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


  • Allan Pack Center for Sleep and Circadian Neurobiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
    Zimmerman, J. E., Chan, M. T., Lenz, O. T., Keenan, B. T., Maislin, G. and Pack, A. I. (2016). Glutamate is a wake active neurotransmitter in Drosophila melanogaster. Sleep. PubMed ID: 27855736

    Gerstner, J. R., Lenz, O., Vanderheyden, W. M., Chan, M. T., Pfeiffenberger, C. and Pack, A. I. (2016). Amyloid-beta induces sleep fragmentation that is rescued by fatty acid binding proteins in Drosophila. J Neurosci Res. PubMed ID: 27320125

    Zimmerman, J. E., Chan, M. T., Jackson, N., Maislin, G. and Pack, A. I. (2012). Genetic background has a major impact on differences in sleep resulting from environmental influences in Drosophila. Sleep 35(4): 545-557. PubMed ID: Rick Padgett Waksman Institute of Microbiology, Rutgers University
    Kane, N. S., Vora, M., Varre, K. J. and Padgett, R. W. (2016). Efficient Screening of CRISPR/Cas9-Induced Events in Drosophila Using a co-CRISPR Strategy. G3 (Bethesda). PubMed ID: 27793971

    Li, Y. and Padgett, R. W. (2012). bantam is required for optic lobe development and glial cell proliferation. PLoS One 7: e32910. PubMed ID: 22412948

  • Andrea Page-McCaw Vanderbilt University School of Medicine, Nashville, TN
    Waghmare, I. and Page-McCaw, A. (2022). Regulation of Wnt distribution and function by Drosophila glypicans. J Cell Sci 135(3). PubMed ID: 35112708

    O'Connor, J. T., Stevens, A. C., Shannon, E. K., Akbar, F. B., LaFever, K. S., Narayanan, N. P., Gailey, C. D., Hutson, M. S. and Page-McCaw, A. (2021). Proteolytic activation of Growth-blocking peptides triggers calcium responses through the GPCR Mthl10 during epithelial wound detection. Dev Cell. PubMed ID: 34273275

    Wang, X., LaFever, K. S., Waghmare, I. and Page-McCaw, A. (2021). Extracellular spreading of Wingless is required for Drosophila oogenesis. PLoS Genet 17(4): e1009469. PubMed ID: 33798197

    Waghmare, I., Wang, X. and Page-McCaw, A. (2020). Dally-like protein sequesters multiple Wnt ligands in the Drosophila germarium. Dev Biol. PubMed ID: 32473955

    Ramos-Lewis, W., LaFever, K. S. and Page-McCaw, A. (2018). A scar-like lesion is apparent in basement membrane after wound repair in vivo. Matrix Biol. PubMed ID: 29981372

    Waghmare, I. and Page-McCaw, A. (2018). Wnt Signaling in Stem Cell Maintenance and Differentiation in the Drosophila Germarium. Genes (Basel) 9(3). PubMed ID: 29495453

    Wang, X. and Page-McCaw, A. (2018). Wnt6 maintains anterior escort cells as an integral component of the germline stem cell niche. Development [Epub ahead of print]. PubMed ID: 29361569

    LaFever, K. S., Wang, X., Page-McCaw, P., Bhave, G. and Page-McCaw, A. (2017). Both Drosophila matrix metalloproteinases have released and membrane-tethered forms but have different substrates. Sci Rep 7: 44560. PubMed ID: 28300207

    Wang, X. and Page-McCaw, A. (2014). A matrix metalloproteinase mediates long-distance attenuation of stem cell proliferation. J Cell Biol 206: 923-936. PubMed ID: 25267296

    Broderick, S., Wang, X., Simms, N. and Page-McCaw, A. (2012). Drosophila Ninjurin A induces nonapoptotic cell death. PLoS One 7: e44567. PubMed ID: 23028562

  • Li-Mei Pai Department of Biochemistry and Molecular Biology, Chang Gung University, Taiwan
    Wang, P. Y., Lin, W. C., Tsai, Y. C., Cheng, M. L., Lin, Y. H., Tseng, S. H., Chakraborty, A. and Pai, L. M. (2015). Regulation of CTP Synthase Filament Formation During DNA Endoreplication in Drosophila. Genetics. PubMed ID: 26482795

    Chou, H. Y., Lin, Y. H., Shiu, G. L., Tang, H. Y., Cheng, M. L., Shiao, M. S. and Pai, L. M. (2014). ADI1, a methionine salvage pathway enzyme, is required for Drosophila fecundity. J Biomed Sci 21: 64. PubMed ID: 25037729

    Tsai, Y. C., Chiang, W., Liou, W., Lee, W. H., Chang, Y. W., Wang, P. Y., Li, Y. C., Tanaka, T., Nakamura, A. and Pai, L. M. (2014). Endophilin B is required for the Drosophila oocyte to endocytose yolk downstream of Oskar. Development 141: 563-573. PubMed ID: 24401369

  • Isabel Palacios Department of Zoology, University of Cambridge
    Santa-Cruz Mateos, C., Valencia-Exposito, A., Palacios, I. M. and Martin-Bermudo, M. D. (2020). Integrins regulate epithelial cell shape by controlling the architecture and mechanical properties of basal actomyosin networks. PLoS Genet 16(6): e1008717. PubMed ID: 32479493

    Drechsler, M., Lang, L. F., Al-Khatib, L., Dirks, H., Burger, M., Schonlieb, C. B. and Palacios, I. M. (2020). Optical flow analysis reveals that Kinesin-mediated advection impacts on the orientation of microtubules in the Drosophila oocyte. Mol Biol Cell: mbcE19080440. PubMed ID: 32267197

    Francis, D., Chanana, B., Fernandez, B., Gordon, B., Mak, T. and Palacios, I. M. (2019). YAP/Yorkie in the germline modulates the age-related decline of germline stem cells and niche cells. PLoS One 14(4): e0213327. PubMed ID: 30943201

    Drechsler, M., Giavazzi, F., Cerbino, R. and Palacios, I. M. (2017). Active diffusion and advection in Drosophila oocytes result from the interplay of actin and microtubules. Nat Commun 8(1): 1520. PubMed ID: 29142223

    Ng, B. F., Selvaraj, G. K., Mateos, C. S., Grosheva, I., Alvarez-Garcia, I., Martin-Bermudo, M. D. and Palacios, I. M. (2016). Alpha-Spectrin and integrins act together to regulate actomyosin and columnarization, and to maintain a mono-layered follicular epithelium. Development. PubMed ID: 26952981

    Gomez-Lamarca, M. J., Cobreros-Reguera, L., Ibanez-Jimenez, B., Palacios, I. M. and Martin-Bermudo, M. D. (2014). Integrins regulate epithelial cell differentiation by modulating Notch activity. J Cell Sci. PubMed ID: 25179603

    Saavedra, P., Vincent, J. P., Palacios, I. M., Lawrence, P. A. and Casal, J. (2014). Plasticity of both planar cell polarity and cell identity during the development of Drosophila. Elife 3: e01569. PubMed ID: 24520160

    Williams, L. S., Ganguly, S., Loiseau, P., Ng, B. F. and Palacios, I. M. (2013). The auto-inhibitory domain and ATP-independent microtubule-binding region of Kinesin heavy chain are major functional domains for transport in the Drosophila germline. Development. PubMed ID: 24257625

    Ganguly, S., Williams, L. S., Palacios, I. M. and Goldstein, R. E. (2012). Cytoplasmic streaming in Drosophila oocytes varies with kinesin activity and correlates with the microtubule cytoskeleton architecture. Proc Natl Acad Sci U S A 109: 15109-15114. PubMed ID: 22949706

    Avery, P., Vicente-Crespo, M., Francis, D., Nashchekina, O., Alonso, C. R. and Palacios, I. M. (2011). Drosophila Upf1 and Upf2 loss of function inhibits cell growth and causes animal death in a Upf3-independent manner. RNA 17: 624-638. PubMed ID: 21317294

  • Michael Palladino Pharmacology & Chemical Biology, University of Pittsburgh
    Hrizo, S. L., Eicher, S. L., Myers, T. D., McGrath, I., Wodrich, A. P. K., Venkatesh, H., Manjooran, D., Swoger, S., Gagnon, K., Bruskin, M., Lebedev, M. V., Zheng, S., Vitantonio, A., Kim, S., Lamb, Z. J., Vogt, A., Ruzhnikov, M. R. Z. and Palladino, M. J. (2021). Identification of protein quality control regulators using a Drosophila model of TPI deficiency. Neurobiol Dis 152: 105299. PubMed ID: 33600953

    Taverner, A. M., Yang, L., Barile, Z. J., Lin, B., Peng, J., Pinharanda, A. P., Rao, A. S., Roland, B. P., Talsma, A. D., Wei, D., Petschenka, G., Palladino, M. J. and Andolfatto, P. (2019). Adaptive substitutions underlying cardiac glycoside insensitivity in insects exhibit epistasis in vivo. Elife 8. PubMed ID: 31453806

    Thomas, R. E., Vincow, E. S., Merrihew, G. E., MacCoss, M. J., Davis, M. Y. and Pallanck, L. J. (2018). Glucocerebrosidase deficiency promotes protein aggregation through dysregulation of extracellular vesicles. PLoS Genet 14(9): e1007694. PubMed ID: 30256786

    Drombosky, K. W., Rode, S., Kodali, R., Jacob, T. C., Palladino, M. J. and Wetzel, R. (2018). Mutational analysis implicates the amyloid fibril as the toxic entity in Huntington's disease. Neurobiol Dis. PubMed ID: 30171891

    Fogle, K.J., Hertzler, J.I., Shon, J.H. and Palladino, M.J. (2016). The ATP-sensitive K channel is seizure protective and required for effective dietary therapy in a model of mitochondrial encephalomyopathy. J Neurogenet [Epub ahead of print]. PubMed ID: 27868454

    Talsma, A. D., Chaves, J. F., LaMonaca, A., Wieczorek, E. D. and Palladino, M. J. (2014). Genome-wide screen for modifiers of Na (+) /K (+) ATPase alleles identifies critical genetic loci. Mol Brain 7: 89. PubMed ID: 25476251

    Robert, V. J., Garvis, S. and Palladino, F. (2015). Repression of somatic cell fate in the germline. Cell Mol Life Sci [Epub ahead of print]. PubMed ID: 26043973

    Roland, B. P., Amrich, C. G., Kammerer, C. J., Stuchul, K. A., Larsen, S. B., Rode, S., Aslam, A. A., Heroux, A., Wetzel, R., VanDemark, A. P. and Palladino, M. J. (2014). Triosephosphate isomerase I170V alters catalytic site, enhances stability and induces pathology in a Drosophila model of TPI deficiency. Biochim Biophys Acta 1852: 61-69. PubMed ID: 25463631

    Roland, B. P., Stuchul, K. A., Larsen, S. B., Amrich, C. G., Vandemark, A. P., Celotto, A. M. and Palladino, M. J. (2013). Evidence of a triosephosphate isomerase non-catalytic function critical to behavior and longevity. J Cell Sci. PubMed ID: 23641070

  • Leo Pallanck Department of Genome Sciences, University of Washington School of Medicine Seattle
    Jewett, K. A., Thomas, R. E., Phan, C. Q., Lin, B., Milstein, G., Yu, S., Bettcher, L. F., Neto, F. C., Djukovic, D., Raftery, D., Pallanck, L. J. and Davis, M. Y. (2021). Glucocerebrosidase reduces the spread of protein aggregation in a Drosophila melanogaster model of neurodegeneration by regulating proteins trafficked by extracellular vesicles. PLoS Genet 17(2): e1008859. PubMed ID: 33539341

    Vincow, E. S., Thomas, R. E., Merrihew, G. E., MacCoss, M. J. and Pallanck, L. J. (2021). Slowed protein turnover in aging Drosophila reflects a shift in cellular priorities. J Gerontol A Biol Sci Med Sci. PubMed ID: 33453098

    Pareek, G. and Pallanck, L. J. (2020). Inactivation of the mitochondrial protease Afg3l2 results in severely diminished respiratory chain activity and widespread defects in mitochondrial gene expression. PLoS Genet 16(10): e1009118. PubMed ID: 33075064

    Vincow, E. S., Thomas, R. E., Merrihew, G. E., Shulman, N. J., Bammler, T. K., MacDonald, J. W., MacCoss, M. J. and Pallanck, L. J. (2019). Autophagy accounts for approximately one-third of mitochondrial protein turnover and is protein selective. Autophagy: 1-14. PubMed ID: 30865561

    Pareek, G., Thomas, R. E., Vincow, E. S., Morris, D. R. and Pallanck, L. J. (2018). Lon protease inactivation in Drosophila causes unfolded protein stress and inhibition of mitochondrial translation. Cell Death Discov 5: 51. PubMed ID: 30374414

    Burman, J. L., Itsara, L. S., Kayser, E. B., Suthammarak, W., Wang, A. M., Kaeberlein, M., Sedensky, M. M., Morgan, P. G. and Pallanck, L. J. (2014). A Drosophila model of mitochondrial disease caused by a complex I mutation that uncouples proton pumping from electron transfer. Dis Model Mech. PubMed ID: 25085991

    Thomas, R. E., Andrews, L. A., Burman, J. L., Lin, W. Y. and Pallanck, L. J. (2014). PINK1-Parkin Pathway Activity Is Regulated by Degradation of PINK1 in the Mitochondrial Matrix. PLoS Genet 10: e1004279. PubMed ID: 24874806

    Vincow, E. S., Merrihew, G., Thomas, R. E., Shulman, N. J., Beyer, R. P., Maccoss, M. J., Pallanck, L. J. (2013) The PINK1-Parkin pathway promotes both mitophagy and selective respiratory chain turnover in vivo. Proc Natl Acad Sci U S A. PubMed ID: 23509287

    Burman, J. L., Yu, S., Poole, A. C., Decal, R. B. and Pallanck, L. (2012). Analysis of neural subtypes reveals selective mitochondrial dysfunction in dopaminergic neurons from parkin mutants. Proc Natl Acad Sci U S A 109: 10438-10443. PubMed ID: 22691499

    Chakrabarti, L., Zahra, R., Jackson, S. M., Kazemi-Esfarjani, P., Sopher, B. L., Mason, A. G., Toneff, T., Ryu, S., Shaffer, S., Kansy, J. W., Eng, J., Merrihew, G., MacCoss, M. J., Murphy, A., Goodlett, D. R., Hook, V., Bennett, C. L., Pallanck, L. J. and La Spada, A. R. (2010). Mitochondrial dysfunction in NnaD mutant flies and Purkinje cell degeneration mice reveals a role for Nna proteins in neuronal bioenergetics. Neuron 66: 835-847. PubMed ID: 20620870

  • Ruth Palmer Department of Molecular Biology, Umeå University, Sweden
    Mendoza-Garcia, P., Basu, S., Sukumar, S. K., Arefin, B., Wolfstetter, G., Anthonydhason, V., Molander, L., Uçkun, E., Lindehell, H., Lebrero-Fernandez, C., Larsson, J., Larsson, E., Bemark, M. and Palmer, R. H. (2021). DamID transcriptional profiling identifies the Snail/Scratch transcription factor Kahuli as an Alk target in the Drosophila visceral mesoderm. Development 148(23). PubMed ID: 34905617

    Uckun, E., Wolfstetter, G., Anthonydhason, V., Kumar Sukumar, S., Umapathy, G., Molander, L., Fuchs, J. and Palmer, R. H. (2021). In vivo profiling of the Alk proximitome in the developing Drosophila brain. J Mol Biol: 167282. PubMed ID: 34624297

    Wolfstetter, G., Pfeifer, K., Backman, M., Masudi, T. A., Mendoza-García, P., Chen, S., Sonnenberg, H., Sukumar, S. K., Uckun, E., Varshney, G. K., Uv, A. and Palmer, R. H. (2020). Identification of the Wallenda JNKKK as an Alk suppressor reveals increased competitiveness of Alk-expressing cells. Sci Rep 10(1): 14954. PubMed ID: 32917927

    Wolfstetter, G., Pfeifer, K., van Dijk, J. R., Hugosson, F., Lu, X. and Palmer, R. H. (2017). The scaffolding protein Cnk binds to the receptor tyrosine kinase Alk to promote visceral founder cell specification in Drosophila. Sci Signal 10(502). PubMed ID: 29066538

    Mendoza-Garcia, P., Hugosson, F., Fallah, M., Higgins, M. L., Iwasaki, Y., Pfeifer, K., Wolfstetter, G., Varshney, G., Popichenko, D., Gergen, J. P., Hens, K., Deplancke, B. and Palmer, R. H. (2017). The Zic family homologue Odd-paired regulates Alk expression in Drosophila. PLoS Genet 13(4): e1006617. PubMed ID: 28369060

    Yamazaki, Y., Palmer, L., Alexandre, C., Kakugawa, S., Beckett, K., Gaugue, I., Palmer, R.H. and Vincent, J.P. (2016). Godzilla-dependent transcytosis promotes Wingless signalling in Drosophila wing imaginal discs. Nat Cell Biol [Epub ahead of print]. PubMed ID: 26974662

    Hugosson, F., Sjogren, C., Birve, A., Hedlund, L., Eriksson, T. and Palmer, R. H. (2014). The Drosophila Midkine/Pleiotrophin homologues Miple1 and Miple2 affect adult lifespan but are dispensable for Alk signaling during embryonic gut formation. PLoS One 9: e112250. PubMed ID: 25380037

    Popichenko, D., Hugosson, F., Sjogren, C., Dogru, M., Yamazaki, Y., Wolfstetter, G., Schonherr, C., Fallah, M., Hallberg, B., Nguyen, H. and Palmer, R. H. (2013). Jeb/Alk signalling regulates the Lame duck GLI family transcription factor in the Drosophila visceral mesoderm. Development. PubMed ID: 23824577

    Yamazaki, Y., Schonherr, C., Varshney, G. K., Dogru, M., Hallberg, B., Palmer, R. H. (2013) Goliath family E3 ligases regulate the recycling endosome pathway via VAMP3 ubiquitylation. EMBO J. PubMed ID: 23353890

    Gouzi, J. Y., Moressis, A., Walker, J. A., Apostolopoulou, A. A., Palmer, R. H., Bernards, A. and Skoulakis, E. M. (2011). The receptor tyrosine kinase Alk controls neurofibromin functions in Drosophila growth and learning. PLoS Genet 7: e1002281. PubMed ID: 21949657

    Schonherr, C., Ruuth, K., Yamazaki, Y., Eriksson, T., Christensen, J., Palmer, R. H. and Hallberg, B. (2011). Activating ALK mutations found in neuroblastoma are inhibited by Crizotinib and NVP-TAE684. Biochem J 440: 405-413. PubMed ID: 21838707

  • Duojia Pan Department of Molecular Biology & Genetics, Johns Hopkins School of Medicine
    Deng, H., Yang, L., Wen, P., Lei, H., Blount, P. and Pan, D. (2020). Spectrin couples cell shape, cortical tension, and Hippo signaling in retinal epithelial morphogenesis. J Cell Biol 219(4). PubMed ID: 32328630

    Guo, P., Lee, C. H., Lei, H., Zheng, Y., Pulgar Prieto, K. D. and Pan, D. (2019). Nerfin-1 represses transcriptional output of Hippo signaling in cell competition. Elife 8. PubMed ID: 30901309

    Yu, J. and Pan, D. (2018). Validating upstream regulators of Yorkie activity in Hippo signaling through scalloped-based genetic epistasis. Development 145(4). PubMed ID: 29467233

    Zheng, Y., Liu, B., Wang, L., Lei, H., Pulgar Prieto, K. D. and Pan, D. (2017). Homeostatic control of Hpo/MST kinase activity through autophosphorylation-dependent recruitment of the STRIPAK PP2A phosphatase complex. Cell Rep 21(12): 3612-3623. PubMed ID: 29262338

    Liu, B., Zheng, Y., Yin, F., Yu, J., Silverman, N. and Pan, D. (2016). Toll receptor-mediated Hippo signaling controls innate immunity in Drosophila. Cell 164: 406-419. PubMed ID: 26824654

    Liu, B., Zheng, Y., Yin, F., Yu, J., Silverman, N. and Pan, D. (2016). Toll receptor-mediated Hippo signaling controls innate immunity in Drosophila. Cell 164: 406-419. PubMed ID: 26824654

    Deng, H., Wang, W., Yu, J., Zheng, Y., Qing, Y. and Pan, D. (2015). Spectrin regulates Hippo signaling by modulating cortical actomyosin activity. Elife 4: e06567. PubMed ID: 25826608

    Zheng, Y., Wang, W., Liu, B., Deng, H., Uster, E. and Pan, D. (2015). Identification of Happyhour/MAP4K as Alternative Hpo/Mst-like Kinases in the Hippo Kinase Cascade. Dev Cell [Epub ahead of print]. PubMed ID: 26364751

    Qing, Y., Yin, F., Wang, W., Zheng, Y., Guo, P., Schozer, F., Deng, H. and Pan, D. (2014). The Hippo effector Yorkie activates transcription by interacting with a histone methyltransferase complex through Ncoa6. Elife: e02564. PubMed ID: 25027438

    Yin, F., Yu, J., Zheng, Y., Chen, Q., Zhang, N. and Pan, D. (2013). Spatial Organization of Hippo Signaling at the Plasma Membrane Mediated by the Tumor Suppressor Merlin/NF2. Cell. PubMed ID: 24012335

  • Vlad Panin Department of Biochemistry and Biophysics, Texas A&M University
    Chandel, I., Baker, R., Nakamura, N. and Panin, V. (2019). Live imaging and analysis of muscle contractions in Drosophila embryo. J Vis Exp(149). PubMed ID: 31355800

    Baker, R., Nakamura, N., Chandel, I., Howell, B., Lyalin, D. and Panin, V. M. (2017). Protein O-mannosyltransferases affect sensory axon wiring and dynamic chirality of body posture in the Drosophila embryo. J Neurosci. PubMed ID: 29167399

    Scott, H. and Panin, V. M. (2014). N-glycosylation in regulation of the nervous system. Adv Neurobiol 9: 367-394. PubMed ID: 25151388

    Scott, H. and Panin, V. M. (2014). The role of protein N-glycosylation in neural transmission. Glycobiology [Epub ahead of print]. PubMed ID: 24643084

    Nakamura, M., Pandey, D. and Panin, V. M. (2012). Genetic Interactions Between Drosophila sialyltransferase and beta1,4-N-acetylgalactosaminyltransferase-A Genes Indicate Their Involvement in the Same Pathway. G3 (Bethesda) 2: 653-656. PubMed ID: 22690374

    Repnikova, E., Koles, K., Nakamura, M., Pitts, J., Li, H., Ambavane, A., Zoran, M. J. and Panin, V. M. (2010). Sialyltransferase regulates nervous system function in Drosophila. J Neurosci 30: 6466-6476. PubMed ID: 20445073

    Nakamura, N., Lyalin, D. and Panin, V. M. (2010). Protein O-mannosylation in animal development and physiology: from human disorders to Drosophila phenotypes. Semin Cell Dev Biol 21: 622-630. PubMed ID: 20362685

  • Udai Pandey Louisiana State University, School of Medicine, New Orleans
    Scaramuzzino, C., Monaghan, J., Milioto, C., Lanson, N. A., Jr., Maltare, A., Aggarwal, T., Casci, I., Fackelmayer, F. O., Pennuto, M., Pandey, U. B. (2013) Protein Arginine Methyltransferase 1 and 8 Interact with FUS to Modify Its Sub-Cellular Distribution and Toxicity In Vitro and In Vivo. PLoS One 8: e61576. PubMed ID: 23620769

    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

    Pandey, U. B. and Nichols, C. D. (2011). Human disease models in Drosophila melanogaster and the role of the fly in therapeutic drug discovery. Pharmacol Rev 63: 411-436. PubMed ID: 21415126

  • Tibor Pankotai Department of Pathology University of Szeged, Hungary
    Pahi, Z. G., Kovacs, L., Szucs, D., Borsos, B. N., Deak, P. and Pankotai, T. (2022). Usp5, Usp34, and Otu1 deubiquitylases mediate DNA repair in Drosophila melanogaster. Sci Rep 12(1): 5870. PubMed ID: 35393473

    Pahi, Z., Borsos, B. N., Vedelek, B., Shidlovskii, Y. V., Georgieva, S. G., Boros, I. M. and Pankotai, T. (2017). TAF10 and TAF10b partially redundant roles during Drosophila melanogaster morphogenesis. Transcription 8(5): 297-306. PubMed ID: 28841365

    Pahi, Z., Kiss, Z., Komonyi, O., Borsos, B. N., Tora, L., Boros, I. M. and Pankotai, T. (2015). dTAF10- and dTAF10b-Containing Complexes Are Required for Ecdysone-Driven Larval-Pupal Morphogenesis in Drosophila melanogaster. PLoS One 10(11): e0142226. PubMed ID: 26556600

    Borsos, B. N., Pankotai, T., Kovacs, D., Popescu, C., Pahi, Z. and Boros, I. M. (2015). Acetylations of Ftz-F1 and histone H4K5 are required for the fine-tuning of ecdysone biosynthesis during Drosophila metamorphosis. Dev Biol 404(1): 80-87. PubMed ID: 25959239

  • Michael Pankratz Development, Genetics & Molecular Physiology Unit, Laboratory of Molecular Brain Physiology and Behavior, Universität Bonn
    Kottmeier, R., Bittern, J., Schoofs, A., Scheiwe, F., Matzat, T., Pankratz, M. and Klambt, C. (2020). Wrapping glia regulates neuronal signaling speed and precision in the peripheral nervous system of Drosophila. Nat Commun 11(1): 4491. PubMed ID: 32901033

    Miroschnikow, A., Schlegel, P. and Pankratz, M. J. (2020). Making Feeding Decisions in the Drosophila Nervous System. Curr Biol 30(14): R831-r840. PubMed ID: 32693083

    Imura, E., Shimada-Niwa, Y., Nishimura, T., Huckesfeld, S., Schlegel, P., Ohhara, Y., Kondo, S., Tanimoto, H., Cardona, A., Pankratz, M. J. and Niwa, R. (2020). The Corazonin-PTTH Neuronal Axis Controls Systemic Body Growth by Regulating Basal Ecdysteroid Biosynthesis in Drosophila melanogaster. Curr Biol 30(11): 2156-2165.e2155. PubMed ID: 32386525

    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

    Schoofs, A., Huckesfeld, S. and Pankratz, M. J. (2017). Serotonergic network in the subesophageal zone modulates the motor pattern for food intake in Drosophila. J Insect Physiol [Epub ahead of print]. PubMed ID: 28735009

    Surendran, S., Huckesfeld, S., Waschle, B. and Pankratz, M. J. (2017). Pathogen induced food evasion behavior in Drosophila larvae. J Exp Biol. PubMed ID: 28254879

    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

    Hückesfeld, S., Peters, M. and Pankratz, M.J. (2016). Central relay of bitter taste to the protocerebrum by peptidergic interneurons in the Drosophila brain. Nat Commun 7: 12796. PubMed ID: 27619503

    Huckesfeld, S., Schoofs, A., Schlegel, P., Miroschnikow, A. and Pankratz, M. J. (2015). Localization of motor neurons and central pattern generators for motor patterns underlying feeding behavior in Drosophila larvae. PLoS One 10: e0135011. PubMed ID: 26252658

    Alekseyenko, O. V., Chan, Y. B., Fernandez Mde, L., Bulow, T., Pankratz, M. J. and Kravitz, E. A. (2014). Single serotonergic neurons that modulate aggression in Drosophila. Curr Biol 24: 2700-2707. PubMed ID: 25447998

  • Fani Papagiannouli University of Heidelberg
    Schardt, L., Ander, J. J., Lohmann, I. and Papagiannouli, F. (2015). Stage-specific control of niche positioning and integrity in the Drosophila testis. Mech Dev. PubMed ID: 26226434

    Papagiannouli, F. (2013) The internal structure of embryonic gonads and testis development in Drosophila melanogaster requires scrib, lgl and dlg activity in the soma. Int J Dev Biol 57: 25-34. PubMed ID: 23585349

    Sorge, S., Ha, N., Polychronidou, M., Friedrich, J., Bezdan, D., Kaspar, P., Schaefer, M. H., Ossowski, S., Henz, S. R., Mundorf, J., Ratzer, J., Papagiannouli, F. and Lohmann, I. (2012). The cis-regulatory code of Hox function in Drosophila. EMBO J 31: 3323-3333. PubMed ID: 22781127

    Papagiannouli, F. and Lohmann, I. (2012). Shaping the niche: lessons from the Drosophila testis and other model systems. Biotechnol J 7: 723-736. PubMed ID: 22488937

    Zhai, Z., Ha, N., Papagiannouli, F., Hamacher-Brady, A., Brady, N., Sorge, S., Bezdan, D. and Lohmann, I. (2012). Antagonistic regulation of apoptosis and differentiation by the Cut transcription factor represents a tumor-suppressing mechanism in Drosophila. PLoS Genet 8: e1002582. PubMed ID: 22438831

  • Dmitri Papatsenko Skoltech: Skolkovo Institute of Science and Technology
    Kim, Y., Iagovitina, A., Ishihara, K., Fitzgerald, K. M., Deplancke, B., Papatsenko, D. and Shvartsman, S. Y. (2013). Context-dependent transcriptional interpretation of mitogen activated protein kinase signaling in the Drosophila embryo. Chaos 23: 025105. PubMed ID: 23822503

    Papatsenko, D. and Levine, M. (2011). The Drosophila gap gene network is composed of two parallel toggle switches. PLoS One 6: e21145. PubMed ID: 21747931

    Papatsenko, D., Levine, M. and Goltsev, Y. (2011). Clusters of temporal discordances reveal distinct embryonic patterning mechanisms in Drosophila and anopheles. PLoS Biol 9: e1000584. PubMed ID: 21283609

  • Mary-Lou Pardue Department of Biology, MIT
    Pardue, M. L. and Debaryshe, P. (2011). Adapting to life at the end of the line: How Drosophila telomeric retrotransposons cope with their job. Mob Genet Elements 1: 128-134. PubMed ID: 22016861

    Pardue, M. L. and DeBaryshe, P. G. (2011). Retrotransposons that maintain chromosome ends. Proc Natl Acad Sci U S A 108: 20317-20324. PubMed ID: 21821789

    George, J. A., Traverse, K. L., DeBaryshe, P. G., Kelley, K. J. and Pardue, M. L. (2010). Evolution of diverse mechanisms for protecting chromosome ends by Drosophila TART telomere retrotransposons. Proc Natl Acad Sci U S A 107: 21052-21057. PubMed ID: 21088221

  • Nuria Paricio Departamento de Genetica, Facultad CC Biologicas, Universidad de Valencia, Burjasot, Spain
    Solana-Manrique, C., Sanz, F. J., Torregrosa, I., Palomino-Schatzlein, M., Hernandez-Oliver, C., Pineda-Lucena, A. and Paricio, N. (2022). Metabolic Alterations in a Drosophila Model of Parkinson's Disease Based on DJ-1 Deficiency. Cells 11(3). PubMed ID: 35159141

    Solana-Manrique, C., Sanz, F. J., Ripolles, E., Bano, M. C., Torres, J., Munoz-Soriano, V. and Paricio, N. (2020). Enhanced activity of glycolytic enzymes in Drosophila and human cell models of Parkinson's disease based on DJ-1 deficiency. Free Radic Biol Med. PubMed ID: 32726690

    Munoz-Soriano, V., Belacortu, Y., Sanz, F. J., Solana-Manrique, C., Dillon, L., Suay-Corredera, C., Ruiz-Romero, M., Corominas, M. and Paricio, N. (2018). Cbt modulates Foxo activation by positively regulating insulin signaling in Drosophila embryos. Biochim Biophys Acta. PubMed ID: 30055320

    Sanz, F. J., Solana-Manrique, C., Munoz-Soriano, V., Calap-Quintana, P., Molto, M. D. and Paricio, N. (2017). Identification of potential therapeutic compounds for Parkinson's disease using Drosophila and human cell models. Free Radic Biol Med 108: 683-691. PubMed ID: 28455141

    Munoz-Soriano, V., Santos, D., Durupt, F. C., Casani, S. and Paricio, N. (2015). Scabrous overexpression in the eye affects R3/R4 cell fate specification and inhibits Notch signaling. Dev Dyn. PubMed ID: 26505171

    Munoz-Soriano, V., Belacortu, Y. and Paricio, N. (2012). Planar cell polarity signaling in collective cell movements during morphogenesis and disease. Curr Genomics 13: 609-622. PubMed ID: 23730201

    Casani, S., Gomez-Pastor, R., Matallana, E., Paricio, N. (2013) Antioxidant compounds supplementation prevents oxidative damage in a Drosophila model of Parkinson's disease. Free Radic Biol Med. PubMed ID: 23548634
    Munoz-Soriano, V., Ruiz, C., Perez-Alonso, M., Mlodzik, M., Paricio, N. (2013) Nemo regulates cell dynamics and represses the expression of miple, a midkine/pleiotrophin cytokine, during ommatidial rotation. Dev Biol. PubMed ID: 23428616

    Munoz-Soriano, V., Nieto-Arellano, R. and Paricio, N. (2012). Septin 4, the drosophila ortholog of human CDCrel-1, accumulates in parkin mutant brains and is functionally related to the Nedd4 E3 ubiquitin ligase. J Mol Neurosci 48: 136-143. PubMed ID: 22562816

    Belacortu, Y., Weiss, R., Kadener, S. and Paricio, N. (2012). Transcriptional activity and nuclear localization of Cabut, the Drosophila ortholog of vertebrate TGF-beta-inducible early-response gene (TIEG) proteins. PLoS One 7: e32004. PubMed ID: 22359651

  • Jae Park Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville
    Yoo, S., Nair, S., Kim, H. J., Kim, Y., Lee, C., Lee, G. and Park, J. H. (2020). Knock-in mutations of scarecrow, a Drosophila homolog of mammalian Nkx2.1, reveal a novel function required for development of the optic lobe in Drosophila melanogaster. Dev Biol. PubMed ID: 32061586

    Nair, S., Bahn, J. H., Lee, G., Yoo, S. and Park, J. H. (2020). A homeobox transcription factor Scarecrow (SCRO) negatively regulates Pdf neuropeptide expression through binding an identified cis-acting element in Drosophila melanogaster. Mol Neurobiol. PubMed ID: 31950355

    Wang, Z., Lee, G., Vuong, R. and Park, J. H. (2019). Two-factor specification of apoptosis: TGF-beta signaling acts cooperatively with ecdysone signaling to induce cell- and stage-specific apoptosis of larval neurons during metamorphosis in Drosophila melanogaster. Apoptosis 24(11-12): 972-989. PubMed ID: 31641960

    Lee, G., Sehgal, R., Wang, Z. and Park, J. H. (2019). Ultraspiracle-independent anti-apoptotic function of ecdysone receptors is required for the survival of larval peptidergic neurons via suppression of grim expression in Drosophila melanogaster. Apoptosis. PubMed ID: 30637539

    Lee, G., Kim, J., Kim, Y., Yoo, S. and Park, J. H. (2018). Identifying and monitoring neurons that undergo metamorphosis-regulated cell death (metamorphoptosis) by a neuron-specific caspase sensor (Casor) in Drosophila melanogaster. Apoptosis 23(1): 41-53. PubMed ID: 29224041

    Kim, G., Lee, Y., Lee, G., Cho, Y., Lee, Y., Jang, Y., Paik, D. and Park, J. (2014). Overexpression of malic enzyme in the larval stage extends Drosophila lifespan. Biochem Biophys Res Commun. PubMed ID: 25511696

    Sha, K., Choi, S. H., Im, J., Lee, G. G., Loeffler, F. and Park, J. H. (2014). Regulation of Ethanol-Related Behavior and Ethanol Metabolism by the Corazonin Neurons and Corazonin Receptor in Drosophila melanogaster. PLoS One 9: e87062. PubMed ID: 24489834

    Lee, G. G., Kikuno, K., Nair, S. and Park, J. H. (2013). Mechanisms of post-ecdysis-associated programmed cell death of peptidergic neurons in drosophila melanogaster. J Comp Neurol. PubMed ID: 23784845

    Lee, G., Sehgal, R., Wang, Z., Nair, S., Kikuno, K., Chen, C. H., Hay, B., Park, J. H. (2013) Essential role of grim-led programmed cell death for the establishment of corazonin-producing peptidergic nervous system during embryogenesis and metamorphosis in Drosophila melanogaster. Biol Open 2: 283-294. PubMed ID: 23519152

    Lee, G., Kikuno, K., Bahn, J. H., Kim, K. M. and Park, J. H. (2013). Dopamine D2 Receptor as a Cellular Component Controlling Nocturnal Hyperactivities in Drosophila melanogaster. Chronobiol Int. PubMed ID: 23286280

  • Ravi Parkash Department of Genetics, M.D.University, Rohtak, India
    Lambhod, C., Pathak, A., Munjal, A. K. and Parkash, R. (2017). Tropical Drosophila ananassae of wet-dry seasons show cross resistance to heat, drought and starvation. Biol Open 6(11): 1698-1706. PubMed ID: 29141954

    Kalra, B. and Parkash, R. (2014). Trade-off of ovarian lipids and total body lipids for fecundity and starvation resistance in tropical populations of Drosophila melanogaster. J Evol Biol. PubMed ID: 25223796

    Chahal, J., Kataria, S. K., Parkash, R. (2013) Invasion and adaptation of a warm adapted species to montane localities: effect of acclimation potential. J Exp Biol. PubMed ID: 23348937

    Parkash, R., Ramniwas, S., Kajla, B. and Aggarwal, D. D. (2012). Divergence of desiccation-related traits in two Drosophila species of the takahashii subgroup from the western Himalayas. J Exp Biol 215: 2181-2191. PubMed ID: 22675178

    Parkash, R., Aggarwal, D. D., Ranga, P. and Singh, D. (2012). Divergent strategies for adaptation to desiccation stress in two Drosophila species of immigrans group. J Comp Physiol B 182: 751-769. PubMed ID: 22407357

  • Susan Parkhurst Fred Hutchinson Cancer Research Institute, Seattle
    Nakamura, M., Verboon, J. M., Allen, T. E., Abreu-Blanco, M. T., Liu, R., Dominguez, A. N. M., Delrow, J. J. and Parkhurst, S. M. (2020). Autocrine insulin pathway signaling regulates actin dynamics in cell wound repair. PLoS Genet 16(12): e1009186. PubMed ID: 33306674

    Nakamura, M., Verboon, J. M., Prentiss, C. L. and Parkhurst, S. M. (2020). The kinesin-like protein Pavarotti functions noncanonically to regulate actin dynamics. J Cell Biol 219(9). PubMed ID: 32673395

    Verboon, J. M., Decker, J. R., Nakamura, M. and Parkhurst, S. M. (2018). Wash exhibits context dependent phenotypes and, along with the WASH Regulatory Complex, regulates Drosophila oogenesis. J Cell Sci. PubMed ID: 29549166

    Nakamura, M., Verboon, J. M. and Parkhurst, S. M. (2017). Prepatterning by RhoGEFs governs Rho GTPase spatiotemporal dynamics during wound repair. J Cell Biol [Epub ahead of print]. PubMed ID: 28923977

    Pitmon, E., Stephens, G., Parkhurst, S. J., Wolf, F. W., Kehne, G., Taylor, M. and Lebestky, T. (2016). The D1 Family Dopamine Receptor, DopR, potentiates hindleg grooming behavior in Drosophila. Genes Brain Behav [Epub ahead of print]. PubMed ID: 26749475

    Verboon, J. M. and Parkhurst, S. M. (2015). Rho family GTPases bring a familiar ring to cell wound repair. Small GTPases 6: 1-7. PubMed ID: 25862160

    Verboon, J. M. and Parkhurst, S. M. (2015). Rho family GTPase functions in Drosophila epithelial wound repair. Small GTPases 6: 33-40. PubMed ID: 25862164

    Verboon, J.M., Rincon-Arano, H., Werwie, T.R., Delrow, J.J., Scalzo, D., Nandakumar, V., Groudine, M. and Parkhurst, S.M. (2015). Wash interacts with Lamin and affects global nuclear organization. Curr Biol [Epub ahead of print]. PubMed ID: 25754639

    Abreu-Blanco, M. T., Verboon, J. M. and Parkhurst, S. M. (2013). Coordination of Rho Family GTPase Activities to Orchestrate Cytoskeleton Responses during Cell Wound Repair. Curr Biol. PubMed ID: 24388847

    Rincon-Arano, H., Halow, J., Delrow, J. J., Parkhurst, S. M. and Groudine, M. (2012). UpSET Recruits HDAC Complexes and Restricts Chromatin Accessibility and Acetylation at Promoter Regions. Cell 151: 1214-1228. PubMed ID: 23177352

  • Marie-Laure Parmentier Institut de Génomique Fonction, Montpellier, France
    Talmat-Amar, Y., Arribat, Y. and Parmentier, M. L. (2018). Vesicular Axonal Transport is Modified In Vivo by Tau Deletion or Overexpression in Drosophila. Int J Mol Sci 19(3). PubMed ID: 29509687

    Devambez, I., van Dijk, J., Benlefki, S., Layalle, S., Grau, Y., Rogowski, K., Parmentier, M. L. and Soustelle, L. (2017). Identification of DmTTLL5 as a Major Tubulin Glutamylase in the Drosophila Nervous System. Sci Rep 7(1): 16254. PubMed ID: 29176602

    Bouge, A. L. and Parmentier, M. L. (2016). Tau excess impairs mitosis and kinesin-5 function, leading to aneuploidy and cell death. Dis Model Mech [Epub ahead of print]. PubMed ID:

    Lepicard, S., Franco, B., de Bock, F. and Parmentier, M. L. (2014). A presynaptic role of microtubule-associated protein 1/futsch in Drosophila: regulation of active zone number and neurotransmitter release. J Neurosci 34: 6759-6771. PubMed ID: 24828631

    Morel, V., Lepicard, S., A, N. R., Parmentier, M. L. and Schaeffer, L. (2014). Drosophila Nesprin-1 controls glutamate receptor density at neuromuscular junctions. Cell Mol Life Sci. PubMed ID: 24492984

    Devambez, I., Ali Agha, M., Mitri, C., Bockaert, J., Parmentier, M. L., Marion-Poll, F., Grau, Y. and Soustelle, L. (2013). Galphao is required for L-canavanine detection in Drosophila. PLoS One 8: e63484. PubMed ID: 23671680

    Redt-Clouet, C., Trannoy, S., Boulanger, A., Tokmatcheva, E., Savvateeva-Popova, E., Parmentier, M. L., Preat, T. and Dura, J. M. (2012). Mushroom body neuronal remodelling is necessary for short-term but not for long-term courtship memory in Drosophila. Eur J Neurosci 35: 1684-1691. PubMed ID: 22571719

  • Moshe Parnas Tel Aviv University, Israel
    Rozenfeld, E., Tauber, M., Ben-Chaim, Y. and Parnas, M. (2021). GPCR voltage dependence controls neuronal plasticity and behavior. Nat Commun 12(1): 7252. PubMed ID: 34903750

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

    Rozenfeld, E., Lerner, H. and Parnas, M. (2019). Muscarinic modulation of antennal lobe GABAergic local neurons shapes odor coding and behavior. Cell Rep 29(10): 3253-3265.e3254. PubMed ID: 31801087

    Bielopolski, N., Amin, H., Apostolopoulou, A. A., Rozenfeld, E., Lerner, H., Huetteroth, W., Lin, A. C. and Parnas, M. (2019). Inhibitory muscarinic acetylcholine receptors enhance aversive olfactory learning in adult Drosophila. Elife 8. PubMed ID: 31215865

    Bentzur, A., Shmueli, A., Omesi, L., Ryvkin, J., Knapp, J. M., Parnas, M., Davis, F. P. and Shohat-Ophir, G. (2018). Odorant binding protein 69a connects social interaction to modulation of social responsiveness in Drosophila. PLoS Genet 14(4): e1007328. PubMed ID: 29630598

    Parnas, M., Lin, A. C., Huetteroth, W. and Miesenbock, G. (2013). Odor discrimination in Drosophila: from neural population codes to behavior. Neuron 79(5): 932-944. PubMed ID: 24012006

  • Renato Paro ETH Zürich, Basel, Switzerland
    Akmammedov, A., Geigges, M. and Paro, R. (2019). Bivalency in Drosophila embryos is associated with strong inducibility of Polycomb target genes. Fly (Austin). PubMed ID: 31094269

    Jiang, Y., Seimiya, M., Schlumpf, T. B. and Paro, R. (2018). An intrinsic tumour eviction mechanism in Drosophila mediated by steroid hormone signalling. Nat Commun 9(1): 3293. PubMed ID: 30120247

    Torres, J., Monti, R., Moore, A. L., Seimiya, M., Jiang, Y., Beerenwinkel, N., Beisel, C., Beira, J. V. and Paro, R. (2018). A switch in transcription and cell fate governs the onset of an epigenetically-deregulated tumor in Drosophila. Elife 7. PubMed ID: 29560857

    Pereira, A. and Paro, R. (2017). Pho dynamically interacts with Spt5 to facilitate transcriptional switches at the hsp70 locus. Epigenetics Chromatin 10(1): 57. PubMed ID: 29208012

    Akmammedov, A., Geigges, M. and Paro, R. (2017). Single vector non-leaky gene expression system for Drosophila melanogaster. Sci Rep 7(1): 6899. PubMed ID: 28761084

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

    Comoglio, F., Schlumpf, T., Schmid, V., Rohs, R., Beisel, C. and Paro, R. (2015). High-resolution profiling of Drosophila replication start sites reveals a DNA shape and chromatin signature of metazoan origins. Cell Rep [Epub ahead of print]. PubMed ID: 25921534

    Katsuyama, T., Comoglio, F., Seimiya, M., Cabuy, E. and Paro, R. (2015). During Drosophila disc regeneration, JAK/STAT coordinates cell proliferation with Dilp8-mediated developmental delay. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 25902518

    Sievers, C., Comoglio, F., Seimiya, M., Merdes, G. and Paro, R. (2014). A Deterministic Analysis of Genome Integrity during Neoplastic Growth in Drosophila. PLoS One 9: e87090. PubMed ID: 24516544

    Comoglio, F. and Paro, R. (2014). Combinatorial modeling of chromatin features quantitatively predicts DNA replication timing in Drosophila. PLoS Comput Biol 10: e1003419. PubMed ID: 24465194

  • Zeev Paroush School of Medicine. Hebrew University of Jerusalem
    Kushnir, T., Bar-Cohen, S., Mooshayef, N., Lange, R., Bar-Sinai, A., Rozen, H., Salzberg, A., Engelberg, D. and Paroush, Z. (2019). An activating mutation in ERK causes hyperplastic tumors in a scribble mutant tissue in Drosophila. Genetics. PubMed ID: 31740452

    Kushnir, T., Mezuman, S., Bar-Cohen, S., Lange, R., Paroush, Z. and Helman, A. (2017). Novel interplay between JNK and Egfr signaling in Drosophila dorsal closure. PLoS Genet 13(6): e1006860. PubMed ID: 28628612

    Grossman, R. and Paroush, Z. (2017). High-throughput in vitro identification of direct MAPK/Erk substrates. Methods Mol Biol 1487: 127-135. PubMed ID: 27924563

    Cinnamon, E., Makki, R., Sawala, A., Wickenberg, L. P., Blomquist, G. J., Tittiger, C., Paroush, Z. and Gould, A. P. (2016). Drosophila Spidey/Kar Regulates Oenocyte Growth via PI3-Kinase Signaling. PLoS Genet 12(8): e1006154. PubMed ID: 27500738

    Nir, R., Grossman, R., Paroush, Z. and Volk, T. (2012). Phosphorylation of the Drosophila melanogaster RNA-binding protein HOW by MAPK/ERK enhances its dimerization and activity. PLoS Genet 8(3): e1002632. PubMed ID: 22479211

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

    Jiang, N., Rasmussen, J. P., Clanton, J. A., Rosenberg, M. F., Luedke, K. P., Cronan, M. R., Parker, E. D., Kim, H. J., Vaughan, J. C., Sagasti, A. and Parrish, J. Z. (2019). A conserved morphogenetic mechanism for epidermal ensheathment of nociceptive sensory neurites. Elife 8. PubMed ID: 30855229

    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

    Hoyer, N., Zielke, P., Hu, C., Petersen, M., Sauter, K., Scharrenberg, R., Peng, Y., Kim, C. C., Han, C., Parrish, J. Z. and Soba, P. (2018). Ret and Substrate-Derived TGF-beta Maverick Regulate Space-Filling Dendrite Growth in Drosophila Sensory Neurons. Cell Rep 24(9): 2261-2272.e2265. PubMed ID: 30157422

    Meltzer, S., Yadav, S., Lee, J., Soba, P., Younger, S. H., Jin, P., Zhang, W., Parrish, J., Jan, L. Y. and Jan, Y. N. (2016). Epidermis-derived Semaphorin promotes dendrite self-avoidance by regulating dendrite-substrate adhesion in Drosophila sensory neurons. Neuron 89: 741-755. PubMed ID: 26853303

    Lin, W. Y., Williams, C. R., Yan, C. and Parrish, J. Z. (2016). Functions of the SLC36 transporter Pathetic in growth control. Fly (Austin): 0. PubMed ID: 26735916

    Peng, Y., Lee, J., Rowland, K., Wen, Y., Hua, H., Carlson, N., Lavania, S., Parrish, J. Z. and Kim, M. D. (2015). Regulation of dendrite growth and maintenance by exocytosis. J Cell Sci 128: 4279-4292. PubMed ID: 26483382

    Lin, W. Y., Williams, C., Yan, C., Koledachkina, T., Luedke, K., Dalton, J., Bloomsburg, S., Morrison, N., Duncan, K. E., Kim, C. C. and Parrish, J. Z. (2015). The SLC36 transporter Pathetic is required for extreme dendrite growth in Drosophila sensory neurons. Genes Dev 29: 1120-1135. PubMed ID: 26063572

    Lee, J., Peng, Y., Lin, W. Y. and Parrish, J. Z. (2015). Coordinate control of terminal dendrite patterning and dynamics by the membrane protein Raw. Development 142: 162-173. PubMed ID: 25480915

    Jiang, N., Soba, P., Parker, E., Kim, C. C. and Parrish, J. Z. (2014). The microRNA bantam regulates a developmental transition in epithelial cells that restricts sensory dendrite growth. Development 141: 2657-2668. PubMed ID: 24924190

  • John Parsch Department of Biology II, University of Munich (LMU), Planegg-Martinsried, Germany
    Ramnarine, T. J. S., Grath, S. and Parsch, J. (2021). Natural variation in the transcriptional response of Drosophila melanogaster to oxidative stress. G3 (Bethesda). PubMed ID: 34747443

    Glaser-Schmitt, A., Wittmann, M. J., Ramnarine, T. J. S. and Parsch, J. (2021). Sexual antagonism, temporally fluctuating selection, and variable dominance affect a regulatory polymorphism in Drosophila melanogaster. Mol Biol Evol. PubMed ID: 34289067

    Belyi, A., Argyridou, E. and Parsch, J. (2020). The influence of chromosomal environment on X-linked gene expression in Drosophila melanogaster. Genome Biol Evol. PubMed ID: 33104185

    Ramnarine, T. J. S., Glaser-Schmitt, A., Catalan, A. and Parsch, J. (2019). Population genetic and functional analysis of a cis-regulatory polymorphism in the Drosophila melanogaster Metallothionein A gene. Genes (Basel) 10(2). PubMed ID: 30769915

    Argyridou, E. and Parsch, J. (2018). Regulation of the X chromosome in the germline and soma of Drosophila melanogaster males. Genes (Basel) 9(5). PubMed ID: 29734690

    AGlaser-Schmitt, A. and Parsch, J. (2018). Functional characterization of adaptive variation within a cis-regulatory element influencing Drosophila melanogaster growth. PLoS Biol 16(1): e2004538. PubMed ID: 29324742

    Catalan, A., Glaser-Schmitt, A., Argyridou, E., Duchen, P. and Parsch, J. (2016). An indel polymorphism in the MtnA 3' untranslated region is associated with gene expression variation and Local Adaptation in Drosophila melanogaster. PLoS Genet 12: e1005987. PubMed ID: 27120580

    Huylmans, A.K. and Parsch, J. (2015). Variation in the X:autosome distribution of male-biased genes among Drosophila melanogaster tissues and its relationship with dosage compensation. Genome Biol Evol [Epub ahead of print]. PubMed ID: 26108491

    Huylmans, A. K. and Parsch, J. (2014). Population- and Sex-Biased Gene Expression in the Excretion Organs of Drosophila melanogaster. G3 (Bethesda). PubMed ID: 25246242

    Glaser-Schmitt, A., Catalan, A. and Parsch, J. (2013). Adaptive divergence of a transcriptional enhancer between populations of Drosophila melanogaster. Philos Trans R Soc Lond B Biol Sci 368: 20130024. PubMed ID: 24218636

    Kemkemer, C., Catalan, A. and Parsch, J. (2013). 'Escaping' the X chromosome leads to increased gene expression in the male germline of Drosophila melanogaster. Heredity (Edinb). PubMed ID: 24022496

  • Jose Pastor-Pareja Tsinghua University, Beijing
    Cao, X., Rojas, M. and Pastor-Pareja, J. C. (2022). Intrinsic and damage-induced JAK/STAT signaling regulate developmental timing by the Drosophila prothoracic gland. Dis Model Mech 15(1). PubMed ID: 34842272

    Sun, T., Song, Y., Teng, D., Chen, Y., Dai, J., Ma, M., Zhang, W. and Pastor-Pareja, J. C. (2021). Atypical laminin spots and pull-generated microtubule-actin projections mediate Drosophila wing adhesion. Cell Rep 36(10): 109667. PubMed ID: 34496252

    Yang, K., Liu, M., Feng, Z., Rojas, M., Zhou, L., Ke, H. and Pastor-Pareja, J. C. (2021). ER exit sites in Drosophila display abundant ER-Golgi vesicles and pearled tubes but no megacarriers. Cell Rep 36(11): 109707. PubMed ID: 34525362

    Chen, Y., Sun, T., Bi, Z., Ni, J. Q., Pastor-Pareja, J. C. and Javid, B. (2020). Premature termination codon readthrough in Drosophila varies in a developmental and tissue-specific manner. Sci Rep 10(1): 8485. PubMed ID: 32444687

    Sun, T., Song, Y., Dai, J., Mao, D., Ma, M., Ni, J. Q., Liang, X. and Pastor-Pareja, J. C., (2019). Spectraplakin Shot maintains perinuclear microtubule organization in Drosophila polyploid cells. Dev Cell. PubMed ID: 31006649

    Ke, H., Feng, Z., Liu, M., Sun, T., Dai, J., Ma, M., Liu, L. P., Ni, J. Q. and Pastor-Pareja, J. C. (2018). Collagen secretion screening in Drosophila supports a common secretory machinery and multiple Rab requirements. J Genet Genomics. PubMed ID: 29935791

    Ma, M., Cao, X., Dai, J. and Pastor-Pareja, J. C. (2017). Basement Membrane Manipulation in Drosophila Wing Discs Affects Dpp Retention but Not Growth Mechanoregulation. Dev Cell 42(1): 97-106.e104. PubMed ID: 28697337

    Dai, J., Ma, M., Feng, Z. and Pastor-Pareja, J. C. (2017). Inter-adipocyte Adhesion and Signaling by Collagen IV Intercellular Concentrations in Drosophila. Curr Biol 27(18): 2729-2740.e2724. PubMed ID: 28867208

    Ma, M., Cao, X., Dai, J. and Pastor-Pareja, J. C. (2017). Basement Membrane Manipulation in Drosophila Wing Discs Affects Dpp Retention but Not Growth Mechanoregulation. Dev Cell 42(1): 97-106.e104. PubMed ID: 28697337

    Liu, M., Feng, Z., Ke, H., Liu, Y., Sun, T., Dai, J., Cui, W. and Pastor-Pareja, J. C. (2017). Tango1 spatially organizes ER exit sites to control ER export. J Cell Biol. PubMed ID: 28280122

    Pastor-Pareja, J. C. and Xu, T. (2013). Dissecting Social Cell Biology and Tumors Using Drosophila Genetics. Annu Rev Genet. PubMed ID: 23988119

  • Linda Partridge Max Planck Institute for Biology of Ageing, Cologne
    Tain, L. S., Sehlke, R., Meilenbrock, R. L., Leech, T., Paulitz, J., Chokkalingam, M., Nagaraj, N., Gronke, S., Frohlich, J., Atanassov, I., Mann, M., Beyer, A. and Partridge, L. (2021). Tissue-specific modulation of gene expression in response to lowered insulin signalling in Drosophila. Elife 10. PubMed ID: 33879316

    Niccoli, T., Kerr, F., Snoeren, I., Fabian, D., Aleyakpo, B., Ivanov, D., Sofola-Adesakin, O., Cryar, A., Adcott, J., Thornton, J. and Partridge, L. (2021). Activating transcription factor 4-dependent lactate dehydrogenase activation as a protective response to amyloid beta toxicity. Brain Commun 3(2): fcab053. PubMed ID: 33977265

    Lu, Y. X., Regan, J. C., Eßer, J., Drews, L. F., Weinseis, T., Stinn, J., Hahn, O., Miller, R. A., Grönke, S. and Partridge, L. (2021). A TORC1-histone axis regulates chromatin organisation and non-canonical induction of autophagy to ameliorate ageing. Elife 10. PubMed ID: 33988501

    Atilano, M. L., Grpnke, S., Niccoli, T., Kempthorne, L., Hahn, O., Moron-Oset, J., Hendrich, O., Dyson, M., Adams, M. L., Hull, A., Salcher-Konrad, M. T., Monaghan, A., Bictash, M., Glaria, I., Isaacs, A. M. and Partridge, L. (2021). Enhanced insulin signalling ameliorates C9orf72 hexanucleotide repeat expansion toxicity in Drosophila. Elife 10. PubMed ID: 33739284

    Fabian, D. K., Melike Donertaş, 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

    Bjedov, I., Cocheme, H. M., Foley, A., Wieser, D., Woodling, N. S., Castillo-Quan, J. I., Norvaisas, P., Lujan, C., Regan, J. C., Toivonen, J. M., Murphy, M. P., Thornton, J., Kinghorn, K. J., Neufeld, T. P., Cabreiro, F. and Partridge, L. (2020). Fine-tuning autophagy maximises lifespan and is associated with changes in mitochondrial gene expression in Drosophila. PLoS Genet 16(11): e1009083. PubMed ID: 33253201

    Scholes, H. M., Cryar, A., Kerr, F., Sutherland, D., Gethings, L. A., Vissers, J. P. C., Lees, J. G., Orengo, C. A., Partridge, L. and Thalassinos, K. (2020). Dynamic changes in the brain protein interaction network correlates with progression of Ab42 pathology in Drosophila. Sci Rep 10(1): 18517. PubMed ID: 33116184

    Woodling, N. S., Rajasingam, A., Minkley, L. J., Rizzo, A. and Partridge, L. (2020). Independent glial subtypes delay development and extend healthy lifespan upon reduced insulin-PI3K signalling. BMC Biol 18(1): 124. PubMed ID: 32928209

    Weigelt, C. M., Sehgal, R., Tain, L. S., Cheng, J., Eßer, J., Pahl, A., Dieterich, C., Gronke, S. and Partridge, L. (2020). An Insulin-Sensitive Circular RNA that Regulates Lifespan in Drosophila. Mol Cell 79(2): 268-279.e265. PubMed ID: 32592682

    Yu, Y., Niccoli, T., Ren, Z., Woodling, N. S., Aleyakpo, B., Szabadkai, G. and Partridge, L. (2020). PICALM rescues glutamatergic neurotransmission, behavioural function, and survival in a Drosophila model of Abeta42 toxicity. Hum Mol Genet. PubMed ID: 32592479

  • Nipam Patel Marine Biological Laboratory, Woods Hole, MA
    Bruce, H. S. and Patel, N. H. (2020). Knockout of crustacean leg patterning genes suggests that insect wings and body walls evolved from ancient leg segments. Nat Ecol Evol 4(12): 1703-1712. PubMed ID: 33262517

    Gu, S., Wang, F., Patel, N. P., Bourgeois, J. A. and Huang, J. H. (2019). A Model for Basic Emotions Using Observations of Behavior in Drosophila. Front Psychol 10: 781. PubMed ID: 31068849

    Patel, R. S., Carter, G., Cooper, D. R., Apostolatos, H. and Patel, N. A. (2014). Transformer 2beta homolog (Drosophila) (TRA2B) regulates protein kinase C deltaI (PKCdeltaI) splice variant expression during 3T3L1 preadipocyte cell cycle. J Biol Chem 289(46): 31662-31672. PubMed ID: 25261467

    Hoxha, V., Lama, C., Chang, P. L., Saurabh, S., Patel, N., Olate, N. and Dauwalder, B. (2013). Sex-specific signaling in the blood-brain barrier is required for male courtship in Drosophila. PLoS Genet 9(1): e1003217. PubMed ID: 23359644

  • Achim Paululat Department of Zoology & Developmental Biology, Osnabrück/Germany
    Togel, M., Pass, G. and Paululat, A. (2021). Wing Hearts in Four-Winged Ultrabithorax-mutant Flies-the role of Hox genes in wing heart specification. Genetics. PubMed ID: 34791231

    Dehnen, L., Janz, M., Verma, J. K., Psathaki, O. E., Langemeyer, L., Fröhlich, F., Heinisch, J. J., Meyer, H., Ungermann, C. and Paululat, A. (2020). A trimeric metazoan Rab7 GEF complex is crucial for endocytosis and scavenger function. J Cell Sci. PubMed ID: 32499409

    Beyenbach, K. W., Schoene, F., Breitsprecher, L. F., Tiburcy, F., Furuse, M., Izumi, Y., Meyer, H., Jonusaite, S., Rodan, A. R. and Paululat, A. (2020). The Septate Junction Protein Tetraspanin 2A is critical to the Structure and Function of Malpighian tubules in Drosophila melanogaster. Am J Physiol Cell Physiol. PubMed ID: 32267718

    Schiemann, R., Lammers, K., Janz, M., Lohmann, J., Paululat, A. and Meyer, H. (2018). Identification and in vivo characterisation of cardioactive peptides in Drosophila melanogaster. Int J Mol Sci 20(1). PubMed ID: 30577424

    Wilmes, A. C., Klinke, N., Rotstein, B., Meyer, H. and Paululat, A. (2018). Biosynthesis and assembly of the Collagen IV-like protein Pericardin in Drosophila melanogaster. Biol Open 7(4). PubMed ID: 29685999

    Rotstein, B., Post, Y., Reinhardt, M., Lammers, K., Buhr, A., Heinisch, J. J., Meyer, H. and Paululat, A. (2018). Distinct domains in the matricellular protein Lonely heart are crucial for cardiac extracellular matrix formation and heart function in Drosophila. J Biol Chem [Epub ahead of print]. PubMed ID: 29599288

    Psathaki, O. E., Dehnen, L., Hartley, P. S. and Paululat, A. (2018). Drosophila pericardial nephrocyte ultrastructure changes during ageing. Mech Ageing Dev 173: 9-20. PubMed ID: 29702130

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

    Lammers, K., Abeln, B., Husken, M., Lehmacher, C., Psathaki, O. E., Alcorta, E., Meyer, H. and Paululat, A. (2017). Formation and function of intracardiac valve cells in the Drosophila heart. J Exp Biol [Epub ahead of print]. PubMed ID: 28254880

    Hallier, B., Schiemann, R., Cordes, E., Vitos-Faleato, J., Walter, S., Heinisch, J. J., Malmendal, A., Paululat, A. and Meyer, H. (2016). Drosophila neprilysins control insulin signaling and food intake via cleavage of regulatory peptides. Elife 5. PubMed ID: 27919317

  • Franç Payre Centre de Biologie du Développement, Université de Toulouse, France
    Al Hayek, S., Alsawadi, A., Kambris, Z., Boquete, J. P., Bohère, J., Immarigeon, C., Ronsin, B., Plaza, S., Lemaitre, B., Payre, F. and Osman, D. (2020). Steroid-dependent switch of OvoL/Shavenbaby controls self-renewal versus differentiation of intestinal stem cells. Embo j: e104347. PubMed ID: 33372708

    Ray, S., Rosenberg, M. I., Chanut-Delalande, H., Decaras, A., Schwertner, B., Toubiana, W., Auman, T., Schnellhammer, I., Teuscher, M., Valenti, P., Khila, A., Klingler, M. and Payre, F. (2019). The mlpt/Ubr3/Svb module comprises an ancient developmental switch for embryonic patterning. Elife 8. PubMed ID: 30896406

    Bohere, J., Mancheno-Ferris, A., Al Hayek, S., Zanet, J., Valenti, P., Akino, K., Yamabe, Y., Inagaki, S., Chanut-Delalande, H., Plaza, S., Kageyama, Y., Osman, D., Polesello, C. and Payre, F. (2018). Shavenbaby and Yorkie mediate Hippo signaling to protect adult stem cells from apoptosis. Nat Commun 9(1): 5123. PubMed ID: 30504772

    Gomez-Lamarca, M. J., Falo-Sanjuan, J., Stojnic, R., Abdul Rehman, S., Muresan, L., Jones, M. L., Pillidge, Z., Cerda-Moya, G., Yuan, Z., Baloul, S., Valenti, P., Bystricky, K., Payre, F., O'Holleran, K., Kovall, R. and Bray, S. J. (2018). Activation of the Notch Signaling Pathway In Vivo Elicits Changes in CSL Nuclear Dynamics. Dev Cell 44(5): 611-623 e617. PubMed ID: 29478922

    Zanet, J., Benrabah, E., Li, T., Pelissier-Monier, A., Chanut-Delalande, H., Ronsin, B., Bellen, H. J., Payre, F. and Plaza, S. (2015). Pri sORF peptides induce selective proteasome-mediated protein processing. Science 349(6254): 1356-1358. PubMed ID: 26383956

    Crocker, J., Abe, N., Rinaldi, L., McGregor, A. P., Frankel, N., Wang, S., Alsawadi, A., Valenti, P., Plaza, S., Payre, F., Mann, R. S. and Stern, D. L. (2015). Low affinity binding site clusters confer hox specificity and regulatory robustness. Cell 160(1-2): 191-203. PubMed ID: 25557079

  • Mark Peifer Department of Biology, University of North Carolina at Chapel Hill
    Perez-Vale, K. Z., Yow, K. D., Johnson, R. I., Byrnes, A. E., Finegan, T. M., Slep, K. C. and Peifer, M. (2021). Multivalent interactions make adherens junction-cytoskeletal linkage robust during morphogenesis. J Cell Biol 220(12). PubMed ID: 34762121

    Rogers, E. M., Allred, S. C. and Peifer, M. (2021). Abelson kinase's intrinsically disordered region plays essential roles in protein function and protein stability. Cell Commun Signal 19(1): 27. PubMed ID: 33627133

    Schaefer, K. N., Pronobis, M., Williams, C. E., Zhang, S., Bauer, L., Goldfarb, D., Yan, F., Major, M. B. and Peifer, M. (2020). Wnt Regulation: Exploring Axin-Disheveled interactions and defining mechanisms by which the SCF E3 ubiquitin ligase is recruited to the destruction complex. Mol Biol Cell: mbcE19110647. PubMed ID: 32129710

    Bonello, T. T., Choi, W. and Peifer, M. (2019). Scribble and Discs-large direct initial assembly and positioning of adherens junctions during the establishment of apical-basal polarity. Development 146(22). PubMed ID: 31628110

    Spracklen, A. J., Thornton-Kolbe, E. M., Bonner, A. N., Florea, A., Compton, P. J., Fernandez-Gonzalez, R. and Peifer, M. (2019). The Crk adapter protein is essential for Drosophila embryogenesis, where it regulates multiple actin-dependent morphogenic events. Mol Biol Cell: mbcE19050302. PubMed ID: 31318326

    Manning, L. A., Perez-Vale, K. Z., Schaefer, K. N., Sewell, M. T. and Peifer, M. (2019). The Drosophila Afadin and ZO-1 homologs Canoe and Polychaetoid act in parallel to maintain epithelial integrity when challenged by adherens junction remodeling. Mol Biol Cell: mbcE19040209. PubMed ID: 31188739

    Poulton, J. S., McKay, D. J. and Peifer, M. (2019). Centrosome loss triggers a transcriptional program to counter apoptosis-induced oxidative stress. Genetics. PubMed ID: 30867197

    Bonello, T. T., Vale, K. Z., Sumigray, K. D. and Peifer, M. (2018). Rap1 acts via multiple mechanisms to position Canoe and adherens junctions and mediate apical-basal polarity establishment. Development 145(2). PubMed ID: 29361565

    Poulton, J.S., Cuningham, J.C. and Peifer, M. (2017). Centrosome and spindle assembly checkpoint loss leads to neural apoptosis and reduced brain size. J Cell Biol 216: 1255-1265. PubMed ID: 28351851

    Rogers, E.M, Spracklen, A.J., Bilancia, C.G., Sumigray, K.D., Allred, S.C., Nowotarski, S.H., Schaefer, K.N., Ritchie, B.J. and Peifer, M. (2016). Abelson kinase acts as a robust, multifunctional scaffold in regulating embryonic morphogenesis. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27385341

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

  • Jun Wei Pek Department of Biological Sciences, National University of Singapore
    Voo, K., Ching, J. W. H., Lim, J. W. H., Chan, S. N., Ng, A. Y. E., Heng, J. Y. Y., Lim, S. S. and Pek, J. W. (2021). Maternal starvation primes progeny response to nutritional stress. PLoS Genet 17(11): e1009932. PubMed ID: 34843464

    Jia Ng, S. S., Zheng, R. T., Osman, I. and Pek, J. W. (2018). Generation of Drosophila sisRNAs by Independent Transcription from Cognate Introns. iScience 4: 68-75. PubMed ID: 30240754

    Osman, I. and Pek, J. W. (2018). A sisRNA/miRNA Axis Prevents Loss of Germline Stem Cells during Starvation in Drosophila. Stem Cell Reports 11(1): 4-12. PubMed ID: 30008327

    Ng, A. Y. E., Peralta, K. R. G. and Pek, J. W. (2018). Germline Stem Cell Heterogeneity Supports Homeostasis in Drosophila. Stem Cell Reports 11(1): 13-21. PubMed ID: 29887366

  • Josef Penninger Institute of Molecular Biotechnology, Vienna
    Orthofer, M., Valsesia, A......Penninger, J. M. (2020). Identification of ALK in Thinness. Cell 181(6): 1246-1262. PubMed ID: 32442405

    Zoranovic, T., Manent, J., Willoughby, L., Matos de Simoes, R., La Marca, J. E., Golenkina, S., Cuiping, X., Gruber, S., Angjeli, B., Kanitz, E. E., Cronin, S. J. F., Neely, G. G., Wernitznig, A., Humbert, P. O., Simpson, K. J., Mitsiades, C. S., Richardson, H. E. and Penninger, J. M. (2018). A genome-wide Drosophila epithelial tumorigenesis screen identifies Tetraspanin 29Fb as an evolutionarily conserved suppressor of Ras-driven cancer. PLoS Genet 14(10): e1007688. PubMed ID: 30325918

    Nagy, V., Cole, T., Van Campenhout, C., Khoung, T. M., Leung, C., Vermeiren, S., Novatchkova, M., Wenzel, D., Cikes, D., Polyansky, A. A., Kozieradzki, I., Meixner, A., Bellefroid, E. J., Neely, G. G. and Penninger, J. M. (2015). The evolutionarily conserved transcription factor PRDM12 controls sensory neuron development and pain perception. Cell Cycle: 0. PubMed ID: 25891934

    Neely, G. G., et al. (2012). Construction of a global pain systems network highlights phospholipid signaling as a regulator of heat nociception. PLoS Genet 8: e1003071. PubMed ID: 23236288

    Neely, G. G., Keene, A. C., Duchek, P., Chang, E. C., Wang, Q. P., Aksoy, Y. A., Rosenzweig, M., Costigan, M., Woolf, C. J., Garrity, P. A. and Penninger, J. M. (2011). TrpA1 regulates thermal nociception in Drosophila. PLoS One 6: e24343. PubMed ID: 21909389

    Neely, G. G., et al. (2010). A genome-wide Drosophila screen for heat nociception identifies alpha2delta3 as an evolutionarily conserved pain gene. Cell 143: 628-638. PubMed ID: 21074052

  • Steve Perlman Department of Biology, University of Victoria, Canada
    Ballinger, M. J. and Perlman, S. J. (2017). Generality of toxins in defensive symbiosis: Ribosome-inactivating proteins and defense against parasitic wasps in Drosophila. PLoS Pathog 13(7): e1006431. PubMed ID: 28683136

    Keais, G. L., Hanson, M. A., Gowen, B. E. and Perlman, S. J. (2017). X chromosome drive in a widespread Palearctic woodland fly, Drosophila testacea. J Evol Biol. PubMed ID: 28402000

    Hamilton, P. T., Peng, F., Boulanger, M. J. and Perlman, S. J. (2015). A ribosome-inactivating protein in a Drosophila defensive symbiont. Proc Natl Acad Sci U S A. PubMed ID: 26712000

    Hamilton, P. T., Leong, J. S., Koop, B. F. and Perlman, S. J. (2013). Transcriptional Responses in a Drosophila Defensive Symbiosis. Mol Ecol. PubMed ID: 24274471

    Haselkorn, T. S., Cockburn, S. N., Hamilton, P. T., Perlman, S. J. and Jaenike, J. (2013). Infectious adaptation: potential host range of a defensive endosymbiont in Drosophila. Evolution 67: 934-945. PubMed ID: 23550746

    Cockburn, S. N., Haselkorn, T. S., Hamilton, P. T., Landzberg, E., Jaenike, J. and Perlman, S. J. (2013). Dynamics of the continent-wide spread of a Drosophila defensive symbiont. Ecol Lett 16: 609-616. PubMed ID: 23517577

  • Frédérique Peronnet Developmental Biology, Sorbonne Universités, Institut de Biologie Paris-Seine
    Dardalhon-Cumenal, D., Deraze, J., Dupont, C. A., Ribeiro, V., Coleno-Costes, A., Pouch, J., Le Crom, S., Thomassin, H., Debat, V., Randsholt, N. B. and Peronnet, F. (2018). Cyclin G and the Polycomb Repressive complexes PRC1 and PR-DUB cooperate for developmental stability. PLoS Genet 14(7): e1007498. PubMed ID: 29995890

    Gibert, J. M., Mouchel-Vielh, E. and Peronnet, F. (2018). Pigmentation pattern and developmental constraints: flight muscle attachment sites delimit the thoracic trident of Drosophila melanogaster. Sci Rep 8(1): 5328. PubMed ID: 29593305

    Gibert, J. M., Blanco, J., Dolezal, M., Nolte, V., Peronnet, F. and Schlotterer, C. (2017). Strong epistatic and additive effects of linked candidate SNPs for Drosophila pigmentation have implications for analysis of genome-wide association studies results. Genome Biol 18(1): 126. PubMed ID: 28673357

    Gibert, J. M., Mouchel-Vielh, E. and Peronnet, F. (2017). Modulation of yellow expression contributes to thermal plasticity of female abdominal pigmentation in Drosophila melanogaster. Sci Rep 7: 43370. PubMed ID: 28230190

    Gibert, J. M., Mouchel-Vielh, E., De Castro, S. and Peronnet, F. (2016). Phenotypic Plasticity through Transcriptional Regulation of the Evolutionary Hotspot Gene tan in Drosophila melanogaster. PLoS Genet 12: e1006218. PubMed ID: 27508387

    Dupont, C. A., Dardalhon-Cumenal, D., Kyba, M., Brock, H. W., Randsholt, N. B. and Peronnet, F. (2015). Drosophila Cyclin G and epigenetic maintenance of gene expression during development. Epigenetics Chromatin 8: 18. PubMed ID: 25995770

    Rougeot, J., Renard, M., Randsholt, N. B., Peronnet, F. and Mouchel-Vielh, E. (2013). The elongin complex antagonizes the chromatin factor Corto for vein versus intervein cell identity in Drosophila wings. PLoS One 8: e77592. PubMed ID: 24204884

    Debat, V. and Peronnet, F. (2013). Asymmetric flies: the control of developmental noise in Drosophila. Fly (Austin) 7: 70-77. PubMed ID: 23519089

    Debat, V., Bloyer, S., Faradji, F., Gidaszewski, N., Navarro, N., Orozco-Terwengel, P., Ribeiro, V., Schlotterer, C., Deutsch, J. S. and Peronnet, F. (2011). Developmental stability: a major role for cyclin G in drosophila melanogaster. PLoS Genet 7: e1002314. PubMed ID: 21998598

  • Norbert Perrimon Harvard Medical School
    Xu, J., Kim, A. R., Cheloha, R. W., Fischer, F. A., Li, J. S. S., Feng, Y., Stoneburner, E., Binari, R., Mohr, S. E., Zirin, J., Ploegh, H. L. and Perrimon, N. (2022). Protein visualization and manipulation in Drosophila through the use of epitope tags recognized by nanobodies. Elife 11. PubMed ID: 35076390

    Liu, Y., Li, J. S. S., Rodiger, J., Comjean, A., Attrill, H., Antonazzo, G., Brown, N. H., Hu, Y. and Perrimon, N. (2022). FlyPhoneDB: an integrated web-based resource for cell-cell communication prediction in Drosophila. Genetics 220(3). PubMed ID: 35100387

    Zhao, H., Shi, L., Li, Z., Kong, R., Ren, X., Ma, R., Jia, L., Ma, M., Lu, S., Xu, R., Binari, R., Wang, J. H., Dong, M. Q., Perrimon, N. and Li, Z. (2022). The Yun/Prohibitin complex regulates adult Drosophila intestinal stem cell proliferation through the transcription factor E2F1. Proc Natl Acad Sci U S A 119(6). PubMed ID: 35115400

    Kanca, O., Zirin, J., Hu, Y., Tepe, B., Dutta, D., Lin, W. W., Ma, L., Ge, M., Zuo, Z., Liu, L. P., Levis, R. W., Perrimon, N. and Bellen, H. J. (2022). An expanded toolkit for Drosophila gene tagging using synthesized homology donor constructs for CRISPR-mediated homologous recombination. Elife 11. PubMed ID: 35723254

    Jouandin, P., Marelja, Z., Shih, Y. H., Parkhitko, A. A., Dambowsky, M., Asara, J. M., Nemazanyy, I., Dibble, C. C., Simons, M. and Perrimon, N. (2022). Lysosomal cystine mobilization shapes the response of TORC1 and tissue growth to fasting. Science 375(6582): eabc4203. PubMed ID: 35175796

    Xu, J., Kim, A. R., Cheloha, R. W., Fischer, F. A., Li, J. S. S., Feng, Y., Stoneburner, E., Binari, R., Mohr, S. E., Zirin, J., Ploegh, H. L. and Perrimon, N. (2022). Protein visualization and manipulation in Drosophila through the use of epitope tags recognized by nanobodies. Elife 11. PubMed ID: 35076390

    Parkhitko, A. A., Wang, L., Filine, E., Jouandin, P., Leshchiner, D., Binari, R., Asara, J. M., Rabinowitz, J. D. and Perrimon, N. (2021). A genetic model of methionine restriction extends Drosophila health- and lifespan. Proc Natl Acad Sci U S A 118(40). PubMed ID: 34588310

    Ding, G., Xiang, X., Hu, Y., Xiao, G., Chen, Y., Binari, R., Comjean, A., Li, J., Rushworth, E., Fu, Z., Mohr, S. E., Perrimon, N. and Song, W. (2021). Coordination of tumor growth and host wasting by tumor-derived Upd3. Cell Rep 36(7): 109553. PubMed ID: 34407411

    Droujinine, I. A., Meyer, A. S., Wang, D., Udeshi, N. D., Hu, Y., Rocco, D., McMahon, J. A., Yang, R., Guo, J., Mu, L., Carey, D. K., Svinkina, T., Zeng, R., Branon, T., Tabatabai, A., Bosch, J. A., Asara, J. M., Ting, A. Y., Carr, S. A., McMahon, A. P. and Perrimon, N. (2021). Proteomics of protein trafficking by in vivo tissue-specific labeling. Nat Commun 12(1): 2382. PubMed ID: 33888706

    Tang, H. W., Weng, J. H., Lee, W. X., Hu, Y., Gu, L., Cho, S., Lee, G., Binari, R., Li, C., Cheng, M. E., Kim, A. R., Xu, J., Shen, Z., Xu, C., Asara, J. M., Blenis, J. and Perrimon, N. (2021). mTORC1-chaperonin CCT signaling regulates m(6)A RNA methylation to suppress autophagy. Proc Natl Acad Sci U S A 118(10). PubMed ID: 33649236

  • Anthony Percival-Smith Department of Biology, London, Ontario
    Banerjee, A. and Percival-Smith, A. (2020). Post-translational modifications of Drosophila melanogaster HOX protein, Sex combs reduced. PLoS One 15(1): e0227642. PubMed ID: 31931520

    Newman, C. E., Toxopeus, J., Udaka, H., Ahn, S., Martynowicz, D. M., Graether, S. P., Sinclair, B. J. and Percival-Smith, A. (2017). CRISPR-induced null alleles show that Frost protects Drosophila melanogaster reproduction after cold exposure. J Exp Biol 220(Pt 18): 3344-3354. PubMed ID: 28705828

    Percival-Smith, A. (2016). Non-specificity of transcription factor function in Drosophila melanogaster. Dev Genes Evol [Epub ahead of print]. PubMed ID: 27848019

    Sivanantharajah, L. and Percival-Smith, A. (2015). Differential pleiotropy and HOX functional organization. Dev Biol 398: 1-10. PubMed ID: 25448696

    Sivanantharajah, L. and Percival-Smith, A. (2014). Acquisition of a Leucine Zipper Motif as a Mechanism of Antimorphy for an Allele of the Drosophila Hox Gene Sex combs reduced. G3 (Bethesda). PubMed ID: 24622333

    Kanca, O., Caussinus, E., Denes, A. S., Percival-Smith, A. and Affolter, M. (2014). Raeppli: a whole-tissue labeling tool for live imaging of Drosophila development. Development 141: 472-480. PubMed ID: 24335257

    Percival-Smith, A., Sivanantharajah, L., Pelling, J. J. and Teft, W. A. (2013). Developmental competence and the induction of ectopic proboscises in Drosophila melanogaster. Dev Genes Evol 223: 375-387. PubMed ID: 24121940

    Udaka, H., Percival-Smith, A. and Sinclair, B. J. (2013). Increased abundance of Frost mRNA during recovery from cold stress is not essential for cold tolerance in adult Drosophila melanogaster. Insect Mol Biol 22: 541-550. PubMed ID: 23901849

  • Pedro Pereira MRC Laboratory for Molecular Cell Biology, London
    Tavares, L., Gracio, P., Ramos, R., Traquete, R., Relvas, J. B. and Pereira, P. S. (2021). The Pebble/Rho1/Anillin pathway controls polyploidization and axonal wrapping activity in the glial cells of the Drosophila eye. Dev Biol 473: 90-96. PubMed ID: 33581137

    Eusebio, N., Tavares, L. and Pereira, P. S. (2018). CtBP represses Dpp-dependent Mad activation during Drosophila eye development. Dev Biol. PubMed ID: 30031756

    Tavares, L., Correia, A., Santos, M. A., Relvas, J. B. and Pereira, P. S. (2017). dMyc is required in retinal progenitors to prevent JNK-mediated retinal glial activation. PLoS Genet 13(3): e1006647. PubMed ID: 28267791

    Martins, T., Eusebio, N., Correia, A., Marinho, J., Casares, F. and Pereira, P. S. (2017). TGFbeta/Activin signalling is required for ribosome biogenesis and cell growth in Drosophila salivary glands. Open Biol 7(1). PubMed ID: 28123053

    Tavares, L., Pereira, E., Correia, A., Santos, M. A., Amaral, N., Martins, T., Relvas, J. B. and Pereira, P. S. (2015). Drosophila PS2 and PS3 integrins play distinct roles in retinal photoreceptors-glia interactions. Glia 63(7): 1155-1165. PubMed ID: 25731761

  • Trent Perry University of Melbourne
    Christesen, D., Yang, Y. T., Chen, W., Batterham, P. and Perry, T. (2021). Loss of the Dbeta1 nicotinic acetylcholine receptor subunit disrupts bursicon-driven wing expansion and diminishes adult viability in Drosophila melanogaster. Genetics 219(1). PubMed ID: 34849910

    Somers, J., Luong, H. N., Mitchell, J., Batterham, P. and Perry, T. (2017). Pleiotropic Effects of Loss of the Dalpha1 Subunit in Drosophila melanogaster: Implications for Insecticide Resistance. Genetics 205(1): 263-271. PubMed ID: 28049707

    Christesen, D., Yang, Y. T., Somers, J., Robin, C., Sztal, T., Batterham, P. and Perry, T. (2016). Transcriptome Analysis of Drosophila melanogaster Third Instar Larval Ring Glands Points to Novel Functions and Uncovers a Cytochrome p450 Required for Development. G3 (Bethesda). PubMed ID: 27974438

    Remnant, E. J., Williams, A., Lumb, C., Yang, Y. T., Chan, J., Duchene, S., Daborn, P. J., Batterham, P. and Perry, T. (2016). Evolution, Expression, and Function of Nonneuronal Ligand-Gated Chloride Channels in Drosophila melanogaster. G3 (Bethesda) 6(7): 2003-2012. PubMed ID: 27172217

  • Dmitri Petrov Department of Biology, Stanford University
    >Machado, H. E., Bergland, A. O., Taylor, R., Tilk, S., Behrman, E., Dyer, K., Fabian, D. K., Flatt, T., Gonzalez, J., Karasov, T. L., Kim, B., Kozeretska, I., Lazzaro, B. P., Merritt, T. J., Pool, J. E., O'Brien, K., Rajpurohit, S., Roy, P. R., Schaeffer, S. W., Serga, S., Schmidt, P. and Petrov, D. A. (2021). Broad geographic sampling reveals the shared basis and environmental correlates of seasonal adaptation in Drosophila. Elife 10. PubMed ID: 34155971

    Garud, N. R., Messer, P. W. and Petrov, D. A. (2021). Detection of hard and soft selective sweeps from Drosophila melanogaster population genomic data. PLoS Genet 17(2): e1009373. PubMed ID: 33635910

    Machado, H. E., Lawrie, D. S. and Petrov, D. A. (2019). Pervasive strong selection at the level of codon usage bias in Drosophila melanogaster. Genetics. PubMed ID: 31871131

    Zhao, X., Bergland, A. O., Behrman, E. L., Gregory, B. D., Petrov, D. A. and Schmidt, P. S. (2015). Global transcriptional profiling of diapause and climatic adaptation in Drosophila melanogaster. Mol Biol Evol [Epub ahead of print]. PubMed ID: 26568616

    Bergland, A. O., Tobler, R., Gonzalez, J., Schmidt, P. and Petrov, D. (2015). Secondary contact and local adaptation contribute to genome-wide patterns of clinal variation in Drosophila melanogaster. Mol Ecol [Epub ahead of print]. PubMed ID: 26547394

    Garud, N. R., Messer, P. W., Buzbas, E. O. and Petrov, D. A. (2015). Recent selective sweeps in North American Drosophila melanogaster show signatures of soft sweeps. PLoS Genet 11: e1005004. PubMed ID: 25706129

    Bergland, A. O., Behrman, E. L., O'Brien, K. R., Schmidt, P. S. and Petrov, D. A. (2014). Genomic evidence of rapid and stable adaptive oscillations over seasonal time scales in Drosophila. PLoS Genet 10: e1004775. PubMed ID: 25375361

    Barron, M. G., Fiston-Lavier, A. S., Petrov, D. A. and Gonzalez, J. (2014). Population genomics of transposable elements in Drosophila. Annu Rev Genet. PubMed ID: 25292358

    Staubach, F., Baines, J. F., Kunzel, S., Bik, E. M. and Petrov, D. A. (2013). Host species and environmental effects on bacterial communities associated with Drosophila in the laboratory and in the natural environment. PLoS One 8: e70749. PubMed ID: 23967097

    Lawrie, D. S., Messer, P. W., Hershberg, R. and Petrov, D. A. (2013). Strong Purifying Selection at Synonymous Sites in D. melanogaster. PLoS Genet 9: e1003527. PubMed ID: 23737754

    Zhu, Y., Bergland, A. O., Gonzalez, J. and Petrov, D. A. (2012). Empirical validation of pooled whole genome population re-sequencing in Drosophila melanogaster. PLoS One 7: e41901. PubMed ID: 22848651

  • Cathie Pfleger Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York
    Jahanshahi, M., Hsiao, K., Jenny, A. and Pfleger, C. M. (2016). The Hippo pathway targets Rae1 to regulate mitosis and organ size and to feed back to regulate upstream components Merlin, Hippo, and Warts. PLoS Genet 12: e1006198. PubMed ID: 27494403

    Reimels, T. A. and Pfleger, C. M. (2015). Drosophila Rabex-5 restricts Notch activity in hematopoietic cells and maintains hematopoietic homeostasis. J Cell Sci [Epub ahead of print]. PubMed ID: 26567216

    Ilanges, A., Jahanshahi, M., Balobin, D. M. and Pfleger, C. M. (2013). Alcohol interacts with genetic alteration of the hippo tumor suppressor pathway to modulate tissue growth in Drosophila. PLoS One 8: e78880. PubMed ID: 24205337

    Liu, H. Y., Pfleger, C. M. (2013) Mutation in e1, the ubiquitin activating enzyme, reduces Drosophila lifespan and results in motor impairment. PLoS One 8: e32835. PubMed ID: 23382794

    Pfleger, C. M., Wang, J., Friedman, L., Vittorino, R., Conley, L. M., Ho, L., Fivecoat, H. C. and Pasinetti, G. M. (2010). Grape-seed polyphenolic extract improves the eye phenotype in a Drosophila model of tauopathy. Int J Alzheimers Dis 2010. PubMed ID: 20871666

    Yan, H., Jahanshahi, M., Horvath, E. A., Liu, H. Y. and Pfleger, C. M. (2010). Rabex-5 ubiquitin ligase activity restricts Ras signaling to establish pathway homeostasis in Drosophila. Curr Biol 20: 1378-1382. PubMed ID: 20655224

  • Gert Pflugfelder Institute of Molecular Biology, University of Mainz
    Liu, S., Sun, J., Wang, D., Pflugfelder, G. O. and Shen, J. (2016). Fold formation at the compartment boundary of Drosophila wing requires Yki signaling to suppress JNK dependent apoptosis. Sci Rep 6: 38003. PubMed ID: 27897227

    Tsai, Y.C., Grimm, S., Chao, J.L., Wang, S.C., Hofmeyer, K., Shen, J., Eichinger, F., Michalopoulou, T., Yao, C.K., Chang, C.H., Lin, S.H., Sun, Y.H. and Pflugfelder, G.O. (2015). Optomotor-blind negatively regulates Drosophila eye development by blocking Jak/STAT signaling. PLoS One 10: e0120236. PubMed ID: 25781970

    Shen, J., Lu, J., Sui, L., Wang, D., Yin, M., Hoffmann, I., Legler, A. and Pflugfelder, G. O. (2014). The orthologous Tbx transcription factors Omb and TBX2 induce epithelial cell migration and extrusion in vivo without involvement of matrix metalloproteinases. Oncotarget. PubMed ID: 25344916

    Sen, A., Grimm, S., Hofmeyer, K. and Pflugfelder, G. O. (2014). Optomotor-blind in the development of the Drosophila HS and VS lobula plate tangential cells. J Neurogenet: 1-49. PubMed ID: 24912380

    Zhang, X., Luo, D., Pflugfelder, G. O. and Shen, J. (2013). Dpp signaling inhibits proliferation in the Drosophila wing by Omb-dependent regional control of bantam. Development 140: 2917-2922. PubMed ID: 23821035

    Mayer, L. R., Diegelmann, S., Abassi, Y., Eichinger, F., Pflugfelder, G. O. (2013) Enhancer trap infidelity in Drosophila optomotor-blind. Fly (Austin) 7. PubMed ID: 23519069

    Sui, L., Pflugfelder, G. O. and Shen, J. (2012). The Dorsocross T-box transcription factors promote tissue morphogenesis in the Drosophila wing imaginal disc. Development 139: 2773-2782. PubMed ID: 22782723

    Shen, J., Dahmann, C. and Pflugfelder, G. O. (2010). Spatial discontinuity of optomotor-blind expression in the Drosophila wing imaginal disc disrupts epithelial architecture and promotes cell sorting. BMC Dev Biol 10: 23. PubMed ID: 20178599

    Sen, A., Gadomski, C., Balles, J., Abassi, Y., Dorner, C. and Pflugfelder, G. O. (2010). Null mutations in Drosophila Optomotor-blind affect T-domain residues conserved in all Tbx proteins. Mol Genet Genomics 283: 147-156. PubMed ID: 20033428

  • Nitin Phadnis Department of Biology, University of Utah, Salt Lake City
    Cooper, J. C., Lukacs, A., Reich, S., Schauer, T., Imhof, A. and Phadnis, N. (2019). Altered Localization of Hybrid Incompatibility Proteins in Drosophila. Mol Biol Evol 36(8): 1783-1792. PubMed ID: 31038678

    Fuller, Z. L., Koury, S. A., Phadnis, N. and Schaeffer, S. W. (2019). How chromosomal rearrangements shape adaptation and speciation: Case studies in Drosophila pseudoobscura and its sibling species Drosophila persimilis. Mol Ecol 28(6): 1283-1301. PubMed ID: 30402909

    Phadnis, N., Baker, E. P., Cooper, J. C., Frizzell, K. A., Hsieh, E., de la Cruz, A. F., Shendure, J., Kitzman, J. O. and Malik, H. S. (2015). An essential cell cycle regulation gene causes hybrid inviability in Drosophila. Science 350(6267): 1552-1555. PubMed ID: 26680200

  • Lucia Piacentini Sapienza University of Rome - Department of Biology and Biotechnology "Charles Darwin" BBCD
    Casale, A. M., Liguori, F., Ansaloni, F., Cappucci, U., Finaurini, S., Spirito, G., Persichetti, F., Sanges, R., Gustincich, S. and Piacentini, L. (2022). Transposable element activation promotes neurodegeneration in a Drosophila model of Huntington's disease. iScience 25(1): 103702. PubMed ID: 35036881

    Casale, A. M., Cappucci, U., Fanti, L. and Piacentini, L. (2019). Heterochromatin protein 1 (HP1) is intrinsically required for post-transcriptional regulation of Drosophila Germline Stem Cell (GSC) maintenance. Sci Rep 9(1): 4372. PubMed ID: 30867469

    Laneve, P., Piacentini, L., Casale, A. M., Capauto, D., Gioia, U., Cappucci, U., Di Carlo, V., Bozzoni, I., Di Micco, P., Morea, V., Di Franco, C. A. and Caffarelli, E. (2017). Drosophila CG3303 is an essential endoribonuclease linked to TDP-43-mediated neurodegeneration. Sci Rep 7: 41559. PubMed ID: 28139767

    Caizzi, R., Moschetti, R., Piacentini, L., Fanti, L., Marsano, R. M. and Dimitri, P. (2016). Comparative Genomic Analyses Provide New Insights into the Evolutionary Dynamics of Heterochromatin in Drosophila. PLoS Genet 12(8): e1006212. PubMed ID: 27513559

  • Franck Pichaud Laboratory for Molecular Cell Biology, University College London
    Blackie, L., Walther, R. F., Staddon, M. F., Banerjee, S. and Pichaud, F. (2020). Cell-type specific mechanical response and myosin dynamics during retinal lens development in Drosophila. Mol Biol Cell: mbcE19090523. PubMed ID: 32320320

    Nunes de Almeida, F., Walther, R. F., Presse, M. T., Vlassaks, E. and Pichaud, F. (2019). Cdc42 defines apical identity and regulates epithelial morphogenesis by promoting apical recruitment of Par6-aPKC and Crumbs. Development 146(15). PubMed ID: 31405903

    Walther, R. F., Burki, M., Pinal, N., Rogerson, C. and Pichaud, F. (2018). Rap1, canoe and Mbt cooperate with Bazooka to promote zonula adherens assembly in the fly photoreceptor. J Cell Sci. PubMed ID: 29507112

    Pichaud, F. (2013). Transcriptional regulation of tissue organization and cell morphogenesis: the fly retina as a case study. Dev Biol. PubMed ID: 24099926

    Fichelson, P., Brigui, A. and Pichaud, F. (2012). Orthodenticle and Kruppel homolog 1 regulate Drosophila photoreceptor maturation. Proc Natl Acad Sci U S A 109: 7893-7898. PubMed ID: 22547825

    Pinal, N. and Pichaud, F. (2011). Dynamin- and Rab5-dependent endocytosis is required to prevent Drosophila photoreceptor degeneration. J Cell Sci 124: 1564-1570. PubMed ID: 21486953

  • Nicolas Pichaud Department of Chemistry and Biochemistry Universite de Moncton, New Brunswick, Canada Department:
    Jorgensen, L. B., Overgaard, J., Hunter-Manseau, F. and Pichaud, N. (2021). Dramatic changes in mitochondrial substrate use at critically high temperatures: a comparative study using Drosophila. J Exp Biol. PubMed ID: 33563650

    Cormier, R. J., Strang, R., Menail, H., Touaibia, M. and Pichaud, N. (2021). Systemic and mitochondrial effects of metabolic inflexibility induced by high fat diet in Drosophila melanogaster. Insect Biochem Mol Biol: 103556. PubMed ID: 33626368

    Simard, C. J., Touaibia, M., Allain, E. P., Hebert-Chatelain, E. and Pichaud, N. (2020). Role of the Mitochondrial Pyruvate Carrier in the Occurrence of Metabolic Inflexibility in Drosophila melanogaster Exposed to Dietary Sucrose. Metabolites 10(10). PubMed ID: 33066485

    Simard, C., Lebel, A., Allain, E. P., Touaibia, M., Hebert-Chatelain, E. and Pichaud, N. (2020). Metabolic Characterization and Consequences of Mitochondrial Pyruvate Carrier Deficiency in Drosophila melanogaster. Metabolites 10(9). PubMed ID: 32899962

  • Leslie Pick University of Maryland, Entomology Department, College Park, MD
    Graham, P. L., Fischer, M. D., Giri, A. and Pick, L. (2021). The fushi tarazu zebra element is not required for Drosophila viability or fertility. G3 (Bethesda) 11(11). PubMed ID: 34518886

    Cheatle Jarvela, A. M., Trelstad, C. S. and Pick, L. (2021). Anterior-posterior patterning of segments in Anopheles stephensi offers insights into the transition from sequential to simultaneous segmentation in holometabolous insects. J Exp Zool B Mol Dev Evol. PubMed ID: 34734470

    Cheatle Jarvela, A. M., Trelstad, C. S. and Pick, L. (2020). Regulatory gene function handoff allows essential gene loss in mosquitoes. Commun Biol 3(1): 540. PubMed ID: 32999445

    Reding, K., Chen, M., Lu, Y., Cheatle Jarvela, A. M. and Pick, L. (2019). Shifting roles of Drosophila pair-rule gene orthologs: segmental expression and function in the milkweed bug Oncopeltus fasciatus. Development 146(17). PubMed ID: 31444220

    Graham, P. L., Anderson, W. R., Brandt, E. A., Xiang, J. and Pick, L. (2019). Dynamic expression of Drosophila segmental cell surface-encoding genes and their pair-rule regulators. Dev Biol. PubMed ID: 30695684

    Daffern, N., Chen, Z., Zhang, Y., Pick, L. and Radhakrishnan, I. (2018). Solution NMR studies of the ligand-binding domain of an orphan nuclear receptor reveals a dynamic helix in the ligand-binding pocket. Biochemistry. PubMed ID: 29547262

    Field, A., Xiang, J., Anderson, W. R., Graham, P. and Pick, L. (2016). Activation of Ftz-F1-responsive genes through Ftz/Ftz-F1 dependent enhancers. PLoS One 11: e0163128. PubMed ID: 27723822

    Xiang, J., Reding, K., Heffer, A. and Pick, L. (2017). Conservation and variation in pair-rule gene expression and function in the intermediate-germ beetle, Dermestes maculatus. Development 144(24):4625-4636. PubMed ID: 29084804

    Li, C. R., Guo, D. and Pick, L. (2013). Independent signaling by Drosophila insulin receptor for axon guidance and growth. Front Physiol 4: 385. PubMed ID: 24478707

    Heffer, A., Grubbs, N., Mahaffey, J. and Pick, L. (2013). The evolving role of the orphan nuclear receptor ftz-f1, a pair-rule segmentation gene. Evol Dev 15: 406-417. PubMed ID: 24261442

    Crivat, G., Lizunov, V. A., Li, C. R., Stenkula, K. G., Zimmerberg, J., Cushman, S. W. and Pick, L. (2013). Insulin Stimulates Translocation of Human GLUT4 to the Membrane in Fat Bodies of Transgenic Drosophila melanogaster. PLoS One 8: e77953. PubMed ID: 24223128

  • Eugenia Piddini University of Bristol, U. K.
    Langton, P. F., Baumgartner, M. E., Logeay, R. and Piddini, E. (2021). Xrp1 and Irbp18 trigger a feed-forward loop of proteotoxic stress to induce the loser status. PLoS Genet 17(12): e1009946. PubMed ID: 34914692

    Baumgartner, M. E., Dinan, M. P., Langton, P. F., Kucinski, I. and Piddini, E. (2021). Proteotoxic stress is a driver of the loser status and cell competition. Nat Cell Biol 23(2): 136-146. PubMed ID: 33495633

    Kucinski, I., Dinan, M., Kolahgar, G. and Piddini, E. (2017). Chronic activation of JNK JAK/STAT and oxidative stress signalling causes the loser cell status. Nat Commun 8(1): 136. PubMed ID: 28743877

    Wagstaff, L., Goschorska, M., Kozyrska, K., Duclos, G., Kucinski, I., Chessel, A., Hampton-O'Neil, L., Bradshaw, C. R., Allen, G. E., Rawlins, E. L., Silberzan, P., Carazo Salas, R. E. and Piddini, E. (2016). Mechanical cell competition kills cells via induction of lethal p53 levels. Nat Commun 7: 11373. PubMed ID: 27109213

  • Jan Pielage Friedrich Miescher Institute for Biomedical Research, Basel
    Weber, T., Stephan, R., Moreno, E. and Pielage, J. (2019). The ankyrin repeat domain controls presynaptic localization of Drosophila Ankyrin2 and is essential for synaptic stability. Front Cell Dev Biol 7: 148. PubMed ID: 31475145

    Siegenthaler, D., Escribano, B., Brauler, V. and Pielage, J. (2019). Selective suppression and recall of long-term memories in Drosophila. PLoS Biol 17(8): e3000400. PubMed ID: 31454345

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

    Siegenthaler, D., Enneking, E. M., Moreno, E. and Pielage, J. (2015). L1CAM/Neuroglian controls the axon-axon interactions establishing layered and lobular mushroom body architecture. J Cell Biol 208: 1003-1018. PubMed ID: 25825519

    Stephan, R., Goellner, B., Moreno, E., Frank, C. A., Hugenschmidt, T., Genoud, C., Aberle, H. and Pielage, J. (2015). Hierarchical microtubule organization controls axon caliber and transport and determines synaptic structure and stability. Dev Cell 33: 5-21. PubMed ID: 25800091

    Bulat, V., Rast, M. and Pielage, J. (2014). Presynaptic CK2 promotes synapse organization and stability by targeting Ankyrin2. J Cell Biol 204: 77-94. PubMed ID: 24395637

  • Francesca Pignoni Center for Vision Research, Upstate Medical University, Syracuse, NY
    Neal, S. J., Zhou, Q. and Pignoni, F. (2020). STRIPAK-PP2A regulates Hippo-Yorkie signaling to suppress retinal fate in the Drosophila eye disc peripodial epithelium. J Cell Sci. PubMed ID: 32184260

    Neal, S. J., Dolezal, D., Jusic, N. and Pignoni, F. (2019). Drosophila ML-DmD17-c3 cells respond robustly to Dpp and exhibit complex transcriptional feedback on BMP signaling components. BMC Dev Biol 19(1): 1. PubMed ID: 30669963

    Zhou, Q., Yu, L., Friedrich, M. and Pignoni, F. (2016). Distinct regulation of atonal in a visual organ of Drosophila: organ-specific enhancer and lack of autoregulation in the larval eye. Dev Biol [Epub ahead of print]. PubMed ID: 27693434

    Dolezal, D., Liu, Z., Zhou, Q. and Pignoni, F. (2015). Fly LMBR1/LIMR-type protein Lilipod promotes germ-line stem cell self-renewal by enhancing BMP signaling. Proc Natl Acad Sci U S A 112: 13928-13933. PubMed ID: 26512105

    Zhang, T., Zhou, Q., Ogmundsdottir, M. H., Moller, K., Siddaway, R., Larue, L., Hsing, M., Kong, S. W., Goding, C., Palsson, A., Steingrimsson, E. and Pignoni, F. (2015). Mitf is a master regulator of the v-ATPase forming an Mitf/v-ATPase/TORC1 control module for cellular homeostasis. J Cell Sci. PubMed ID: 26092939

    Zhou, Q., Zhang, T., Jemc, J. C., Chen, Y., Chen, R., Rebay, I. and Pignoni, F. (2013). Onset of atonal expression in Drosophila retinal progenitors involves redundant and synergistic contributions of Ey/Pax6 and So bindings sites within two distant enhancers. Dev Biol. PubMed ID: 24247006

    Yu, L., Zhou, Q., Zhang, C. and Pignoni, F. (2012). Identification of Bombyx atonal and functional comparison with the Drosophila atonal proneural factor in the developing fly eye. Genesis 50: 393-403. PubMed ID: 21998072

    Zhang, T., Zhou, Q. and Pignoni, F. (2011). Yki/YAP, Sd/TEAD and Hth/MEIS control tissue specification in the Drosophila eye disc epithelium. PLoS One 6: e22278. PubMed ID: 21811580

  • Claudio Pikielny Department of Genetics, Dartmouth Medical School
    Vijayan, V., Thistle, R., Liu, T., Starostina, E. and Pikielny, C. W. (2014). Drosophila Pheromone-Sensing Neurons Expressing the ppk25 Ion Channel Subunit Stimulate Male Courtship and Female Receptivity. PLoS Genet 10: e1004238. PubMed ID: 24675786

    Liu, T., Starostina, E., Vijayan, V. and Pikielny, C. W. (2012). Two Drosophila DEG/ENaC channel subunits have distinct functions in gustatory neurons that activate male courtship. J Neurosci 32: 11879-11889. PubMed ID: 22915128

    Starostina, E., Liu, T., Vijayan, V., Zheng, Z., Siwicki, K. K. and Pikielny, C. W. (2012). A Drosophila DEG/ENaC subunit functions specifically in gustatory neurons required for male courtship behavior. J Neurosci 32: 4665-4674. PubMed ID: 22457513

  • Lori Pile Wayne State University, Detroit
    Liu, M., Saha, N., Gajan, A., Saadat, N., Gupta, S. V. and Pile, L. A. (2019). A complex interplay between SAM synthetase and the epigenetic regulator SIN3 controls metabolism and transcription. J Biol Chem. PubMed ID: 31776190

    Liu, M. and Pile, L. A. (2016). The transcriptional corepressor SIN3 directly regulates genes involved in methionine catabolism and affects histone methylation, linking epigenetics and metabolism. J Biol Chem [Epub ahead of print]. PubMed ID: 28028175

    Chaubal, A., Todi, S.V. and Pile, L.A. (2016). Inter-isoform-dependent regulation of the Drosophila master transcriptional regulator SIN3. J Biol Chem [Epub ahead of print]. PubMed ID: 27129248

    Saha, N., Liu, M., Gajan, A. and Pile, L. A. (2016). Genome-wide studies reveal novel and distinct biological pathways regulated by SIN3 isoforms. BMC Genomics 17: 111. PubMed ID: 26872827

    Gajan, A., Barnes, V. L., Liu, M., Saha, N. and Pile, L. A. (2016). The histone demethylase dKDM5/LID interacts with the SIN3 histone deacetylase complex and shares functional similarities with SIN3. Epigenetics Chromatin 9: 4. PubMed ID: 26848313

    Liu, M., Barnes, V. L. and Pile, L. A. (2015). Disruption of Methionine Metabolism in Drosophila melanogaster Impacts Histone Methylation and Results in Loss of Viability. G3 (Bethesda) 6: 121-132. PubMed ID: 26546310

    Holowatyj, A., Yang, Z. Q. and Pile, L. A. (2015). Histone lysine demethylases in Drosophila melanogaster. Fly (Austin) 9: 36-44. PubMed ID: 26207949

  • Sergio Pimpinelli Department of Biology and Biotechnology - Sapienza University of Rome

    Tritto, P., Palumbo, V., Micale, L., Marzulli, M., Bozzetti, M. P., Specchia, V., Palumbo, G., Pimpinelli, S. and Berloco, M. (2015). Loss of Pol32 in Drosophila melanogaster causes chromosome instability and suppresses variegation. PLoS One 10(3): e0120859. PubMed ID: 25826374

    Berloco, M., Palumbo, G., Piacentini, L., Pimpinelli, S. and Fanti, L. (2014). Position effect variegation and viability are both sensitive to dosage of constitutive heterochromatin in Drosophila. G3 (Bethesda) 4(9): 1709-1716. PubMed ID: 25053704

    Piacentini, L., Fanti, L., Negri, R., Del Vescovo, V., Fatica, A., Altieri, F. and Pimpinelli, S. (2009). Heterochromatin protein 1 (HP1a) positively regulates euchromatic gene expression through RNA transcript association and interaction with hnRNPs in Drosophila. PLoS Genet 5(10): e1000670. PubMed ID: 19798443


  • Matt Piper Monash University, Clayton Campus, Victoria, Australia
    Zanco, B., Mirth, C. K., Sgro, C. M. and Piper, M. D. (2021). A dietary sterol trade-off determines lifespan responses to dietary restriction in Drosophila melanogaster females. Elife 10. PubMed ID: 33494859

    Ma, C., Mirth, C. K., Hall, M. D. and Piper, M. D. W. (2020). Amino acid quality modifies the quantitative availability of protein for reproduction in Drosophila melanogaster. J Insect Physiol: 104050. PubMed ID: 32229142

    Wu, Q., Yu, G., Cheng, X., Gao, Y., Fan, X., Yang, D., Xie, M., Wang, T., Piper, M. D. W. and Yang, M. (2020). Sexual dimorphism in the nutritional requirement for adult lifespan in Drosophila melanogaster. Aging Cell 19(3): e13120. PubMed ID: 32069521

    Camilleri-Carter, T. L., Dowling, D. K., R, L. R. and Piper, M. D. W. (2019). Transgenerational Obesity and Healthy Aging in Drosophila. J Gerontol A Biol Sci Med Sci 74(10): 1582-1589. PubMed ID: 31231757

  • Vincenzo Pirrotta Molecular Biology & Biochemistry, Rutgers University
    Kahn, T. G., Dorafshan, E., Schultheis, D., Zare, A., Stenberg, P., Reim, I., Pirrotta, V. and Schwartz, Y. B. (2016). Interdependence of PRC1 and PRC2 for recruitment to Polycomb response elements. Nucleic Acids Res. PubMed ID: 27557709

    Lee, H. G., Kahn, T. G., Simcox, A., Schwartz, Y. B. and Pirrotta, V. (2015). Genome-wide activities of Polycomb complexes control pervasive transcription. Genome Res 25: 1170-1181. PubMed ID: 25986499

    Li, H. B., Ohno, K., Gui, H. and Pirrotta, V. (2013). Insulators target active genes to transcription factories and polycomb-repressed genes to polycomb bodies. PLoS Genet 9: e1003436. PubMed ID: 23637616
    Kahn, T. G., Stenberg, P., Pirrotta, V. and Schwartz, Y. B. (2014). Combinatorial Interactions Are Required for the Efficient Recruitment of Pho Repressive Complex (PhoRC) to Polycomb Response Elements. PLoS Genet 10: e1004495. PubMed ID: 25010632

    Park, S. Y., Schwartz, Y. B., Kahn, T. G., Asker, D. and Pirrotta, V. (2012). Regulation of Polycomb group genes Psc and Su(z)2 in Drosophila melanogaster. Mech. Dev. 128(11-12): 536-47. PubMed ID: 22289633

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

    Schwartz, Y. B., Kahn, T. G., Stenberg, P., Ohno, K., Bourgon, R. and Pirrotta, V. (2010). Alternative epigenetic chromatin states of polycomb target genes. PLoS Genet. 6(1): e1000805. PubMed ID: 20062800

  • Alison Pischedda Department of Biology ​Barnard College, Columbia University
    Sinclair, C. S., Lisa, S. F. and Pischedda, A. (2021). Does sexual experience affect the strength of male mate choice for high-quality females in Drosophila melanogaster? Ecol Evol 11(23): 16981-16992. PubMed ID: 34938486

    Pischedda, A., Shahandeh, M. P. and Turner, T. L. (2020). 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 37(3): 864-880. PubMed ID: 31774527

    Shahandeh, M. P., Pischedda, A., Rodriguez, J. M. and Turner, T. L. (2020). The Genetics of Male Pheromone Preference Difference Between Drosophila melanogaster and Drosophila simulans. G3 (Bethesda) 10(1): 401-415. PubMed ID: 31748379

    Shahandeh, M. P., Pischedda, A. and Turner, T. L. (2018). Male mate choice via cuticular hydrocarbon pheromones drives reproductive isolation between Drosophila species. Evolution 72(1): 123-135. PubMed ID: 29098691

  • Scott Pitnick Biology at Syracuse University
    Lüpold, S., Manier, M.K., Puniamoorthy, N., Schoff, C., Starmer, W.T., Luepold, S.H., Belote, J.M. and Pitnick, S. (2016). How sexual selection can drive the evolution of costly sperm ornamentation. Nature 533: 535-538. PubMed ID: 27225128

    Ala-Honkola, O., Manier, M. K., Lupold, S., Droge-Young, E. M., Collins, W. F., Belote, J. M. and Pitnick, S. (2013). No inbreeding depression in sperm storage ability or offspring viability in Drosophila melanogaster females. J Insect Physiol. PubMed ID: 24188987

    Manier, M. K., Lupold, S., Belote, J. M., Starmer, W. T., Berben, K. S., Ala-Honkola, O., Collins, W. F. and Pitnick, S. (2013). Postcopulatory Sexual Selection Generates Speciation Phenotypes in Drosophila. Curr Biol. PubMed ID: 24076241

    Ala-Honkola, O., Hosken, D. J., Manier, M. K., Lupold, S., Droge-Young, E. M., Berben, K. S., Collins, W. F., Belote, J. M. and Pitnick, S. (2013). Inbreeding reveals mode of past selection on male reproductive characters in Drosophila melanogaster. Ecol Evol 3: 2089-2102. PubMed ID: 23919154

    Manier, M. K., Belote, J. M., Berben, K. S., Lupold, S., Ala-Honkola, O., Collins, W. F. and Pitnick, S. (2013). Rapid diversification of sperm precedence traits and processes among three sibling Drosophila species. Evolution 67: 2348-2362. PubMed ID: 23888856

    Lupold, S., Pitnick, S., Berben, K. S., Blengini, C. S., Belote, J. M. and Manier, M. K. (2013). Female mediation of competitive fertilization success in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 23757499

    Belote, J. M., Lupold, S., Manier, M. K., Pitnick, S. (2013) Opening a window onto sperm competition. Mol Reprod Dev. PubMed ID: 23341262

    Lupold, S., Manier, M. K., Berben, K. S., Smith, K. J., Daley, B. D., Buckley, S. H., Belote, J. M. and Pitnick, S. (2012). How multivariate ejaculate traits determine competitive fertilization success in Drosophila melanogaster. Curr Biol 22: 1667-1672. PubMed ID: 22840512

  • Chrysoula Pitsouli Department of Biological Sciences - University of Cyprus
    Neophytou, C. and Pitsouli, C. (2022). Biotin controls intestinal stem cell mitosis and host-microbiome interactions. Cell Rep 38(10): 110505. PubMed ID: 35263602

    ouli, C. (2022). How Gut Microbes Nurture Intestinal Stem Cells: A Drosophila Perspective. Metabolites 12(2). PubMed ID: 35208243

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

    Tamamouna, V., Panagi, M., Theophanous, A., Demosthenous, M., Michail, M., Papadopoulou, M., Teloni, S., Pitsouli, C. and Apidianakis, Y. (2020). Evidence of two types of balance between stem cell mitosis and enterocyte nucleus growth in the Drosophila midgut. Development 147(11). PubMed ID: 32513656

  • Barry Pittendrigh School of Integrative Biology, Department of Entomology, University of Illinois, Urbana-Champaign
    Zhang, C., Seong, K. M., Sun, W., Mittapalli, O., Qiu, B., Clark, J. M. and Pittendrigh, B. R. (2021). The insulin signaling pathway in Drosophila melanogaster: A nexus revealing an "Achilles' heel" in DDT resistance. Pestic Biochem Physiol 171: 104727. PubMed ID: 33357549

    Seong, K. M., Coates, B. S. and Pittendrigh, B. R. (2020). Post-transcriptional modulation of cytochrome P450s, Cyp6g1 and Cyp6g2, by miR-310s cluster is associated with DDT-resistant Drosophila melanogaster strain 91-R. Sci Rep 10(1): 14394. PubMed ID: 32873850

    Abdu-Allah, G. A. M., Seong, K. M., Mittapalli, O., Ojo, J. A., Sun, W., Posos-Parra, O., Mota-Sanchez, D., Clark, J. M. and Pittendrigh, B. R. (2020). Dietary antioxidants impact DDT resistance in Drosophila melanogaster. PLoS One 15(8): e0237986. PubMed ID: 32841282

    Huang, J., Sun, W., Seong, K. M., Mittapalli, O., Ojo, J., Coates, B., Paige, K. N., Clark, J. M. and Pittendrigh, B. R. (2020). Dietary antioxidant vitamin C influences the evolutionary path of insecticide resistance in Drosophila melanogaster. Pestic Biochem Physiol 168: 104631. PubMed ID: 32711765

    Seong, K. M., Coates, B. S. and Pittendrigh, B. R. (2019). Cytochrome P450s Cyp4p1 and Cyp4p2 associated with the DDT tolerance in the Drosophila melanogaster strain 91-R. Pestic Biochem Physiol 159: 136-143. PubMed ID: 31400775

    Seong, K. M., Sun, W., Clark, J. M. and Pittendrigh, B. R. (2016). Splice form variant and amino acid changes in MDR49 confers DDT resistance in transgenic Drosophila. Sci Rep 6: 23355. PubMed ID: 27003579

    Sun, W., Valero, M. C., Seong, K. M., Steele, L. D., Huang, I. T., Lee, C. H., Clark, J. M., Qiu, X. and Pittendrigh, B. R. (2015). A glycine insertion in the Estrogen-Related Receptor (ERR) is associated with enhanced expression of three cytochrome P450 genes in transgenic Drosophila melanogaster. PLoS One 10: e0118779. PubMed ID: 25761142

    Qiu, X., Sun, W., McDonnell, C. M., Li-Byarlay, H., Steele, L. D., Wu, J., Xie, J., Muir, W. M., Pittendrigh, B. R. (2012) Genome-wide analysis of genes associated with moderate and high DDT resistance in Drosophila melanogaster. Pest Manag Sci. PubMed ID: 23371854

    Walters, K. R., Jr., Rupassara, S. I., Markelz, R. J., Leakey, A. D., Muir, W. M. and Pittendrigh, B. R. (2012). Methamphetamine causes anorexia in Drosophila melanogaster, exhausting metabolic reserves and contributing to mortality. J Toxicol Sci 37: 773-790. PubMed ID: 22863857

    McDonnell, C. M., King, D., Comeron, J. M., Li, H., Sun, W., Berenbaum, M. R., Schuler, M. A. and Pittendrigh, B. R. (2012). Evolutionary toxicogenomics: diversification of the Cyp12d1 and Cyp12d3 genes in Drosophila species. J Mol Evol 74: 281-296. PubMed ID: 22811321

  • Serge Plaza Paul Sabatier University - Toulouse
    Montigny, A., Tavormina, P., Duboe, C., San Clemente, H., Aguilar, M., Valenti, P., Lauressergues, D., Combier, J. P. and Plaza, S. (2021). Drosophila primary microRNA-8 encodes a microRNA-encoded peptide acting in parallel of miR-8. Genome Biol 22(1): 118. PubMed ID: 33892772

    Scheffer, L. K., Xu, C. S., Januszewski, M., Lu, Z., Takemura, S. Y., Hayworth, K. J., Huang, G. B., Shinomiya, K., Maitlin-Shepard, J., Berg, S., Clements, J., Hubbard, P. M., Katz, W. T., Umayam, L., Zhao, T., Ackerman, D., Blakely, T., Bogovic, J., Dolafi, T., Kainmueller, D., Kawase, T., Khairy, K. A., Leavitt, L., Li, P. H., Lindsey, L., Neubarth, N., Olbris, D. J., Otsuna, H., Trautman, E. T., Ito, M., Bates, A. S., Goldammer, J., Wolff, T., Svirskas, R., Schlegel, P., Neace, E., Knecht, C. J., Alvarado, C. X., Bailey, D. A., Ballinger, S., Borycz, J. A., Canino, B. S., Cheatham, N., Cook, M., Dreher, M., Duclos, O., Eubanks, B., Fairbanks, K., Finley, S., Forknall, N., Francis, A., Hopkins, G. P., Joyce, E. M., Kim, S., Kirk, N. A., Kovalyak, J., Lauchie, S., Lohff, A., Maldonado, C., Manley, E. A., McLin, S., Mooney, C., Ndama, M., Ogundeyi, O., Okeoma, N., Ordish, C., Padilla, N., Patrick, C. M., Paterson, T., Phillips, E. E., Phillips, E. M., Rampally, N., Ribeiro, C., Robertson, M. K., Rymer, J. T., Ryan, S. M., Sammons, M., Scott, A. K., Scott, A. L., Shinomiya, A., Smith, C., Smith, K., Smith, N. L., Sobeski, M. A., Suleiman, A., Swift, J., Takemura, S., Talebi, I., Tarnogorska, D., Tenshaw, E., Tokhi, T., Walsh, J. J., Yang, T., Horne, J. A., Li, F., Parekh, R., Rivlin, P. K., Jayaraman, V., Costa, M., Jefferis, G. S., Ito, K., Saalfeld, S., George, R., Meinertzhagen, I., Rubin, G. M., Hess, H. F., Jain, V. and Plaza, S. M. (2020). A connectome and analysis of the adult Drosophila central brain. Elife 9. PubMed ID: 32880371

    Zanet, J., Benrabah, E., Li, T., Pelissier-Monier, A., Chanut-Delalande, H., Ronsin, B., Bellen, H. J., Payre, F. and Plaza, S. (2015). Pri sORF peptides induce selective proteasome-mediated protein processing. Science 349: 1356-1358. PubMed ID: 26383956

    Chanut-Delalande, H., Hashimoto, Y., Pelissier-Monier, A., Spokony, R., Dib, A., Kondo, T., Bohere, J., Niimi, K., Latapie, Y., Inagaki, S., Dubois, L., Valenti, P., Polesello, C., Kobayashi, S., Moussian, B., White, K. P., Plaza, S., Kageyama, Y. and Payre, F. (2014). Pri peptides are mediators of ecdysone for the temporal control of development. Nat Cell Biol 16: 1035-1044. PubMed ID: 25344753

    Menoret, D., Santolini, M., Fernandes, I., Spokony, R., Zanet, J., Gonzalez, I., Latapie, Y., Ferrer, P., Rouault, H., White, K. P., Besse, P., Hakim, V., Aerts, S., Payre, F. and Plaza, S. (2013). Genome-wide analyses of Shavenbaby target genes reveals distinct features of enhancer organization. Genome Biol 14: R86. PubMed ID: 23972280

    Zanet, J., Payre, F. and Plaza, S. (2009). Fascin for cell migration in Drosophila. Fly (Austin) 3: 281-282. PubMed ID: 19901519

  • Anne Plessis Development, Signalling and Trafficking, Institut Jacques Monod, Paris
    Bruzzone, L., Argüelles, C., Sanial, M., Miled, S., Alvisi, G., Gonçalves-Antunes, M., Qasrawi, F., Holmgren, R. A., Smibert, C. A., Lipshitz, H. D., Boccaccio, G. L., Plessis, A. and Bécam, I. (2020). Regulation of the RNA-binding protein Smaug by the GPCR Smoothened via the kinase Fused. EMBO Rep: e48425. PubMed ID: 32383557

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

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

    Yanai, H., Yoshioka, Y., Yoshida, H., Nakao, Y., Plessis, A. and Yamaguchi, M. (2014). Drosophila myeloid leukemia factor acts with DREF to activate the JNK signaling pathway. Oncogenesis 3: e98. PubMed ID: 24752236

    Marchal, C., Vinatier, G., Sanial, M., Plessis, A., Pret, A. M., Limbourg-Bouchon, B., Theodore, L. and Netter, S. (2012). The HIV-1 Vpu protein induces apoptosis in Drosophila via activation of JNK signaling. PLoS One 7: e34310. PubMed ID: 22479597

  • Scott Pletcher Department of Molecular and Integrative Physiology and Geriatrics Center, University of Michigan, Ann Arbor
    Lyu, Y., Promislow, D. E. L. and Pletcher, S. D. (2021). Serotonin signaling modulates aging-associated metabolic network integrity in response to nutrient choice in Drosophila melanogaster. Commun Biol 4(1): 740. PubMed ID: 34131274

    Chakraborty, T. S., Gendron, C. M., Lyu, Y., Munneke, A. S., DeMarco, M. N., Hoisington, Z. W. and Pletcher, S. D. (2019). Sensory perception of dead conspecifics induces aversive cues and modulates lifespan through serotonin in Drosophila. Nat Commun 10(1): 2365. PubMed ID: 31147540

    Gendron, C. M. and Pletcher, S. D. (2017). MicroRNAs mir-184 and let-7 alter Drosophila metabolism and longevity. Aging Cell 16(6): 1434-1438. PubMed ID: 28963741

    Fedina, T. Y., Arbuthnott, D., Rundle, H. D., Promislow, D. E. L. and Pletcher, S. D. (2017). Tissue-specific insulin signaling mediates female sexual attractiveness. PLoS Genet 13(8): e1006935. PubMed ID: 28817572

    Lee, J. E., Rayyan, M., Liao, A., Edery, I. and Pletcher, S. D. (2017). Acute dietary restriction acts via TOR, PP2A, and Myc signaling to boost innate immunity in Drosophila. Cell Rep 20(2): 479-490. PubMed ID: 28700947

    Harvanek, Z. M., Lyu, Y., Gendron, C. M., Johnson, J. C., Kondo, S., Promislow, D. E. L. and Pletcher, S. D. (2017). Perceptive costs of reproduction drive ageing and physiology in male Drosophila. Nat Ecol Evol 1(6): 152. PubMed ID: 28812624

    Fedina, T. Y., Arbuthnott, D., Rundle, H. D., Promislow, D. E. L. and Pletcher, S. D. (2017). Tissue-specific insulin signaling mediates female sexual attractiveness. PLoS Genet 13(8): e1006935. PubMed ID: 28817572

    Chung, B. Y., Ro, J., Hutter, S. A., Miller, K. M., Guduguntla, L. S., Kondo, S. and Pletcher, S. D. (2017). Drosophila Neuropeptide F signaling independently regulates feeding and sleep-wake behavior. Cell Rep 19(12): 2441-2450. PubMed ID: 28636933

    Arbuthnott, D., Fedina, T.Y., Pletcher, S.D. and Promislow, D.E. (2017). Mate choice in fruit flies is rational and adaptive. Nat Commun 8: 13953. PubMed ID: 28094789

    Harvanek, Z. M., Mourao, M. A., Schnell, S. and Pletcher, S. D. (2016). A computational approach to studying ageing at the individual level. Proc Biol Sci 283. PubMed ID: 26865300

    Waterson, M. J., Chan, T. P. and Pletcher, S. D. (2015). Adaptive physiological response to perceived scarcity as a mechanism of sensory modulation of life span. J Gerontol A Biol Sci Med Sci. PubMed ID: 25878032

  • Nancy Jo Jo Pokrywka Department of Biology, Vassar College, Poughkeepsie, New York
    Starble, R. and Pokrywka, N. J. (2017). The retromer subunit Vps26 mediates Notch signaling during Drosophila oogenesis. Mech Dev. PubMed ID: 29031909

    Pokrywka, N. J., Zhang, H. and Raley-Susman, K. (2014). Distinct roles for hu li tai shao and swallow in cytoskeletal organization during Drosophila oogenesis. Dev Dyn 243(7): 906-916. PubMed ID: 24677508

    Pokrywka, N. J. (2013). Live imaging of GFP-labeled proteins in Drosophila oocytes. J Vis Exp(73). PubMed ID: 23567977

  • Michal Polak University of Cincinnati
    Rodriguez-Exposito, E., Garcia-Gonzalez, F. and Polak, M. (2019). Individual and synergistic effects of male external genital traits in sexual selection. J Evol Biol. PubMed ID: 31554023

    LeVasseur-Viens, H., Polak, M. and Moehring, A. J. (2015). No evidence for external genital morphology affecting cryptic female choice and reproductive isolation in Drosophila. Evolution 69(7): 1797-1807. PubMed ID: 25962316

    Grieshop, K. and Polak, M. (2014). Evaluating the post-copulatory sexual selection hypothesis for genital evolution reveals evidence for pleiotropic harm exerted by the male genital spines of Drosophila ananassae. J Evol Biol 27(12): 2676-2686. PubMed ID: 25403358

  • John Pool Laboratory of Genetics, University of Wisconsin, Madison
    Sprengelmeyer, Q. D., Lack, J. B., Braun, D. T., Monette, M. J. and Pool, J. E. (2022). The evolution of larger size in high-altitude Drosophila melanogaster has a variable genetic architecture. G3 (Bethesda) 12(3). PubMed ID: 35100377

    Lange, J. D., Bastide, H., Lack, J. B. and Pool, J. E. (2021). A Population Genomic Assessment of Three Decades of Evolution in a Natural Drosophila Population. Mol Biol Evol. PubMed ID: 34971382

    Huang, Y., Lack, J. B., Hoppel, G. T. and Pool, J. E. (2022). Gene regulatory evolution in cold-adapted fly populations neutralizes plasticity and may undermine genetic canalization. Genome Biol Evol. PubMed ID: 35380655

    Sprengelmeyer, Q. D. and Pool, J. E. (2021). Ethanol resistance in Drosophila melanogaster has increased in parallel cold-adapted populations and shows a variable genetic architecture within and between populations. Ecol Evol 11(21): 15364-15376. PubMed ID: 34765183

    Huang, Y., Lack, J. B., Hoppel, G. T. and Pool, J. E. (2021). Parallel and Population-specific Gene Regulatory Evolution in Cold-Adapted Fly Populations. Genetics. PubMed ID: 33989401

    Sprengelmeyer, Q. D., Mansourian, S., Lange, J. D., Matute, D. R., Cooper, B. S., Jirle, E. V., Stensmyr, M. C. and Pool, J. E. (2019). Recurrent collection of Drosophila melanogaster from Wild african environments and genomic insights into species history. Mol Biol Evol. PubMed ID: 31730190

    Lange, J. D. and Pool, J. E. (2018). Impacts of Recurrent Hitchhiking on Divergence and Demographic Inference in Drosophila. Genome Biol Evol. PubMed ID: 30010915

    Groth, B. R., Huang, Y., Monette, M. J. and Pool, J. E. (2018). Directional selection reduces developmental canalization against genetic and environmental perturbations in Drosophila wings. Evolution. PubMed ID: 29985527

    Bastide, H., Lange, J. D., Lack, J. B., Yassin, A. and Pool, J. E. (2016). A Variable Genetic architecture of melanic evolution in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 27638419

    Lack, J. B., Yassin, A., Sprengelmeyer, Q. D., Johanning, E. J., David, J. R. and Pool, J. E. (2016). Life history evolution and cellular mechanisms associated with increased size in high-altitude Drosophila. Ecol Evol 6: 5893-5906. PubMed ID: 27547363

    Yassin, A., Debat, V., Bastide, H., Gidaszewski, N., David, J.R. and Pool, J.E. (2016). Recurrent specialization on a toxic fruit in an island Drosophila population. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27044093

    Yassin, A., Bastide, H., Chung, H., Veuille, M., David, J. R. and Pool, J. E. (2016). Ancient balancing selection at tan underlies female colour dimorphism in Drosophila erecta. Nat Commun 7: 10400. PubMed ID: 26778363

    Lack, J. B., Monette, M. J., Johanning, E. J., Sprengelmeyer, Q. D. and Pool, J. E. (2016). Decanalization of wing development accompanied the evolution of large wings in high-altitude Drosophila. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26755605

  • John Poulton Department of Medicine, University of North Carolina
    Mysh, M. and Poulton, J. S. (2021). The Basolateral Polarity Module Promotes Slit Diaphragm Formation in Drosophila Nephrocytes, a Model of Vertebrate Podocytes. J Am Soc Nephrol. PubMed ID: 33795424

    Poulton, J. S., McKay, D. J. and Peifer, M. (2019). Centrosome loss triggers a transcriptional program to counter apoptosis-induced oxidative stress. Genetics. PubMed ID: 30867197

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

  • James Posakony Section of Cell and Developmental Biology, UC San Diego
    Miller, S. W. and Posakony, J. W. (2020). Disparate expression specificities coded by a shared Hox-C enhancer. Elife 9. PubMed ID: 32342858

    Miller, S. W., Movsesyan, A., Zhang, S., Fernandez, R. and Posakony, J. W. (2019). Evolutionary emergence of Hairless as a novel component of the Notch signaling pathway. Elife 8. PubMed ID: 31545167

    Miller, S. W. and Posakony, J. W. (2018). Lateral inhibition: Two modes of non-autonomous negative autoregulation by neuralized. PLoS Genet 14(7): e1007528. PubMed ID: 30028887

    Miller, S. W., Rebeiz, M., Atanasov, J. E. and Posakony, J. W. (2014). Neural precursor-specific expression of multiple Drosophila genes is driven by dual enhancer modules with overlapping function. Proc Natl Acad Sci U S A. PubMed ID: 25404315

    Liu, F. and Posakony, J. W. (2014). An enhancer composed of interlocking submodules controls transcriptional autoregulation of suppressor of hairless. Dev Cell 29: 88-101. PubMed ID: 24735880

    Liu, F., and Posakony, J. W. (2012). Role of architecture in the function and specificity of two Notch-regulated transcriptional enhancer modules. PLoS Genet. 8: e1002796. PubMed ID: 22792075

    Rebeiz, M., Castro, B., Liu, F., Yue, F., and Posakony, J. W. (2012). Ancestral and conserved cis-regulatory architectures in developmental control genes. Dev. Biol. 362: 282-294. PubMed ID: 22185795

    Rebeiz, M., Miller, S. W. and Posakony, J. W. (2011). Notch regulates numb: integration of conditional and autonomous cell fate specification. Development 138: 215-225. PubMed ID: 21148185

  • Nico Posnien Department of Developmental Biology, Georg-August-University Goettingen
    Buchberger, E., Bilen, A., Ayaz, S., Salamanca, D., Matas de Las Heras, C., Niksic, A., Almudi, I., Torres-Oliva, M., Casares, F. and Posnien, N. (2021). Variation in pleiotropic hub gene expression is associated with interspecific differences in head shape and eye size in Drosophila. Mol Biol Evol. PubMed ID: 33386848

    Reis, M., Wiegleb, G., Claude, J., Lata, R., Horchler, B., Ha, N. T., Reimer, C., Vieira, C. P., Vieira, J. and Posnien, N. (2020). Multiple loci linked to inversions are associated with eye size variation in species of the Drosophila virilis phylad. Sci Rep 10(1): 12832. PubMed ID: 32732947

    Torres-Oliva, M., Schneider, J., Wiegleb, G., Kaufholz, F. and Posnien, N. (2018). Dynamic genome wide expression profiling of Drosophila head development reveals a novel role of Hunchback in retinal glia cell development and blood-brain barrier integrity. PLoS Genet 14(1): e1007180. PubMed ID: 29360820

    Al Khatib, A., Siomava, N., Iannini, A., Posnien, N. and Casares, F. (2017). Specific expression and function of the Six3 optix in Drosophila serially homologous organs. Biol Open 6(8): 1155-1164. PubMed ID: 28642242

    Siomava, N., Wimmer, E. A. and Posnien, N. (2016). Size relationships of different body parts in the three dipteran species Drosophila melanogaster, Ceratitis capitata and Musca domestica. Dev Genes Evol 226(3): 245-256. PubMed ID: 27116604

  • Chris Potter Neurosciences, Johns Hopkins, Baltimore, Md.
    Marr, E. and Potter, C. J. (2021). Base Editing of Somatic Cells Using CRISPR-Cas9 in Drosophila. Crispr j 4(6): 836-845. PubMed ID: 34813372

    Maguire, S. E., Afify, A., Goff, L. A. and Potter, C. J. (2022). Odorant-receptor-mediated regulation of chemosensory gene expression in the malaria mosquito Anopheles gambiae. Cell Rep 38(10): 110494. PubMed ID: 35263579

    Raji, J. I. and Potter, C. J. (2021). The number of neurons in Drosophila and mosquito brains. PLoS One 16(5): e0250381. PubMed ID: 33989293

    Chin, S. G., Maguire, S. E., Huoviala, P., Jefferis, G. and Potter, C. J. (2018). Olfactory neurons and brain centers directing oviposition decisions in Drosophila. Cell Rep 24(6): 1667-1678. PubMed ID: 30089274

    Lin, C. C., Riabinina, O. and Potter, C. J. (2016). Olfactory behaviors assayed by computer tracking of Drosophila in a four-quadrant olfactometer. J Vis Exp. PubMed ID: 27585032

    Lin, C. C., Prokop-Prigge, K. A., Preti, G. and Potter, C. J. (2015). Food odors trigger males to deposit a pheromone that guides aggregation and female oviposition decisions. Elife 4. PubMed ID: 26422512

    Gao, X. J., Riabinina, O., Li, J., Potter, C. J., Clandinin, T. R. and Luo, L. (2015). A transcriptional reporter of intracellular Ca(2+) in Drosophila. Nat Neurosci 18: 917-925. PubMed ID: 25961791

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

    Riabinina, O., Luginbuhl, D., Marr, E., Liu, S., Wu, M. N., Luo, L. and Potter, C. J. (2015). Improved and expanded Q-system reagents for genetic manipulations. Nat Methods. PubMed ID: 25581800

    Ronderos, D. S., Lin, C. C., Potter, C. J. and Smith, D. P. (2014). Farnesol-detecting olfactory neurons in Drosophila. J Neurosci 34: 3959-3968. PubMed ID: 24623773

  • Mohit Prasad Department of Biological Sciences, IISER Kolkata, India
    Felix, M. and Prasad, M. (2022). Mapping Asymmetry in Collective Cell Migration: Lessons from Border Cells in Drosophila Oogenesis. Methods Mol Biol 2438: 483-494. PubMed ID: 35147959
    Basu, I., Bar, S., Prasad, M. and Datta, R. (2022). Adipose deficiency and aberrant autophagy in a Drosophila model of MPS VII is corrected by pharmacological stimulators of mTOR. Biochim Biophys Acta Mol Basis Dis 1868(7): 166399. PubMed ID: 35318126

    Felix, M. and Prasad, M. (2022). Mapping Asymmetry in Collective Cell Migration: Lessons from Border Cells in Drosophila Oogenesis. Methods Mol Biol 2438: 483-494. PubMed ID: 35147959

    Sharma, A., Halder, S., Felix, M., Nisaa, K., Deshpande, G. and Prasad, M. (2018). Insulin signaling modulates border cell movement in Drosophila oogenesis. Development 145(14). PubMed ID: 29950391

  • N. G. Prasad Evolutionary Biology Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Mohali
    Singh, K., Kochar, E., Gahlot, P., Bhatt, K. and Prasad, N. G. (2021). Evolution of reproductive traits have no apparent life-history associated cost in populations of Drosophila melanogaster selected for cold shock resistance. BMC Ecol Evol 21(1): 219. PubMed ID: 34872492

    Geeta Arun, M., Agarwala, A., Syed, Z. A., Jigisha, Kashyap, M., Venkatesan, S., Chechi, T. S., Gupta, V. and Prasad, N. G. (2021). Experimental evolution reveals sex-specific dominance for surviving bacterial infection in laboratory populations of Drosophila melanogaster. Evol Lett 5(6): 657-671. PubMed ID: 34919096

    Ahlawat, N., Geeta Arun, M., Maggu, K. and Prasad, N. G. (2021). Enemies make you stronger: Coevolution between fruit fly host and bacterial pathogen increases postinfection survivorship in the host. Ecol Evol 11(14): 9563-9574. PubMed ID: 34306643

    Kapila, R., Kashyap, M., Gulati, A., Narasimhan, A., Poddar, S., Mukhopadhaya, A. and Prasad, N. G. (2021). Evolution of sex-specific heat stress tolerance and larval Hsp70 expression in populations of Drosophila melanogaster adapted to larval crowding. J Evol Biol. PubMed ID: 34197669

    Kapila, R., Kashyap, M., Poddar, S., Gangwal, S. and Prasad, N. G. G. (2021). Evolution of pathogen-specific improved survivorship post-infection in populations of Drosophila melanogaster adapted to larval crowding. PLoS One 16(4): e0250055. PubMed ID: 33852596

    Maggu, K., Ahlawat, N., Geeta Arun, M., Meena, A. and Prasad, N. G. (2020). Divergence of responses to variable socio-sexual environments in laboratory populations of Drosophila melanogaster evolving under altered operational sex ratios. Evolution. PubMed ID: 33319380

    Syed, Z. A., Gupta, V., Arun, M. G., Dhiman, A., Nandy, B. and Prasad, N. G. (2020). Absence of reproduction-immunity trade-off in male Drosophila melanogaster evolving under differential sexual selection. BMC Evol Biol 20(1): 13. PubMed ID: 31992187

    Gogna, N., Sharma, R., Gupta, V., Dorai, K. and Prasad, N. G. (2017). Evolution of the metabolome in response to selection for increased immunity in populations of Drosophila melanogaster. PLoS One 12(11): e0188089. PubMed ID: 29149207

    Syed, Z. A., Chatterjee, M., Samant, M. A. and Prasad, N. G. (2017). Reproductive isolation through experimental manipulation of sexually antagonistic coevolution in Drosophila melanogaster. Sci Rep 7(1): 3330. PubMed ID: 28611437

    Shenoi, V. N., Syed, è. A. and Prasad, N. G. (2015). Evolution of increased adult longevity in Drosophila melanogaster populations selected for adaptation to larval crowding. J Evol Biol [Epub ahead of print]. PubMed ID: 26575793

    Singh, K., Kochar, E. and Prasad, N. G. (2015). Egg Viability, Mating Frequency and Male Mating Ability Evolve in Populations of Drosophila melanogaster Selected for Resistance to Cold Shock. PLoS One 10: e0129992. PubMed ID: 26065704

    Nandy, B., Gupta, V., Udaykumar, N., Samant, M. A., Sen, S. and Prasad, N. G. (2014). Experimental evolution of female traits under different levels of intersexual conflict in Drosophila melanogaster. Evolution 68: 412-425. PubMed ID: 24117169

  • Thomas Preat Le Centre national de la recherche scientifique, Paris
    Silva, B., Mantha, O. L., Schor, J., Pascual, A., Plaçais, P. Y., Pavlowsky, A. and Preat, T. (2022). Glia fuel neurons with locally synthesized ketone bodies to sustain memory under starvation. Nat Metab 4(2): 213-224. PubMed ID: 35177854
    de Tredern, E., Rabah, Y., Pasquer, L., Minatchy, J., Placais, P. Y. and Preat, T. (2021). Glial glucose fuels the neuronal pentose phosphate pathway for long-term memory. Cell Rep 36(8): 109620. PubMed ID: 34433052

    Hadjieconomou, D., King, G., Gaspar, P., Mineo, A., Blackie, L., Ameku, T., Studd, C., de Mendoza, A., Diao, F., White, B. H., Brown, A. E. X., Placais, P. Y., Preat, T. and Miguel-Aliaga, I. (2020). Enteric neurons increase maternal food intake during reproduction. Nature 587(7834): 455-459. PubMed ID: 33116314

    Turrel, O., Rabah, Y., Placais, P. Y., Goguel, V. and Preat, T. (2020). Drosophila middle-term memory: Amnesiac is required for PKA activation in the mushroom bodies, a function modulated by Neprilysin 1. J Neurosci. PubMed ID: 32303647

    Delestro, F., Scheunemann, L., Pedrazzani, M., Tchenio, P., Preat, T. and Genovesio, A. (2020). In vivo large-scale analysis of Drosophila neuronal calcium traces by automated tracking of single somata. Sci Rep 10(1): 7153. PubMed ID: 32346011

    Silva, B., Niehage, C., Maglione, M., Hoflack, B., Sigrist, S. J., Wassmer, T., Pavlowsky, A. and Preat, T. (2020). Interactions between amyloid precursor protein-like (APPL) and MAGUK scaffolding proteins contribute to appetitive long-term memory in Drosophila melanogaster. J Neurogenet: 1-14. PubMed ID: 31965876

    Scheunemann, L., Lampin-Saint-Amaux, A., Schor, J. and Preat, T. (2019). A sperm peptide enhances long-term memory in female Drosophila. Sci Adv 5(11): eaax3432. PubMed ID: 31799390

    Hudry, B., de Goeij, E., Mineo, A., Gaspar, P., Hadjieconomou, D., Studd, C., Mokochinski, J. B., Kramer, H. B., Placais, P. Y., Preat, T. and Miguel-Aliaga, I. (2019). Sex Differences in intestinal carbohydrate metabolism promote food intake and sperm maturation. Cell 178(4): 901-918. PubMed ID: 31398343

    Dolan, M. J., Belliart-Guerin, G., Bates, A. S., Frechter, S., Lampin-Saint-Amaux, A., Aso, Y., Roberts, R. J. V., Schlegel, P., Wong, A., Hammad, A., Bock, D., Rubin, G. M., Preat, T., Placais, P. Y. and Jefferis, G. (2018). Communication from learned to innate olfactory processing centers Is required for memory retrieval in Drosophila. Neuron. PubMed ID: 30244885

    Pavlowsky, A., Schor, J., Placais, P. Y. and Preat, T. (2018). A GABAergic feedback shapes dopaminergic input on the Drosophila mushroom body to promote appetitive long-term memory. Curr Biol. Pubmed ID: 29779874

    Scheunemann, L., Placais, P. Y., Dromard, Y., Schwarzel, M. and Preat, T. (2018). Dunce phosphodiesterase acts as a checkpoint for Drosophila long-term memory in a pair of serotonergic neurons. Neuron 98(2): 350-365.e355. PubMed ID: 29673482

    Musso, P. Y., Lampin-Saint-Amaux, A., Tchenio, P. and Preat, T. (2017). Ingestion of artificial sweeteners leads to caloric frustration memory in Drosophila. Nat Commun 8(1): 1803. PubMed ID: 29180783

    Turrel, O., Goguel, V. and Preat, T. (2017). Drosophila neprilysin 1 rescues memory deficits caused by amyloid-beta peptide. J Neurosci [Epub ahead of print]. PubMed ID: 28931572

    Placais, P. Y., de Tredern, E., Scheunemann, L., Trannoy, S., Goguel, V., Han, K. A., Isabel, G. and Preat, T. (2017). Upregulated energy metabolism in the Drosophila mushroom body is the trigger for long-term memory. Nat Commun 8: 15510. PubMed ID: 28580949

  • Ken Prehoda Institute of Molecular Biology and Department of Chemistry, University of Oregon, Eugene
    Oon, C. H. and Prehoda, K. E. (2021). Phases of cortical actomyosin dynamics coupled to the neuroblast polarity cycle. Elife 10. PubMed ID: 34779402

    Holly, R. W., Jones, K. and Prehoda, K. E. (2020). A Conserved PDZ-Binding Motif in aPKC Interacts with Par-3 and Mediates Cortical Polarity. Curr Biol 30(5): 893-898. PubMed ID: 32084408

    Golub, O., Wee, B., Newman, R. A., Paterson, N. M. and Prehoda, K. E. (2017). Activation of Discs large by aPKC aligns the mitotic spindle to the polarity axis during asymmetric cell division. Elife 6. PubMed ID: 29185419

    Whitney, D. S., Peterson, F. C., Kittell, A. W., Egner, J. M., Prehoda, K. E. and Volkman, B. F. (2016). Binding of Crumbs to the Par-6 CRIB-PDZ module is regulated by Cdc42. Biochemistry 55: 1455-1461. PubMed ID: 26894406

    Bailey, M. J. and Prehoda, K. E. (2015). Establishment of par-polarized cortical domains via phosphoregulated membrane motifs. Dev Cell 35: 199-210. PubMed ID: 26481050

    Graybill, C. and Prehoda, K. E. (2014). Ordered multi-site phosphorylation of Lethal giant larvae by atypical Protein Kinase C. Biochemistry. PubMed ID: 25000553

    Lu, M. S. and Prehoda, K. E. (2013). A NudE/14-3-3 pathway coordinates dynein and the kinesin Khc73 to position the mitotic spindle. Dev Cell 26: 369-380. PubMed ID: 23987511

  • Anette Preiss Institut für Genetik, Universität Hohenheim
    Nagel, A. C., Muller, D., Zimmermann, M. and Preiss, A. (2021). The Membrane-Bound Notch Regulator Mnr Supports Notch Cleavage and Signaling Activity in Drosophila melanogaster. Biomolecules 11(11). PubMed ID: 34827670

    Nagel, A. C., Maier, D., Scharpf, J., Ketelhut, M. and Preiss, A. (2020). Limited Availability of General Co-Repressors Uncovered in an Overexpression Context during Wing Venation in Drosophila melanogaster. Genes (Basel) 11(10). PubMed ID: 32998295

    Maier, D., Nagel, A. C. and Preiss, A. (2019). Genetic interactions between Protein Kinase D and Lobe mutants during eye development of Drosophila melanogaster. Hereditas 156: 37. PubMed ID: 31889943

    Wolf, D., Smylla, T. K., Reichmuth, J., Hoffmeister, P., Kober, L., Zimmermann, M., Turkiewicz, A., Borggrefe, T., Nagel, A. C., Oswald, F., Preiss, A. and Maier, D. (2019). Nucleo-cytoplasmic shuttling of Drosophila Hairless/Su(H) heterodimer as a means of regulating Notch dependent transcription. Biochim Biophys Acta Mol Cell Res 1866(10): 1520-1532. PubMed ID: 31326540

    Smylla, T. K., Meier, M., Preiss, A. and Maier, D. (2019). The Notch repressor complex in Drosophila: in vivo analysis of Hairless mutants using overexpression experiments. Dev Genes Evol. PubMed ID: 30612166

    Bayer, F. E., Zimmermann, M., Preiss, A. and Nagel, A. C. (2018). Overexpression of the Drosophila ATR homologous checkpoint kinase Mei-41 induces a G2/M checkpoint in Drosophila imaginal tissue. Hereditas 155: 27. PubMed ID: 30202398

    Preiss, A., Nagel, A. C., Praxenthaler, H. and Maier, D. (2018). Complex genetic interactions of novel Suppressor of Hairless alleles deficient in co-repressor binding. PLoS One 13(3): e0193956. PubMed ID: 29509808

    Nagel, A. C., Auer, J. S., Schulz, A., Pfannstiel, J., Yuan, Z., Collins, C. E., Kovall, R. A. and Preiss, A. (2017). Phosphorylation of Suppressor of Hairless impedes its DNA-binding activity. Sci Rep 7(1): 11820. PubMed ID: 28928428

    Praxenthaler, H., Nagel, A. C., Schulz, A., Zimmermann, M., Meier, M., Schmid, H., Preiss, A. and Maier, D. (2017). Hairless-binding deficient Suppressor of Hairless alleles reveal Su(H) protein levels are dependent on complex formation with Hairless. PLoS Genet 13(5): e1006774. PubMed ID: 28475577

    Smylla, T.K., Preiss, A. and Maier, D. (2016). In vivo analysis of internal ribosome entry at the Hairless locus by genome engineering in Drosophila. Sci Rep 6: 34881. PubMed ID: 27713501

    Yuan, Z., Praxenthaler, H., Tabaja, N., Torella, R., Preiss, A., Maier, D. and Kovall, R. A. (2016). Structure and Function of the Su(H)-Hairless Repressor Complex, the Major Antagonist of Notch Signaling in Drosophila melanogaster. PLoS Biol 14: e1002509. PubMed ID: 27404588

  • Daven Presgraves Department of Biology, University of Rochester
    Brand, C. L., Wright, L. and Presgraves, D. C. (2019). Positive selection and functional divergence at meiosis genes that mediate crossing over across the Drosophila phylogeny. G3 (Bethesda). PubMed ID: 31362974

    Meiklejohn, C. D., Landeen, E. L., Gordon, K. E., Rzatkiewicz, T., Kingan, S. B., Geneva, A. J., Vedanayagam, J. P., Muirhead, C. A., Garrigan, D., Stern, D. L. and Presgraves, D. C. (2018). Gene flow mediates the role of sex chromosome meiotic drive during complex speciation. Elife 7. PubMed ID: 30543325

    Landeen, E. L., Muirhead, C. A., Wright, L., Meiklejohn, C. D. and Presgraves, D. C. (2016). Sex chromosome-wide transcriptional suppression and compensatory cis-regulatory evolution mediate gene expression in the Drosophila male germline. PLoS Biol 14: e1002499. PubMed ID: 27404402

    Brand, C. L., Larracuente, A. M. and Presgraves, D. C. (2015). Origin, evolution, and population genetics of the selfish Segregation Distorter gene duplication in European and African populations of Drosophila melanogaster. Evolution 69: 1271-1283. PubMed ID: 25828399

    Tang, S. and Presgraves, D. C. (2015). Lineage-Specific Evolution of the Complex Nup160 Hybrid Incompatibility Between Drosophila melanogaster and Its Sister Species. Genetics 200: 1245-1254. PubMed ID: 26022241

    Garrigan, D., Kingan, S. B., Geneva, A. J., Vedanayagam, J. P. and Presgraves, D. C. (2015). Genome Diversity and Divergence in Drosophila mauritiana: Multiple Signatures of Faster X Evolution. Genome Biol Evol 7: 1331. PubMed ID: 25991815

    Brand, C. L., Larracuente, A. M. and Presgraves, D. C. (2015). Origin, evolution, and population genetics of the selfish Segregation Distorter gene duplication in European and African populations of Drosophila melanogaster. Evolution 69: 1271-1283. PubMed ID: 25828399

    Garrigan, D., Kingan, S. B., Geneva, A. J., Vedanayagam, J. P. and Presgraves, D. C. (2014). Genome diversity and divergence in Drosophila mauritiana: multiple signatures of faster X evolution. Genome Biol Evol 6: 2444-2458. PubMed ID: 25193308

  • Jeffrey Price School of Biological Sciences, University of Missouri-Kansas City
    Wang, J., Fan, J. Y., Zhao, Z., Dissel, S. and Price, J. (2022). DBT affects sleep in both circadian and non-circadian neurons. PLoS Genet 18(2): e1010035. PubMed ID: 35139068

    Venkatesan, A., Fan, J. Y., Bouyain, S. and Price, J. L. (2019). The circadian tau mutation in Casein Kinase 1 is part of a larger domain that can be mutated to shorten circadian period. Int J Mol Sci 20(4). PubMed ID: 30769795

    Venkatesan, A., Fan, J. Y., Nauman, C. and Price, J. L. (2015). A Doubletime nuclear localization signal mediates an interaction with Bride of Doubletime to promote circadian function. J Biol Rhythms [Epub ahead of print]. PubMed ID: 26082158

    Fan, J. Y., Means, J. C., Bjes, E. S. and Price, J. L. (2015). Drosophila DBT autophosphorylation of its C terminal domain antagonized by SPAG and involved in UV-induced apoptosis. Mol Cell Biol. PubMed ID: 25939385

    Means, J.C., Venkatesan, A., Gerdes, B., Fan, J.Y., Bjes, E.S. and Price, J.L. (2015). Drosophila Spaghetti and Doubletime link the circadian clock and light to caspases, apoptosis and tauopathy. PLoS Genet 11: e1005171. PubMed ID: 25951229

    He, C., Yang, Y., Zhang, M., Price, J. L. and Zhao, Z. (2013). Regulation of Sleep by Neuropeptide Y-Like System in Drosophila melanogaster. PLoS One 8: e74237. PubMed ID: 24040211

    Yu, W., Zheng, H., Price, J. L. and Hardin, P. E. (2009). DOUBLETIME plays a noncatalytic role to mediate CLOCK phosphorylation and repress CLOCK-dependent transcription within the Drosophila circadian clock. Mol Cell Biol 29: 1452-1458. PubMed ID: 19139270

    Fan, J. Y., Preuss, F., Muskus, M. J., Bjes, E. S. and Price, J. L. (2009). Drosophila and vertebrate casein kinase Idelta exhibits evolutionary conservation of circadian function. Genetics 181: 139-152. PubMed ID: 18957703

  • Thomas Price Institute of Integrative Biology, University of Liverpool
    Walsh, B. S., Parratt, S. R., Snook, R. R., Bretman, A., Atkinson, D. and Price, T. A. R. (2022). Female fruit flies cannot protect stored sperm from high temperature damage. J Therm Biol 105: 103209. PubMed ID: 35393050

    Heys, C., Lize, A., Lewis, Z. and Price, T. A. R. (2020). Drosophila sexual attractiveness in older males is mediated by their microbiota. Microorganisms 8(2). PubMed ID: 31991698

    Verspoor, R. L., Smith, J. M. L., Mannion, N. L. M., Hurst, G. D. D. and Price, T. A. R. (2018). Strong hybrid male incompatibilities impede the spread of a selfish chromosome between populations of a fly. Evol Lett 2(3): 169-179. PubMed ID: 30283674

    Giraldo-Perez, P., Herrera, P., Campbell, A., Taylor, M. L., Skeats, A., Aggio, R., Wedell, N. and Price, T. A. (2015). Winter is coming: hibernation reverses the outcome of sperm competition in a fly. J Evol Biol. PubMed ID: 26565889

    Verspoor, R. L., Heys, C. and Price, T. A. (2015). Dyeing insects for behavioral assays: the mating behavior of anesthetized Drosophila. J Vis Exp. PubMed ID: 25938821

    Maguire, C. P., Lize, A. and Price, T. A. (2015). Assessment of rival males through the use of multiple sensory cues in the fruitfly Drosophila pseudoobscura. PLoS One 10: e0123058. PubMed ID: 25849643

    Bretman, A., Lize, A., Walling, C. A. and Price, T. A. (2014). The heritability of mating behaviour in a fly and its plasticity in response to the threat of sperm competition. PLoS One 9: e90236. PubMed ID: 24587294

    Fisher, D. N., Doff, R. J. and Price, T. A. (2013). True polyandry and pseudopolyandry: why does a monandrous fly remate? BMC Evol Biol 13: 157. PubMed ID: 23885723

    Price, T. A., Lize, A., Marcello, M. and Bretman, A. (2012). Experience of mating rivals causes males to modulate sperm transfer in the fly Drosophila pseudoobscura. J Insect Physiol 58: 1669-1675. PubMed ID: 23085556

  • Marina Prigol Universidade Federal do Pampa, Brazil
    Poetini, M. R., Musachio, E. A. S., Araujo, S. M., Bortolotto, V. C., Meichtry, L. B., Silva, N. C., Janner, D. E., La Rosa Novo, D., Mesko, M. F., Roehrs, R., Ramborger, B. P. and Prigol, M. (2022). Improvement of non-motor and motor behavioral alterations associated with Parkinson-like disease in Drosophila melanogaster: Comparative effects of treatments with hesperidin and L-dopa. Neurotoxicology 89: 174-183. PubMed ID: 35167856

    Poetini, M. R., Musachio, E. A. S., Araujo, S. M., Almeida, F. P., Dahleh, M. M. M., Bortolotto, V. C., Janner, D. E., Pinheiro, F. C., Ramborger, B. P., Roehrs, R., La Rosa Novo, D., Mesko, M. F., Guerra, G. P. and Prigol, M. (2021). Iron overload during the embryonic period develops hyperactive like behavior and dysregulation of biogenic amines in Drosophila melanogaster. Dev Biol 475: 80-90. PubMed ID: 33741348

    Meichtry, L. B., Poetini, M. R., Dahleh, M. M. M., Araujo, S. M., Musachio, E. A. S., Bortolotto, V. C., de Freitas Couto, S., Somacal, S., Emanuelli, T., Gayer, M. C., Roehrs, R., Guerra, G. P. and Prigol, M. (2020). Addition of Saturated and Trans-fatty Acids to the Diet Induces Depressive and Anxiety-like Behaviors in Drosophila melanogaster. Neuroscience 443: 164-175. PubMed ID: 32738432

    Araujo, S. M., Bortolotto, V. C., Poetini, M. R., Dahleh, M. M. M., Couto, S. F., Pinheiro, F. C., Meichtry, L. B., Musachio, E. A. S., Ramborger, B. P., Roehrs, R., Guerra, G. P. and Prigol, M. (2021). gamma-Oryzanol produces an antidepressant-like effect in a chronic unpredictable mild stress model of depression in Drosophila melanogaster. Stress 24(3): 282-293. PubMed ID: 32723199

  • Daniel Promislow Department of Genetics, University of Georgia, Athens
    Zhao, X., Golic, F. T., Harrison, B. R., Manoj, M., Hoffman, E. V., Simon, N., Johnson, R., MacCoss, M. J., McIntyre, L. M. and Promislow, D. E. L. (2022). The metabolome as a biomarker of aging in Drosophila melanogaster. Aging Cell: e13548. PubMed ID: 35019203

    Harrison, B. R., Hoffman, J. M., Samuelson, A., Raftery, D. and Promislow, D. E. L. (2021). Modular Evolution of the Drosophila Metabolome. Mol Biol Evol. PubMed ID: 34662414

    Jin, K., Wilson, K. A., Beck, J. N., Nelson, C. S., Brownridge, G. W., 3rd, Harrison, B. R., Djukovic, D., Raftery, D., Brem, R. B., Yu, S., Drton, M., Shojaie, A., Kapahi, P. and Promislow, D. (2020). Genetic and metabolomic architecture of variation in diet restriction-mediated lifespan extension in Drosophila. PLoS Genet 16(7): e1008835. PubMed ID: 32644988

    Harrison, B. R., Wang, L., Gajda, E., Hoffman, E. V., Chung, B. Y., Pletcher, S. D., Raftery, D. and Promislow, D. E. L. (2020). The metabolome as a link in the genotype-phenotype map for peroxide resistance in the fruit fly, Drosophila melanogaster. BMC Genomics 21(1): 341. PubMed ID: 32366330

    Arbuthnott, D., Fedina, T.Y., Pletcher, S.D. and Promislow, D.E. (2017). Mate choice in fruit flies is rational and adaptive. Nat Commun 8: 13953. PubMed ID: 28094789

    Harvanek, Z. M., Mourao, M. A., Schnell, S. and Pletcher, S. D. (2016). A computational approach to studying ageing at the individual level. Proc Biol Sci 283. PubMed ID: 26865300

    Arbuthnott, D., Levin, T. C. and Promislow, D. E. (2015). The impacts of Wolbachia and the microbiome on mate choice in Drosophila melanogaster. J Evol Biol [Epub ahead of print]. PubMed ID: 26548557

    Laye, M. J., Tran, V., Jones, D. P., Kapahi, P. and Promislow, D. E. (2015). The effects of age and dietary restriction on the tissue-specific metabolome of Drosophila. Aging Cell [Epub ahead of print]. PubMed ID: 26085309

    Hoffman, J. M., Soltow, Q. A., Li, S., Sidik, A., Jones, D. P. and Promislow, D. E. (2014). Effects of age, sex, and genotype on high-sensitivity metabolomic profiles in the fruit fly, Drosophila melanogaster. Aging Cell. PubMed ID: 24636523

    Linder, J. E. and Promislow, D. E. (2009). Cross-generational fitness effects of infection in Drosophila melanogaster. Fly (Austin) 3: 143-150. PubMed ID: 19242114

  • Andreas Prokop Manchester University
    Hahn, I., Voelzmann, A., Parkin, J., Fulle, J. B., Slater, P. G., Lowery, L. A., Sanchez-Soriano, N. and Prokop, A. (2021). Tau, XMAP215/Msps and Eb1 co-operate interdependently to regulate microtubule polymerisation and bundle formation in axons. PLoS Genet 17(7): e1009647. PubMed ID: 34228717

    Qu, Y., Hahn, I., Lees, M., Parkin, J., Voelzmann, A., Dorey, K., Rathbone, A., Friel, C. T., Allan, V. J., Okenve-Ramos, P., Sanchez-Soriano, N. and Prokop, A. (2019). Efa6 protects axons and regulates their growth and branching by inhibiting microtubule polymerisation at the cortex. Elife 8. PubMed ID: 31718774

    Qu, Y., Hahn, I., Webb, S., Pearce, S. P. and Prokop, A. (2016). Periodic actin structures in neuronal axons are required to maintain microtubules. Mol Biol Cell [Epub ahead of print]. PubMed ID: 27881663

    Voelzmann, A., Okenve-Ramos, P., Qu, Y., Chojnowska-Monga, M., Del Caño-Espinel, M., Prokop, A. and Sanchez-Soriano, N. (2016). Tau and spectraplakins promote synapse formation and maintenance through Jun kinase and neuronal trafficking. Elife 5. PubMed ID: 27501441

    Hahn, I., Ronshaugen, M., Sanchez-Soriano, N. and Prokop, A. (2016). Functional and Genetic Analysis of Spectraplakins in Drosophila. Methods Enzymol 569: 373-405. PubMed ID: 26778568

    Beaven, R., Dzhindzhev, N. S., Qu, Y., Hahn, I., Dajas-Bailador, F., Ohkura, H. and Prokop, A. (2015). Drosophila CLIP-190 and mammalian CLIP-170 display reduced microtubule plus end association in the nervous system. Mol Biol Cell. PubMed ID: 25694447

    Stephan, D., Sanchez-Soriano, N., Loschek, L.F., Gerhards, R., Gutmann, S., Storchova, Z., Prokop, A., and Kadow, I.C. (2012). Drosophila Psidin Regulates Olfactory Neuron Number and Axon Targeting through Two Distinct Molecular Mechanisms. J. Neurosci. 32: 16080-16094. PubMed ID: 23152593

    Alves-Silva, J., Sánchez-Soriano, N., Beaven, R., Klein, M., Parkin, J., Millard, T., Bellen, H., Venken, K. J. T., Ballestrem, C., Kammerer, R.A. and Prokop, A. (2012). Spectraplakins promote microtubule-mediated axonal growth by functioning as structural microtubule-associated proteins and EB1-dependent +TIPs (Tip Interacting Proteins). J. Neurosci. 32: 9143-58. PubMed ID: 22764224

  • Benjamin Prud’homme Institut de Biologie du Developpement de Marseille
    Galouzis, C. C. and Prud'homme, B. (2021). Transvection regulates the sex-biased expression of a fly X-linked gene. Science 371(6527): 396-400. PubMed ID: 33479152

    Le Poul, Y., Xin, Y., Ling, L., Muhling, B., Jaenichen, R., Horl, D., Bunk, D., Harz, H., Leonhardt, H., Wang, Y., Osipova, E., Museridze, M., Dharmadhikari, D., Murphy, E., Rohs, R., Preibisch, S., Prud'homme, B. and Gompel, N. (2020). Regulatory encoding of quantitative variation in spatial activity of a Drosophila enhancer. Sci Adv 6(49). PubMed ID: 33268361

    Paris, M., Boyer, R., Jaenichen, R., Wolf, J., Karageorgi, M., Green, J., Cagnon, M., Parinello, H., Estoup, A., Gautier, M., Gompel, N. and Prud'homme, B. (2020). Near-chromosome level genome assembly of the fruit pest Drosophila suzukii using long-read sequencing. Sci Rep 10(1): 11227. PubMed ID: 32641717

  • Stefan Pulver University of St Andrews, Fife United Kingdom
    Jonaitis, J., MacLeod, J. and Pulver, S. R. (2022). Localization of muscarinic acetylcholine receptor-dependent rhythm-generating modules in the Drosophila larval locomotor network. J Neurophysiol 127(4): 1098-1116. PubMed ID: 35294308

    Zeng, X., Komanome, Y., Kawasaki, T., Inada, K., Jonaitis, J., Pulver, S. R., Kazama, H. and Nose, A. (2021). An electrically coupled pioneer circuit enables motor development via proprioceptive feedback in Drosophila embryos. Curr Biol 31(23): 5327-5340 e5325. PubMed ID: 34666002

    Hiramoto, A., Jonaitis, J., Niki, S., Kohsaka, H., Fetter, R. D., Cardona, A., Pulver, S. R. and Nose, A. (2021). Regulation of coordinated muscular relaxation in Drosophila larvae by a pattern-regulating intersegmental circuit. Nat Commun 12(1): 2943. PubMed ID: 34011945

    Murawski, C., Pulver, S. R. and Gather, M. C. (2020). Segment-specific optogenetic stimulation in Drosophila melanogaster with linear arrays of organic light-emitting diodes. Nat Commun 11(1): 6248. PubMed ID: 33288763

    Morton, A., Murawski, C., Pulver, S. R. and Gather, M. C. (2016). High-brightness organic light-emitting diodes for optogenetic control of Drosophila locomotor behaviour. Sci Rep 6: 31117. PubMed ID: 27484401

  • Giorgos Pyrowolakis Institut für Biologie I, Lehrstuhl für Entwicklungsbiologie, Freiburg
    Chayengia, M., Veikkolainen, V., Jevtic, M. and Pyrowolakis, G. (2019). Sequence environment of BMP-dependent activating elements controls transcriptional responses to Dpp signaling in Drosophila. Development. PubMed ID: 31110028

    Norman, M., Vuilleumier, R., Springhorn, A., Gawlik, J. and Pyrowolakis, G. (2016). Pentagone internalises glypicans to fine-tune multiple signalling pathways. Elife [Epub ahead of print]. PubMed ID: 27269283

    Charbonnier, E., Fuchs, A., Cheung, L.S., Chayengia, M., Veikkolainen, V., Shvartsman, S.Y. and Pyrowolakis, G. (2015). BMP-dependent gene repression cascade in drosophila eggshell patterning. Dev Biol [Epub ahead of print]. PubMed ID: 25704512

    Marmion, R. A., Jevtic, M., Springhorn, A., Pyrowolakis, G. and Yakoby, N. (2012). The Drosophila BMPRII, wishful thinking, is required for eggshell patterning. Dev Biol. PubMed ID: 23274688

    Fuchs, A., Cheung, L. S., Charbonnier, E., Shvartsman, S. Y. and Pyrowolakis, G. (2012). Transcriptional interpretation of the EGF receptor signaling gradient. Proc Natl Acad Sci U S A 109: 1572-1577. PubMed ID: 22307613

    Vuilleumier, R., Affolter, M. and Pyrowolakis, G. (2011). Pentagone: patrolling BMP morphogen signaling. Fly (Austin) 5: 210-214. PubMed ID: 21406970
  • Elzbieta Pyza Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
    Krzeptowski, W., Walkowicz, L., Krzeptowska, E., Motta, E., Witek, K., Szramel, J., Al Abaquita, T., Baster, Z., Rajfur, Z., Rosato, E., Stratoulias, V., Heino, T. I. and Pyza, E. M. (2021). Mesencephalic Astrocyte-Derived Neurotrophic Factor Regulates Morphology of Pigment-Dispersing Factor-Positive Clock Neurons and Circadian Neuronal Plasticity in Drosophila melanogaster. Front Physiol 12: 705183. PubMed ID: 34646147

    Walkowicz, L., Krzeptowski, W., Krzeptowska, E., Warzecha, K., Salek, J., Gorska-Andrzejak, J. and Pyza, E. (2021). Glial expression of DmMANF is required for the regulation of activity, sleep and circadian rhythms in the visual system of Drosophila melanogaster. Eur J Neurosci. PubMed ID: 33666288

    Damulewicz, M., Woznicka, O., Jasinska, M. and Pyza, E. (2020). CRY-dependent plasticity of tetrad presynaptic sites in the visual system of Drosophila at the morning peak of activity and sleep. Sci Rep 10(1): 18161. PubMed ID: 33097794

    Doktor, B., Damulewicz, M. and Pyza, E. (2019). Effects of MUL1 and PARKIN on the circadian clock, brain and behaviour in Drosophila Parkinson's disease models. BMC Neurosci 20(1): 24. PubMed ID: 31138137

    Doktor, B., Damulewicz, M. and Pyza, E. (2019). Overexpression of Mitochondrial Ligases Reverses Rotenone-Induced Effects in a Drosophila Model of Parkinson's Disease. Front Neurosci 13: 94. PubMed ID: 30837828




  • Return to The Interactive Fly