuclear actin. Anat Rec (Hoboken). PubMed ID: 30312534

    Kelpsch, D. J., Groen, C. M., Fagan, T. N., Sudhir, S. and Tootle, T. L. (2016). Fascin regulates nuclear actin during Drosophila oogenesis. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27535426

    Groen, C. M., Jayo, A., Parsons, M. and Tootle, T. L. (2015). Prostaglandins regulate nuclear localization of Fascin and its function in nucleolar architecture. Mol Biol Cell [Epub ahead of print]. PubMed ID: 25808493

    Spracklen, A. J. and Tootle, T. L. (2013). The Utility of Stage-specific Mid-to-late Drosophila Follicle Isolation. J Vis Exp. PubMed ID: 24326735

    Spracklen, A. J., Kelpsch, D. J., Chen, X., Spracklen, C. N. and Tootle, T. L. (2013). Prostaglandins temporally regulate cytoplasmic actin bundle formation during Drosophila oogenesis. Mol Biol Cell. PubMed ID: 24284900

    Groen, C. M., Spracklen, A. J., Fagan, T. N. and Tootle, T. L. (2012). Drosophila Fascin is a novel downstream target of prostaglandin signaling during actin remodeling. Mol Biol Cell 23: 4567-4578. PubMed ID: 23051736

    Tootle, T. L., Williams, D., Hubb, A., Frederick, R. and Spradling, A. (2011). Drosophila eggshell production: identification of new genes and coordination by Pxt. PLoS One 6: e19943. PubMed ID: 21637834

  • Päivi Torkkeli Department of Physiology and Biophysics, Dalhousie University, Halifax
    French, A. S., Meisner, S., Su, C. Y. and Torkkeli, P. H. (2014). Carbon Dioxide and Fruit Odor Transduction in Drosophila Olfactory Neurons. What Controls their Dynamic Properties? PLoS One 9: e86347. PubMed ID: 24466044

    French, A. S., Torkkeli, P. H. and Schuckel, J. (2011). Dynamic characterization of Drosophila antennal olfactory neurons indicates multiple opponent signaling pathways in odor discrimination. J Neurosci 31: 861-869. PubMed ID: 21248109>

  • Laura Torroja Universidad Autonoma de Madrid
    Lopez-Arias, B., Turiegano, E., Monedero, I., Canal, I. and Torroja, L. (2017). Presynaptic Abeta40 prevents synapse addition in the adult Drosophila neuromuscular junction. PLoS One 12(5): e0177541. PubMed ID: 28520784

    Andres, M., Turiegano, E., Gopfert, M. C., Canal, I. and Torroja, L. (2014). The Extracellular Matrix Protein Artichoke Is Required for Integrity of Ciliated Mechanosensory and Chemosensory Organs in Drosophila Embryos. Genetics. PubMed ID: 24496014

    Gonzalo-Gomez, A., Turiegano, E., Leon, Y., Molina, I., Torroja, L. and Canal, I. (2012). Ih current is necessary to maintain normal dopamine fluctuations and sleep consolidation in Drosophila. PLoS One 7: e36477. PubMed ID: 22574167

    Gabilondo, H., Losada-Perez, M., del Saz, D., Molina, I., Leon, Y., Canal, I., Torroja, L. and Benito-Sipos, J. (2011). A targeted genetic screen identifies crucial players in the specification of the Drosophila abdominal Capaergic neurons. Mech Dev 128: 208-221. PubMed ID: 21236339

  • Katalin Fejes Tóth Department of Biology, California Institute of Technology
    Rogers, A. K., Situ, K., Perkins, E. M. and Toth, K. F. (2017). Zucchini-dependent piRNA processing is triggered by recruitment to the cytoplasmic processing machinery. Genes Dev 31(18): 1858-1869. PubMed ID: 29021243

    Le Thomas, A., Rogers, A. K., Webster, A., Marinov, G. K., Liao, S. E., Perkins, E. M., Hur, J. K., Aravin, A. A. and Toth, K. F. (2013). Piwi induces piRNA-guided transcriptional silencing and establishment of a repressive chromatin state. Genes Dev. PubMed ID: 23392610

    Caudron-Herger, M., Muller-Ott, K., Mallm, J. P., Marth, C., Schmidt, U., Fejes-Toth, K. and Rippe, K. (2011). Coding RNAs with a non-coding function: maintenance of open chromatin structure. Nucleus 2: 410-424. PubMed ID: 21983088

    Aravin, A. A., Sachidanandam, R., Bourc'his, D., Schaefer, C., Pezic, D., Toth, K. F., Bestor, T. and Hannon, G. J. (2008). A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice. Mol Cell 31: 785-799. PubMed ID: 18922463

  • John Tower Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California
    Landis, G. N., Hilsabeck, T. A. U., Bell, H. S., Ronnen-Oron, T., Wang, L., Doherty, D. V., Tejawinata, F. I., Erickson, K., Vu, W., Promislow, D. E. L., Kapahi, P. and Tower, J. (2021). Mifepristone Increases Life Span of Virgin Female Drosophila on Regular and High-fat Diet Without Reducing Food Intake. Front Genet 12: 751647. PubMed ID: 34659367

    Landis, G. N., Doherty, D. V., Yen, C. A., Wang, L., Fan, Y., Wang, I., Vroegop, J., Wang, T., Wu, J., Patel, P., Lee, S., Abdelmesieh, M., Shen, J., Promislow, D. E. L., Curran, S. P. and Tower, J. (2020). Metabolic Signatures of Life Span Regulated by Mating, Sex Peptide and Mifepristone/RU486 in Female Drosophila melanogaster. J Gerontol A Biol Sci Med Sci. PubMed ID: 32648907

    Pomatto, L. C., Wong, S., Carney, C., Shen, B., Tower, J. and Davies, K. J. (2017). The age- and sex-specific decline of the 20s proteasome and the Nrf2/CncC signal transduction pathway in adaption and resistance to oxidative stress in Drosophila melanogaster. Aging (Albany NY). PubMed ID: 28373600

    Pomatto, L. C., Carney, C., Shen, B., Wong, S., Halaszynski, K., Salomon, M. P., Davies, K. J. and Tower, J. (2016). The mitochondrial Lon protease is required for age-specific and sex-specific adaptation to oxidative stress. Curr Biol [Epub ahead of print]. PubMed ID: 27916526

    Fear, J.M., Leon-Novelo, L.G., Morse, A.M., Gerken, A.R., Van Lehman, K., Tower, J., Nuzhdin, S.V. and McIntyre, L.M. (2016). Buffering of genetic regulatory networks in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 27194752


    Fear, J. M., Arbeitman, M. N., Salomon, M. P., Dalton, J. E., Tower, J., Nuzhdin, S. V. and McIntyre, L. M. (2015). The Wright stuff: Reimagining path analysis reveals novel components of the sex determination hierarchy in Drosophila melanogaster. BMC Syst Biol 9: 53. PubMed ID: 26335107

    Landis, G. N., Salomon, M. P., Keroles, D., Brookes, N., Sekimura, T. and Tower, J. (2015). The progesterone antagonist mifepristone/RU486 blocks the negative effect on life span caused by mating in female Drosophila. Aging (Albany NY). PubMed ID: 25614682

    Tower, J., Landis, G., Gao, R., Luan, A., Lee, J. and Sun, Y. (2013). Variegated Expression of Hsp22 Transgenic Reporters Indicates Cell-specific Patterns of Aging in Drosophila Oenocytes. J Gerontol A Biol Sci Med Sci. PubMed ID: 23723429

    Landis, G., Shen, J. and Tower, J. (2012). Gene expression changes in response to aging compared to heat stress, oxidative stress and ionizing radiation in Drosophila melanogaster. Aging (Albany NY) 4: 768-789. PubMed ID: 23211361

    Pickering, A. M., Staab, T. A., Tower, J., Sieburth, D. S. and Davies, K. J. (2012). A conserved role for the 20S proteasome and Nrf2 transcription factor in oxidative-stress adaptation in mammals, C. elegans and D. melanogaster. J Exp Biol. PubMed ID: 23038734

  • Dan Tracey Duke University Medical Center
    Song, W., Ressl, S. and Tracey, W. D. (2020). Loss of Pseudouridine Synthases in the RluA Family Causes Hypersensitive Nociception in Drosophila. G3 (Bethesda). PubMed ID: 33028630

    He, L., Gulyanon, S., Mihovilovic Skanata, M., Karagyozov, D., Heckscher, E. S., Krieg, M., Tsechpenakis, G., Gershow, M. and Tracey, W. D. (2019). Direction selectivity in Drosophila proprioceptors requires the mechanosensory channel Tmc. Curr Biol 29(6): 945-956.e943. PubMed ID: 30853433

    Walcott, K. C. E., Mauthner, S. E., Tsubouchi, A., Robertson, J. and Tracey, W. D. (2018). The Drosophila small conductance calcium-activated potassium channel negatively regulates nociception. Cell Rep 24(12): 3125-3132. PubMed ID: 30231996

    Honjo, K., Mauthner, S. E., Wang, Y., Skene, J. H. and Tracey, W. D. (2016). Nociceptor-enriched genes required for normal thermal nociception. Cell Rep. PubMed ID: 27346357

    Mauthner, S. E., Hwang, R. Y., Lewis, A. H., Xiao, Q., Tsubouchi, A., Wang, Y., Honjo, K., Skene, J. H., Grandl, J. and Tracey, W. D., Jr. (2014). Balboa binds to Pickpocket in vivo and is required for mechanical nociception in Drosophila larvae. Curr Biol. PubMed ID: 25454784

    Robertson, J. L., Tsubouchi, A. and Tracey, W. D. (2013). Larval Defense against Attack from Parasitoid Wasps Requires Nociceptive Neurons. PLoS One 8: e78704. PubMed ID: 24205297

    Zhong, L., Bellemer, A., Yan, H., Ken, H., Jessica, R., Hwang, R. Y., Pitt, G. S. and Tracey, W. D. (2012). Thermosensory and nonthermosensory isoforms of Drosophila melanogaster TRPA1 reveal heat-sensor domains of a thermoTRP Channel. Cell Rep 1: 43-55. PubMed ID: 22347718

    Schwartz, N. U., Zhong, L., Bellemer, A. and Tracey, W. D. (2012). Egg laying decisions in Drosophila are consistent with foraging costs of larval progeny. PLoS One 7: e37910. PubMed ID: 22693584

    Zhong, L., Bellemer, A., Yan, H., Ken, H., Jessica, R., Hwang, R. Y., Pitt, G. S. and Tracey, W. D. (2012). Thermosensory and nonthermosensory isoforms of Drosophila melanogaster TRPA1 reveal heat-sensor domains of a thermoTRP Channel. Cell Rep 1: 43-55. PubMed ID: 22347718

  • Jessica TreismanThe Skirball Institute of Biomolecular Medicine, New York University School of Medicine
    Wang, H., Morrison, C. A., Ghosh, N., Tea, J. S., Call, G. B. and Treisman, J. E. (2022). The Blimp-1 transcription factor acts in non-neuronal cells to regulate terminal differentiation of the Drosophila eye. Development 149(7). PubMed ID: 35297965

    Ho, C. H., Paolantoni, C., Bawankar, P., Tang, Z., Brown, S., Roignant, J. Y. and Treisman, J. E. (2021). An exon junction complex-independent function of Barentsz in neuromuscular synapse growth. EMBO Rep: e53231. PubMed ID: 34726300

    Ho, C. H. and Treisman, J. E. (2020). Specific Isoforms of the Guanine-Nucleotide Exchange Factor dPix Couple Neuromuscular Synapse Growth to Muscle Growth. Dev Cell. PubMed ID: 32516570

    Letizia, A., He, D., Astigarraga, S., Colombelli, J., Hatini, V., Llimargas, M. and Treisman, J. E. (2019). Sidekick is a key component of tricellular adherens junctions that acts to resolve cell rearrangements. Dev Cell. PubMed ID: 31353315

    Suisse, A. and Treisman, J. E. (2019). Reduced SERCA function preferentially affects Wnt signaling by retaining E-Cadherin in the endoplasmic reticulum. Cell Rep 26(2): 322-329.e323. PubMed ID: 30625314

    Courgeon, M., He, D., Liu, H. H., Legent, K. and Treisman, J. E. (2018). The Drosophila Epidermal Growth Factor Receptor does not act in the nucleus. J Cell Sci. PubMed ID: 30158176

    Morrison, C. A., Chen, H., Cook, T., Brown, S. and Treisman, J. E. (2018). Glass promotes the differentiation of neuronal and non-neuronal cell types in the Drosophila eye. PLoS Genet 14(1): e1007173. PubMed ID: 29324767

    Astigarraga, S., Douthit, J., Tarnogorska, D., Creamer, M. S., Mano, O., Clark, D. A., Meinertzhagen, I. A. and Treisman, J. E. (2018). Drosophila Sidekick is required in developing photoreceptors to enable visual motion detection. Development. PubMed ID: 29361567

    Suisse, A., He, D., Legent, K. and Treisman, J. E. (2017). COP9 signalosome subunits protect Capicua from MAP kinase-dependent and independent mechanisms of degradation. Development [Epub ahead of print]. PubMed ID: 28619822

    Legent, K., Liu, H.H. and Treisman, J.E. (2015). Drosophila Vps4 promotes Epidermal growth factor receptor signaling independently of its role in receptor degradation. Development [Epub ahead of print]. PubMed ID: 25790850

    Malone, C. D., Mestdagh, C., Akhtar, J., Kreim, N., Deinhard, P., Sachidanandam, R., Treisman, J. and Roignant, J. Y. (2014). The exon junction complex controls transposable element activity by ensuring faithful splicing of the piwi transcript. Genes Dev 28: 1786-1799. PubMed ID: 25104425

  • Hervé Tricoire Unité Biologie Fonctionnelle et Adaptative, Université Paris Descartes
    Heidari, R., Monnier, V., Martin, E. and Tricoire, H. (2015). Methylene blue partially rescues heart defects in a Drosophila model of Huntington's disease. J Huntingtons Dis 4: 173-186. PubMed ID: 26397898

    Bouleau, S. and Tricoire, H. (2015). Drosophila models of Alzheimer's disease: advances, limits, and perspectives. J Alzheimers Dis 45: 1015-1038. PubMed ID: 25697708

    Tricoire, H., Palandri, A., Bourdais, A., Camadro, J. M. and Monnier, V. (2014). Methylene blue rescues heart defects in a Drosophila model of Friedreich's ataxia. Hum Mol Genet 23: 968-979. PubMed ID: 24105471

    Monnier, V., Iche-Torres, M., Rera, M., Contremoulins, V., Guichard, C., Lalevee, N., Tricoire, H. and Perrin, L. (2012). dJun and Vri/dNFIL3 are major regulators of cardiac aging in Drosophila. PLoS Genet 8: e1003081. PubMed ID: 23209438

  • John True Ecology & Evolution Department, SUNY, Stonybrook
    Lachance, J., Jung, L. and True, J. R. (2013). Genetic Background and GxE Interactions Modulate the Penetrance of a Naturally Occurring Wing Mutation in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 24002866

    Yukilevich, R., Turner, T. L., Aoki, F., Nuzhdin, S. V. and True, J. R. (2010). Patterns and processes of genome-wide divergence between North American and African Drosophila melanogaster. Genetics 186: 219-239. PubMed ID: 20551441

    Lachance, J. and True, J. R. (2010). X-autosome incompatibilities in Drosophila melanogaster: tests of Haldane's rule and geographic patterns within species. Evolution 64: 3035-3046. PubMed ID: 20455929

  • Jim Truman Janelia Farm, HHMI
    Eschbach, C., Fushiki, A., Winding, M., Afonso, B., Andrade, I. V., Cocanougher, B. T., Eichler, K., Gepner, R., Si, G., Valdes-Aleman, J., Fetter, R. D., Gershow, M., Jefferis, G. S., Samuel, A. D., Truman, J. W., Cardona, A. and Zlatic, M. (2021). Circuits for integrating learned and innate valences in the insect brain. Elife 10. PubMed ID: 34755599

    Mark, B., Lai, S. L., Zarin, A. A., Manning, L., Pollington, H. Q., Litwin-Kumar, A., Cardona, A., Truman, J. W. and Doe, C. Q. (2021). A developmental framework linking neurogenesis and circuit formation in the Drosophila CNS. Elife 10. PubMed ID: 33973523

    Agrawal, S., Dickinson, E. S., Sustar, A., Gurung, P., Shepherd, D., Truman, J. W. and Tuthill, J. C. (2020). Central processing of leg proprioception in Drosophila. Elife 9. PubMed ID: 33263281

    Lacin, H., Williamson, W. R., Card, G. M., Skeath, J. B. and Truman, J. W. (2020). Unc-4 acts to promote neuronal identity and development of the take-off circuit in the Drosophila CNS. Elife 9. PubMed ID: 32216875

    Miroschnikow, A., Schlegel, P., Schoofs, A., Hueckesfeld, S., Li, F., Schneider-Mizell, C. M., Fetter, R. D., Truman, J. W., Cardona, A. and Pankratz, M. J. (2018). Convergence of monosynaptic and polysynaptic sensory paths onto common motor outputs in a Drosophila feeding connectome. Elife 7. PubMed ID: 30526854

    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

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

    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

    Schlegel, P., Texada, M. J., Miroschnikow, A., Schoofs, A., Huckesfeld, S., Peters, M., Schneider-Mizell, C. M., Lacin, H., Li, F., Fetter, R. D., Truman, J. W., Cardona, A. and Pankratz, M. J. (2016). Synaptic transmission parallels neuromodulation in a central food-intake circuit. Elife 5. PubMed ID: 27845623

    Jovanic, T., Schneider-Mizell, C.M., Shao, M., Masson, J.B., Denisov, G., Fetter, R.D., Mensh, B.D., Truman, J.W., Cardona, A. and Zlatic, M. (2016). Competitive disinhibition mediates behavioral choice and sequences in Drosophila. Cell [Epub ahead of print]. PubMed ID: 27720450

    Hasegawa, E., Truman, J. W. and Nose, A. (2016). Identification of excitatory premotor interneurons which regulate local muscle contraction during Drosophila larval locomotion. Sci Rep 6: 30806. PubMed ID: 27470675

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

    Liang, L., Li, Y., Potter, C. J., Yizhar, O., Deisseroth, K., Tsien, R. W. and Luo, L. (2013). GABAergic projection neurons route selective olfactory inputs to specific higher-order neurons. Neuron 79: 917-931. PubMed ID: 24012005

  • Leo Tsuda National Center for Geriatrics and Gerontology, Laboratory of Experimental Animal Model Research, Obu, Japan
    Yamasaki, Y., Lim, Y. M., Minami, R. and Tsuda, L. (2021). A splicing variant of Charlatan, a Drosophila REST-like molecule, preferentially localizes to axons. Biochem Biophys Res Commun 578: 35-41. PubMed ID: 34536827

    Lim, Y. M., Yagi, Y. and Tsuda, L. (2015). Cellular defense and sensory cell survival require distinct functions of ebi in Drosophila. PLoS One 10: e0141457. PubMed ID: 26524764

    Amcheslavsky, A., Nie, Y., Li, Q., He, F., Tsuda, L., Markstein, M. and Ip, Y. T. (2014). Gene expression profiling identifies the zinc-finger protein Charlatan as a regulator of intestinal stem cells in Drosophila. Development 141: 2621-2632. PubMed ID: 24961799

    Tsuda, L. and Lim, Y. M. (2014). Regulatory system for the G1-arrest during neuronal development in Drosophila. Dev Growth Differ 56: 358-367. PubMed ID: 24738783

    Lim, Y. M., Yamasaki, Y. and Tsuda, L. (2013). Ebi alleviates excessive growth signaling through multiple epigenetic functions in Drosophila. Genes Cells 18: 909-920. PubMed ID: 23919509

  • Susan Tsunoda Department of Biomedical Sciences, Colorado State University. Fort Collins, Colorado
    Vallejos, M. J., Eadaim, A., Hahm, E. T. and Tsunoda, S. (2021). Age-related changes in Kv4/Shal and Kv1/Shaker expression in Drosophila and a role for reactive oxygen species. PLoS One 16(12): e0261087. PubMed ID: 34932577

    Byers, N., Hahm, E. T. and Tsunoda, S. (2021). Slo2/K(Na) Channels in Drosophila Protect Against Spontaneous and Induced Seizure-like Behavior Associated with an Increased Persistent Na(+) Current. J Neurosci. PubMed ID: 34544836

    Eadaim, A., Hahm, E. T., Justice, E. D. and Tsunoda, S. (2020). Cholinergic Synaptic Homeostasis Is Tuned by an NFAT-Mediated alpha7 nAChR-K(v)4/Shal Coupled Regulatory System. Cell Rep 32(10): 108119. PubMed ID: 32905767

    Ping, Y. and Tsunoda, S. (2012). Homeostatic plasticity in Drosophila central neurons, and implications in human diseases. Fly (Austin) 6(3): 153-157. PubMed ID: 22735167

    Lieu, M. H., Vallejos, M. J., Michael, E. and Tsunoda, S. (2012). Mechanisms underlying stage-1 TRPL channel translocation in Drosophila photoreceptors. PLoS One 7(2): e31622. PubMed ID: 22363689

    Ping, Y., Waro, G., Licursi, A., Smith, S., Vo-Ba, D. A. and Tsunoda, S. (2011). Shal/K(v)4 channels are required for maintaining excitability during repetitive firing and normal locomotion in Drosophila. PLoS One 6(1): e16043. PubMed ID: 21264215

  • Alexei Tulin Department of Ecology, Evolution, and Marine Biology, University of California, Santa Barbara
    Bordet, G., Lodhi, N., Kossenkov, A. and Tulin, A. (2021). Age-Related Changes of Gene Expression Profiles in Drosophila. Genes (Basel) 12(12). PubMed ID: 34946931

    Bordet, G., Lodhi, N., Guo, D., Kossenkov, A. and Tulin, A. V. (2020). Poly(ADP-ribose) polymerase 1 in genome-wide expression control in Drosophila. Sci Rep 10(1): 21151. PubMed ID: 33273587

    Thomas, C., Ji, Y., Wu, C., Datz, H., Boyle, C., MacLeod, B., Patel, S., Ampofo, M., Currie, M., Harbin, J., Pechenkina, K., Lodhi, N., Johnson, S. J. and Tulin, A. V. (2019). Hit and run versus long-term activation of PARP-1 by its different domains fine-tunes nuclear processes. Proc Natl Acad Sci U S A 116(20): 9941-9946. PubMed ID: 31028139

    Ji, Y. and Tulin, A. V. (2016). Poly(ADP-ribosyl)ation of hnRNP A1 protein controls translational repression in Drosophila. Mol Cell Biol [Epub ahead of print]. PubMed ID: 27402862

    Thomas, C. J., Kotova, E., Andrake, M., Adolf-Bryfogle, J., Glaser, R., Regnard, C. and Tulin, A. V. (2014). Kinase-mediated changes in nucleosome conformation trigger chromatin decondensation via poly(ADP-ribosyl)ation. Mol Cell 53: 831-842. PubMed ID: 24508391

    Boamah, E. K., Kotova, E., Garabedian, M., Jarnik, M. and Tulin, A. V. (2012). Poly(ADP-Ribose) polymerase 1 (PARP-1) regulates ribosomal biogenesis in Drosophila nucleoli. PLoS Genet 8: e1002442. PubMed ID: 22242017

    Garabedian, M., Jarnik, M., Kotova, E. and Tulin, A. V. (2011). Generating a knockdown transgene against Drosophila heterochromatic Tim17b gene encoding mitochondrial translocase subunit. PLoS One 6: e25945. PubMed ID: 21998726

    Kotova, E., Lodhi, N., Jarnik, M., Pinnola, A. D., Ji, Y. and Tulin, A. V. (2011). Drosophila histone H2A variant (H2Av) controls poly(ADP-ribose) polymerase 1 (PARP1) activation in chromatin. Proc Natl Acad Sci U S A 108: 6205-6210. PubMed ID: 21444826

  • Tim Tully Dart Neuroscience, San Diego
    Chen, A. Y. and Tully, T. (2018). A stress-enhanced model for discovery of disease-modifying gene: Ece1-suppresses the toxicity of alpha-synuclein A30P. Neurobiol Dis 114: 153-163. PubMed ID: 29524599

    Chen, A. Y., Xia, S., Wilburn, P. and Tully, T. (2014). Olfactory deficits in an alpha-synuclein fly model of Parkinson's disease. PLoS One 9: e97758. PubMed ID: 24879013

    Pai, T. P., Chen, C. C., Lin, H. H., Chin, A. L., Lai, J. S., Lee, P. T., Tully, T. and Chiang, A. S. (2013). Drosophila ORB protein in two mushroom body output neurons is necessary for long-term memory formation. Proc Natl Acad Sci U S A 110: 7898-7903. PubMed ID: 23610406

    Chen, C. C., Wu, J. K., Lin, H. W., Pai, T. P., Fu, T. F., Wu, C. L., Tully, T. and Chiang, A. S. (2012). Visualizing long-term memory formation in two neurons of the Drosophila brain. Science 335: 678-685. PubMed ID: 22323813

  • Glenn Turner Cold Spring Harbor Labs
    Hige, T., Aso, Y., Modi, M. N., Rubin, G. M. and Turner, G. C. (2015). Heterosynaptic Plasticity Underlies Aversive Olfactory Learning in Drosophila. Neuron 88: 985-998. PubMed ID: 26637800

    Gruntman, E. and Turner, G. C. (2013). Integration of the olfactory code across dendritic claws of single mushroom body neurons. Nat Neurosci. PubMed ID: 24141312

    Campbell, R. A., Honegger, K. S., Qin, H., Li, W., Demir, E. and Turner, G. C. (2013). Imaging a population code for odor identity in the Drosophila mushroom body. J Neurosci 33: 10568-10581. PubMed ID: 23785169

    Honegger, K. S., Campbell, R. A. and Turner, G. C. (2011). Cellular-resolution population imaging reveals robust sparse coding in the Drosophila mushroom body. J Neurosci 31: 11772-11785. PubMed ID: 21849538

    Wu, C. L., Shih, M. F., Lai, J. S., Yang, H. T., Turner, G. C., Chen, L. and Chiang, A. S. (2011). Heterotypic gap junctions between two neurons in the drosophila brain are critical for memory. Curr Biol 21: 848-854. PubMed ID: 21530256

  • Thomas Turner Department of Ecology, Evolution, and Marine Biology - University of California, Santa Barbara
    Shahandeh, M. P., Brock, C. and Turner, T. L. (2020). Light dependent courtship behavior in Drosophila simulans and D. melanogaster. PeerJ 8: e9499. PubMed ID: 32742789

    Shahandeh, M. P. and Turner, T. L. (2020). The complex genetic architecture of male mate choice evolution between Drosophila species. Heredity (Edinb). PubMed ID: 32203250

    Pischedda, A., Shahandeh, M. P. and Turner, T. L. (2019). The loci of behavioral evolution: evidence that Fas2 and tilB underlie differences in pupation site choice behavior between Drosophila melanogaster and D. simulans. Mol Biol Evol. PubMed ID: 31774527

    Shahandeh, M. P., Pischedda, A., Rodriguez, J. M. and Turner, T. L. (2019). The genetics of male pheromone preference difference between Drosophila melanogaster and Drosophila simulans. G3 (Bethesda). PubMed ID: 31748379

    Shahandeh, M. P., Pischedda, A. and Turner, T. L. (2017). Male mate choice via cuticular hydrocarbon pheromones drives reproductive isolation between Drosophila species. Evolution 72(1):123-135. PubMed ID: 29098691

    Sumethasorn, M. and Turner, T. L. (2016). Oviposition preferences for ethanol depend on spatial arrangement and differ dramatically among closely related Drosophila species. Biol Open. PubMed ID: 27694106

    Najarro, M. A., Sumethasorn, M., Lamoureux, A. and Turner, T. L. (2015). Choosing mates based on the diet of your ancestors: replication of non-genetic assortative mating in Drosophila melanogaster. PeerJ 3: e1173. PubMed ID: 26339551

    Turner, T. L. (2014). Fine-mapping natural alleles: quantitative complementation to the rescue. Mol Ecol 23: 2377-2382. PubMed ID: 24628660

    Pischedda, A., Shahandeh, M. P., Cochrane, W. G., Cochrane, V. A. and Turner, T. L. (2014). Natural Variation in the Strength and Direction of Male Mating Preferences for Female Pheromones in Drosophila melanogaster. PLoS One 9: e87509. PubMed ID: 24489930

    Turner, T. L. and Miller, P. M. (2012). Investigating natural variation in Drosophila courtship song by the evolve and resequence approach. Genetics 191: 633-642. PubMed ID: 22466043

    Turner, T. L., Stewart, A. D., Fields, A. T., Rice, W. R. and Tarone, A. M. (2011). Population-based resequencing of experimentally evolved populations reveals the genetic basis of body size variation in Drosophila melanogaster. PLoS Genet 7: e1001336. PubMed ID: 21437274

  • Hidenobu Tsujimura Tokyo University of Agriculture and Technology
    Nakano, R., Iwamura, M., Obikawa, A., Togane, Y., Hara, Y., Fukuhara, T., Tomaru, M., Takano-Shimizu, T. and Tsujimura, H. (2019). Cortex glia clear dead young neurons via Drpr/dCed-6/Shark and Crk/Mbc/dCed-12 signaling pathways in the developing Drosophila optic lobe. Dev Biol. PubMed ID: 31063730

    Hara, Y., Sudo, T., Togane, Y., Akagawa, H. and Tsujimura, H. (2018). Cell death in neural precursor cells and neurons before neurite formation prevents the emergence of abnormal neural structures in the Drosophila optic lobe. Dev Biol. PubMed ID: 29447906

    Akagawa, H., Hara, Y., Togane, Y., Iwabuchi, K., Hiraoka, T. and Tsujimura, H. (2015). The role of the effector caspases drICE and dcp-1 for cell death and corpse clearance in the developing optic lobe in Drosophila. Dev Biol. PubMed ID: 26022392

    Hara, Y., Hirai, K., Togane, Y., Akagawa, H., Iwabuchi, K. and Tsujimura, H. (2013). Ecdysone-dependent and ecdysone-independent programmed cell death in the developing optic lobe of Drosophila. Dev Biol 374: 127-141. PubMed ID: 23149076

    Togane, Y., Ayukawa, R., Hara, Y., Akagawa, H., Iwabuchi, K. and Tsujimura, H. (2012). Spatio-temporal pattern of programmed cell death in the developing Drosophila optic lobe. Dev Growth Differ 54: 503-518. PubMed ID: 22587328

  • John Tuthill University of Washington
    Karashchuk, P., Rupp, K. L., Dickinson, E. S., Walling-Bell, S., Sanders, E., Azim, E., Brunton, B. W. and Tuthill, J. C. (2021). Anipose: A toolkit for robust markerless 3D pose estimation. Cell Rep 36(13): 109730. PubMed ID: 34592148

    Chen, C., Agrawal, S., Mark, B., Mamiya, A., Sustar, A., Phelps, J. S., Lee, W. A., Dickson, B. J., Card, G. M. and Tuthill, J. C. (2021). Functional architecture of neural circuits for leg proprioception in Drosophila. Curr Biol. PubMed ID: 34637749

    Phelps, J. S., Hildebrand, D. G. C., Graham, B. J., Kuan, A. T., Thomas, L. A., Nguyen, T. M., Buhmann, J., Azevedo, A. W., Sustar, A., Agrawal, S., Liu, M., Shanny, B. L., Funke, J., Tuthill, J. C. and Lee, W. A. (2021). Reconstruction of motor control circuits in adult Drosophila using automated transmission electron microscopy. Cell. PubMed ID: 33400916

    Agrawal, S., Dickinson, E. S., Sustar, A., Gurung, P., Shepherd, D., Truman, J. W. and Tuthill, J. C. (2020). Central processing of leg proprioception in Drosophila. Elife 9. PubMed ID: 33263281

    Azevedo, A. W., Dickinson, E. S., Gurung, P., Venkatasubramanian, L., Mann, R. S. and Tuthill, J. C. (2020). A size principle for recruitment of Drosophila leg motor neurons. Elife 9. PubMed ID: 32490810
    Yan, C., Wang, F., Peng, Y., Williams, C. R., Jenkins, B., Wildonger, J., Kim, H. J., Perr, J. B., Vaughan, J. C., Kern, M. E., Falvo, M. R., O'Brien, E. T., 3rd, Superfine, R., Tuthill, J. C., Xiang, Y., Rogers, S. L. and Parrish, J. Z. (2018). Microtubule Acetylation Is Required for Mechanosensation in Drosophila. Cell Rep 25(4): 1051-1065 e1056. PubMed ID: 30355484

    Mamiya, A., Gurung, P. and Tuthill, J. C. (2018). Neural Coding of Leg Proprioception in Drosophila. Neuron. PubMed ID: 30293823

    Tuthill, J. C. and Wilson, R. I. (2016). Mechanosensation and Adaptive Motor Control in Insects. Curr Biol 26(20): R1022-R1038. PubMed ID: 27780045

  • 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