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


  • Mariano Labrador Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville
    Amankwaa, B., Schoborg, T. and Labrador, M. (2022). Drosophila insulator proteins exhibit in vivo liquid-liquid phase separation properties. Life Sci Alliance 5(12). PubMed ID: 35853678

    Stow, E. C., Simmons, J. R., An, R., Schoborg, T. A., Davenport, N. M. and Labrador, M. (2022). A Drosophila insulator interacting protein suppresses enhancer-blocking function and modulates replication timing. Gene 819: 146208. PubMed ID: 35092858

    Hsu, S. J., Stow, E. C., Simmons, J. R., Wallace, H. A., Lopez, A. M., Stroud, S. and Labrador, M. (2020). Mutations in the insulator protein Suppressor of Hairy wing induce genome instability. Chromosoma 129(3-4): 255-274. PubMed ID: 33140220

    Asadzadeh, J., Neligan, N., Kramer, S.G. and Labrador, J.P. (2016). Tinman regulates NetrinB in the cardioblasts of the Drosophila dorsal vessel. PLoS One 11: e0148526. PubMed ID: 26840059

    Asadzadeh, J., Neligan, N., Canabal-Alvear, J. J., Daly, A. C., Kramer, S. G. and Labrador, J. P. (2015). The Unc-5 Receptor Is Directly Regulated by Tinman in the Developing Drosophila Dorsal Vessel. PLoS One 10: e0137688. PubMed ID: 26356221

    Hsu, S.J., Plata, M.P., Ernest, B., Asgarifar, S. and Labrador, M. (2015). The insulator protein Suppressor of Hairy wing is required for proper ring canal development during oogenesis in Drosophila. Dev Biol [Epub ahead of print]. PubMed ID: 25882370

    Schoborg, T. and Labrador, M. (2014). Expanding the roles of chromatin insulators in nuclear architecture, chromatin organization and genome function. Cell Mol Life Sci 71: 4089-4113. PubMed ID: 25012699

    Zarin, A. A., Asadzadeh, J., Hokamp, K., McCartney, D., Yang, L., Bashaw, G. J. and Labrador, J. P. (2014). A Transcription Factor Network Coordinates Attraction, Repulsion, and Adhesion Combinatorially to Control Motor Axon Pathway Selection. Neuron. PubMed ID: 24560702

    Schoborg, T., Kuruganti, S., Rickels, R. and Labrador, M. (2013). The Drosophila gypsy Insulator Supports Transvection in the Presence of the vestigial Enhancer. PLoS One 8: e81331. PubMed ID: 24236213

    Schoborg, T., Rickels, R., Barrios, J. and Labrador, M. (2013). Chromatin insulator bodies are nuclear structures that form in response to osmotic stress and cell death. J Cell Biol 202: 261-276. PubMed ID: 23878275

  • Andreas Ladurner University of Munich
    Peleg, S., Feller, C., Forne, I., Schiller, E., Sévin, D.C., Schauer, T., Regnard, C., Straub, T., Prestel, M., Klima, C., Schmitt Nogueira, M., Becker, L., Klopstock, T., Sauer, U., Becker, P.B., Imhof, A. and Ladurner, A.G. (2016). Life span extension by targeting a link between metabolism and histone acetylation in Drosophila. EMBO Rep [Epub ahead of print]. PubMed ID: 26781291

    Schauer, T., Schwalie, P. C., Handley, A., Margulies, C. E., Flicek, P. and Ladurner, A. G. (2013). CAST-ChIP Maps Cell-Type-Specific Chromatin States in the Drosophila Central Nervous System. Cell Rep. PubMed ID: 24095734

    Czarna, A., Berndt, A., Singh, H. R., Grudziecki, A., Ladurner, A. G., Timinszky, G., Kramer, A. and Wolf, E. (2013). Structures of Drosophila cryptochrome and mouse cryptochrome1 provide insight into circadian function. Cell 153: 1394-1405. PubMed ID: 23746849

    Murawska, M., Hassler, M., Renkawitz-Pohl, R., Ladurner, A. and Brehm, A. (2011). Stress-induced PARP activation mediates recruitment of Drosophila Mi-2 to promote heat shock gene expression. PLoS Genet 7: e1002206. PubMed ID: 21829383

  • Eric Lai Developmental Biology Program, Memorial Sloan-Kettering Cancer Center, New York
    Lee, S., Chen, Y. C., Gillen, A. E., Taliaferro, J. M., Deplancke, B., Li, H. and Lai, E. C. (2022). Diverse cell-specific patterns of alternative polyadenylation in Drosophila. Nat Commun 13(1): 5372. PubMed ID: 36100597

    Vedanayagam, J., Lin, C. J. and Lai, E. C. (2021). Rapid evolutionary dynamics of an expanding family of meiotic drive factors and their hpRNA suppressors. Nat Ecol Evol 5(12): 1613-1623. PubMed ID: 34862477

    Joseph, B., Scala, C., Kondo, S. and Lai, E. C. (2022). Molecular and genetic dissection of recursive splicing. Life Sci Alliance 5(1). PubMed ID: 34759052
    Garaulet, D. L., Moro, A. and Lai, E. C. (2021). A double-negative gene regulatory circuit underlies the virgin behavioral state. Cell Rep 36(1): 109335. PubMed ID: 34233178

    Lee, S., Wei, L., Zhang, B., Goering, R., Majumdar, S., Wen, J., Taliaferro, J. M. and Lai, E. C. (2021). ELAV/Hu RNA binding proteins determine multiple programs of neural alternative splicing. PLoS Genet 17(4): e1009439. PubMed ID: 33826609

    Kan, L., Ott, S., Joseph, B., Park, E. S., Dai, W., Kleiner, R. E., Claridge-Chang, A. and Lai, E. C. (2021). A neural m(6)A/Ythdf pathway is required for learning and memory in Drosophila. Nat Commun 12(1): 1458. PubMed ID: 33674589

    Bejarano, F., Chang, C. H., Sun, K., Hagen, J. W., Deng, W. M. and Lai, E. C. (2021). A comprehensive in vivo screen for anti-apoptotic miRNAs indicates broad capacities for oncogenic synergy. Dev Biol 475: 10-20. PubMed ID: 33662357

    Wei, L., Lee, S., Majumdar, S., Zhang, B., Sanfilippo, P., Joseph, B., Miura, P., Soller, M. and Lai, E. C. (2020). Overlapping Activities of ELAV/Hu Family RNA Binding Proteins Specify the Extended Neuronal 3' UTR Landscape in Drosophila. Mol Cell 80(1): 140-155.e146. PubMed ID: 33007254

    Mazaud, D., Kottler, B., Goncalves-Pimentel, C., Proelss, S., Tuchler, N., Deneubourg, C., Yuasa, Y., Diebold, C., Jungbluth, H., Lai, E. C., Hirth, F., Giangrande, A. and Fanto, M. (2019). Transcriptional regulation of the Glutamate/GABA/Glutamine cycle in adult glia controls motor activity and seizures in Drosophila. J Neurosci. PubMed ID: 31064860

    Bag, I., Dale, R. K., Palmer, C. and Lei, E. P. (2019). The zinc-finger protein CLAMP promotes gypsy chromatin insulator function in Drosophila. J Cell Sci. PubMed ID: 30718365

    Lin, C. J., Hu, F., Dubruille, R., Vedanayagam, J., Wen, J., Smibert, P., Loppin, B. and Lai, E. C. (2018). The hpRNA/RNAi pathway is essential to resolve intragenomic conflict in the Drosophila male germline. Dev Cell 46(3): 316-326.e315. PubMed ID: 30086302

    Joseph, B., Kondo, S. and Lai, E. C. (2018). Short cryptic exons mediate recursive splicing in Drosophila. Nat Struct Mol Biol. PubMed ID: 29632374

  • Zhi-Chun Lai Department of Biology, Penn State
    Matsui, Y., Zhang, Y., Paulson, R. F. and Lai, Z. C. (2018). Dual role of a C-terminally truncated isoform of large tumor suppressor kinase 1 in the regulation of Hippo signaling and tissue growth. DNA Cell Biol. PubMed ID: 30461308

    Zhang, Y. and Lai, Z. C. (2013). Mob as tumor suppressor is regulated by bantam microRNA through a feedback loop for tissue growth control. Biochem Biophys Res Commun 439: 438-442. PubMed ID: 24016667

    Zhang, Y. and Lai, Z. C. (2013). Mob as tumor suppressor is regulated by bantam microRNA through a feedback loop for tissue growth control. Biochem Biophys Res Commun. PubMed ID: 24016667

    Deng, Y., Matsui, Y., Zhang, Y., Lai, Z. C. (2013) Hippo activation through homodimerization and membrane association for growth inhibition and organ size control. Dev Biol 375: 152-159. PubMed ID: 23298890

    Ye, X., Deng, Y., and Z.-C. Lai. 2012. Akt is negatively regulated by Hippo signaling for growth inhibition in Drosophila. Dev. Biol. 369: 115-123. PubMed ID: 22732571

    Ho, L.-L., X. Wei, T. Shimizu, and Z.-C. Lai. 2010. Mob as tumor suppressor is activated at the cell membrane to control tissue growth and organ size in Drosophila. Dev. Biol 337: 274-283. PubMed Citation: 19913529

  • Subhash Chandra Lakhotia Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University
    Singh, G., Chakraborty, S. and Lakhotia, S. C. (2022). Elevation of major constitutive heat shock proteins is heat shock factor independent and essential for establishment and growth of Lgl loss and Yorkie gain-mediated tumors in Drosophila. Cell Stress Chaperones. PubMed ID: 35704239

    Ray, M. and Lakhotia, S. C. (2019). Activated Ras/JNK driven Dilp8 in imaginal discs adversely affects organismal homeostasis during early pupal stage in Drosophila, a new checkpoint for development. Dev Dyn. PubMed ID: 31415125

    Singh, A.K. and Lakhotia, S.C. (2015). The hnRNP A1 homolog Hrb87F/Hrp36 is important for telomere maintenance in Drosophila melanogaster. Chromosoma [Epub ahead of print]. PubMed ID: 26373285

    Singh, A. K. and Lakhotia, S. C. (2015). Dynamics of hnRNPs and omega speckles in normal and heat shocked live cell nuclei of Drosophila melanogaster. Chromosoma. PubMed ID: 25663367

    Singh, A. K. and Lakhotia, S. C. (2012). The hnRNP A1 homolog Hrp36 is essential for normal development, female fecundity, omega speckle formation and stress tolerance in Drosophila melanogaster. J Biosci 37: 659-678. PubMed ID: 22922191

    Lakhotia, S. C., Mallik, M., Singh, A. K. and Ray, M. (2012). The large noncoding hsromega-n transcripts are essential for thermotolerance and remobilization of hnRNPs, HP1 and RNA polymerase II during recovery from heat shock in Drosophila. Chromosoma 121: 49-70. PubMed ID: 21913129

  • Chanderkala Lambhod Department of Genetics, Maharshi Dayanand University, Rohtak, India
    Parkash, R., Singh, D. and Lambhod, C. (2013). Sex-specific differences in desiccation resistance and the use of energy metabolites as osmolytes in Drosophila melanogaster flies acclimated to dehydration stress. J Comp Physiol B. PubMed ID: 24292242

    Parkash, R., Singh, D. and Lambhod, C. (2013). Divergent strategies for adaptations to stress resistance in two tropical Drosophila species: effects of developmental acclimation for D. bipectinata and the invasive species - D. malerkotliana. J Exp Biol. PubMed ID: 24265421

    Parkash, R., Aggarwal, D. D., Lambhod, C. and Singh, D. (2013). Divergence of water balance mechanisms and acclimation potential in body color morphs of Drosophila ananassae. J Exp Zool A Ecol Genet Physiol. PubMed ID: 24167067

  • Arthur D. Lander Department of Developmental and Cell Biology, University of California, Irvine
    Zhu, Y., Qiu, Y., Chen, W., Nie, Q. and Lander, A. D. (2020). Scaling a Dpp Morphogen Gradient through Feedback Control of Receptors and Co-receptors. Dev Cell 53(6): 724-739 e714. PubMed ID: 32574592

    Lander, A. D., Nie, Q., Sanchez-Tapia, C., Simonyan, A. and Wan, F. Y. M. (2020). Regulatory feedback on receptor and non-receptor synthesis for robust signaling. Dev Dyn 249(3): 383-409. PubMed ID: 32034817

    Newkirk, D. A., Chen, Y. Y., Chien, R., Zeng, W., Biesinger, J., Flowers, E., Kawauchi, S., Santos, R., Calof, A. L., Lander, A. D., Xie, X. and Yokomori, K. (2017). The effect of Nipped-B-like (Nipbl) haploinsufficiency on genome-wide cohesin binding and target gene expression: modeling Cornelia de Lange syndrome. Clin Epigenetics 9: 89. PubMed ID: 28855971

    Wortman, J. C., Nahmad, M., Zhang, P. C., Lander, A. D. and Yu, C. C. (2017). Expanding signaling-molecule wavefront model of cell polarization in the Drosophila wing primordium. PLoS Comput Biol 13(7): e1005610. PubMed ID: 28671940

  • Matthias Landgraf Cambridge Neuroscience
    Wang, Q. Q., You, D. D. and Liu, J. L. (2022). Cytoophidia Maintain the Integrity of Drosophila Follicle Epithelium. Int J Mol Sci 23(23). PubMed ID: 36499609

    Giachello, C. N. G., Fan, Y. N., Landgraf, M. and Baines, R. A. (2021). Nitric oxide mediates activity-dependent change to synaptic excitation during a critical period in Drosophila. Sci Rep 11(1): 20286. PubMed ID: 34645891

    Dhawan, S., Myers, P., Bailey, D. M. D., Ostrovsky, A. D., Evers, J. F. and Landgraf, M. (2021). Reactive Oxygen Species Mediate Activity-Regulated Dendritic Plasticity Through NADPH Oxidase and Aquaporin Regulation. Front Cell Neurosci 15: 641802. PubMed ID: 34290589

    Valdes-Aleman, J., Fetter, R. D., Sales, E. C., Heckman, E. L., Venkatasubramanian, L., Doe, C. Q., Landgraf, M., Cardona, A. and Zlatic, M. (2020). Comparative Connectomics Reveals How Partner Identity, Location, and Activity Specify Synaptic Connectivity in Drosophila. Neuron. PubMed ID: 33120017

    Oswald, M. C. W., Brooks, P. S., Zwart, M. F., Mukherjee, A., West, R. J. H., Giachello, C. N. G., Morarach, K., Baines, R. A., Sweeney, S. T. and Landgraf, M. (2018). Reactive oxygen species regulate activity-dependent neuronal plasticity in Drosophila. Elife 7. PubMed ID: 30540251

    Zwart, M.F., Pulver, S,R., Truman, J.W., Fushiki, A., Cardona, A. and Landgraf, M. (2016). Selective inhibition mediates the sequential recruitment of motor pools. Neuron [Epub ahead of print]. PubMed ID: 27427461

    Peco, E., Davla, S., Camp, D., Stacey, S., Landgraf, M. and van Meyel, D. (2016). Drosophila astrocytes cover specific territories of CNS neuropil and are instructed to differentiate by Prospero, a key effector of Notch. Development [Epub ahead of print]. PubMed ID: 26893340

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

    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

    Couton, L., Mauss, A. S., Yunusov, T., Diegelmann, S., Evers, J. F. and Landgraf, M. (2015). Development of connectivity in a motoneuronal network in Drosophila larvae. Curr Biol 25: 568-576. PubMed ID: 25702582

    Zwart, M. F., Randlett, O., Evers, J. F. and Landgraf, M. (2013). Dendritic growth gated by a steroid hormone receptor underlies increases in activity in the developing Drosophila locomotor system. Proc Natl Acad Sci U S A. PubMed ID: 24043825

  • Gernot Längst Department of Biochemistry, University of Regensburg
    Diermeier, S. D., Schubert, T. and Langst, G. (2014). Deep Sequencing of Small Chromatin-Associated RNA: Isolation and Library Preparation. Methods Mol Biol 1094: 343-353. PubMed ID: 24163001

    Schubert, T., Langst, G. (2013) Changes in higher order structures of chromatin by RNP complexes. RNA Biol 10. PubMed ID: 23353578

    Schubert, T., Pusch, M. C., Diermeier, S., Benes, V., Kremmer, E., Imhof, A. and Langst, G. (2012). Df31 protein and snoRNAs maintain accessible higher-order structures of chromatin. Mol Cell 48: 434-444. PubMed ID: 23022379

  • Patrick Laprise Laval University Cancer Research Center, Quebec
    Biehler, C., Rothenberg, K. E., Jette, A., Gaude, H. M., Fernandez-Gonzalez, R. and Laprise, P. (2021). Pak1 and PP2A antagonize aPKC function to support cortical tension induced by the Crumbs-Yurt complex. Elife 10. PubMed ID: 34212861

    Biehler, C., Wang, L. T., Sevigny, M., Jette, A., Gamblin, C. L., Catterall, R., Houssin, E., McCaffrey, L. and Laprise, P. (2020). Girdin is a component of the lateral polarity protein network restricting cell dissemination. PLoS Genet 16(3): e1008674. PubMed ID: 32196494

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

    Gareau, C., Houssin, E., Martel, D., Coudert, L., Mellaoui, S., Huot, M. E., Laprise, P. and Mazroui, R. (2013). Characterization of fragile X mental retardation protein recruitment and dynamics in Drosophila stress granules. PLoS One 8: e55342. PubMed ID: 23408971

    Chartier, F. J., Hardy, E. J. and Laprise, P. (2012). Crumbs limits oxidase-dependent signaling to maintain epithelial integrity and prevent photoreceptor cell death. J Cell Biol 198: 991-998. PubMed ID: 22965909

    Chartier, F. J., Hardy, E. J. and Laprise, P. (2011). Crumbs controls epithelial integrity by inhibiting Rac1 and PI3K. J Cell Sci 124: 3393-3398. PubMed ID: 21984807

  • Jan LaRocque Department of Human Science, Georgetown University, Washington DC
    Yannuzzi, I., Butler, M. A., Fernandez, J. and LaRocque, J. R. (2021). The Role of Drosophila CtIP in Homology-Directed Repair of DNA Double-Strand Breaks. Genes (Basel) 12(9). PubMed ID: 34573412

    Cox, R. L., Hofley, C. M., Tatapudy, P., Patel, R. K., Dayani, Y., Betcher, M. and LaRocque, J. R. (2019). Functional conservation of RecQ helicase BLM between humans and Drosophila melanogaster. Sci Rep 9(1): 17527. PubMed ID: 31772289

    Fernandez, J., Bloomer, H., Kellam, N. and LaRocque, J. R. (2019). Chromosome preference during homologous recombination repair of DNA double-strand breaks in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 31519746

    Ertl, H. A., Russo, D. P., Srivastava, N., Brooks, J. T., Dao, T. N. and LaRocque, J. R. (2017). The Role of Blm Helicase in homologous recombination, gene conversion tract length, and recombination between diverged sequences in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 28912341

    Delabaere, L., Ertl, H. A., Massey, D. J., Hofley, C. M., Sohail, F., Bienenstock, E. J., Sebastian, H., Chiolo, I. and LaRocque, J. R. (2016). Aging impairs double-strand break repair by homologous recombination in Drosophila germ cells. Aging Cell. PubMed ID: 28000382

    Do, A. T. and LaRocque, J. R. (2015). The role of Drosophila mismatch repair in suppressing recombination between diverged sequences. Sci Rep 5: 17601. PubMed ID: 26616528

    Do, A. T., Brooks, J. T., Le Neveu, M. K. and LaRocque, J. R. (2014). Double-strand break repair assays determine pathway choice and structure of gene conversion events in Drosophila melanogaster. G3 (Bethesda) 4(3): 425-432. PubMed ID: 24368780

  • Amanda Larracuente Department of Biology, River campus, Rochester, NY
    Chen, T., Wei, X., Courret, C., Cui, M., Cheng, L., Wu, J., Ahmad, K., Larracuente, A. M. and Rong, Y. S. (2022). The nanoCUT&RUN technique visualizes telomeric chromatin in Drosophila. PLoS Genet 18(9): e1010351. PubMed ID: 36048878

    Chang, C. H., Gregory, L. E., Gordon, K. E., Meiklejohn, C. D. and Larracuente, A. M. (2022). Unique structure and positive selection promote the rapid divergence of Drosophila Y chromosomes. Elife 11. PubMed ID: 34989337

    Wei, X., Eickbush, D. G., Speece, I. and Larracuente, A. M. (2021). Heterochromatin-dependent transcription of satellite DNAs in the Drosophila melanogaster female germline. Elife 10. PubMed ID: 34259629

    Chakraborty, M., Chang, C. H., Khost, D. E., Vedanayagam, J., Adrion, J. R., Liao, Y., Montooth, K. L., Meiklejohn, C. D., Larracuente, A. M. and Emerson, J. J. (2021). Evolution of genome structure in the Drosophila simulans species complex. Genome Res 31(3): 380-396. PubMed ID: 33563718

  • Erica Larschan Molecular Biology, Cell Biology, & Biochemistry, Brown Universiy
    Jordan, W., 3rd and Larschan, E. (2021). The zinc finger protein CLAMP promotes long-range chromatin interactions that mediate dosage compensation of the Drosophila male X-chromosome. Epigenetics Chromatin 14(1): 29. PubMed ID: 34187599

    Duan, J., Rieder, L., Colonnetta, M. M., Huang, A., McKenney, M., Watters, S., Deshpande, G., Jordan, W., Fawzi, N. and Larschan, E. (2021). CLAMP and Zelda function together to promote Drosophila zygotic genome activation. Elife 10. PubMed ID: 34342574

    Rieder, L. E., Jordan, W. T., 3rd and Larschan, E. N. (2019). Targeting of the dosage-compensated male X-chromosome during early Drosophila development. Cell Rep 29(13): 4268-4275.e4262. PubMed ID: 31875538

    Kaye, E. G., Booker, M., Kurland, J. V., Conicella, A. E., Fawzi, N. L., Bulyk, M. L., Tolstorukov, M. Y. and Larschan, E. (2018). Differential occupancy of two GA-binding proteins promotes targeting of the Drosophila dosage compensation complex to the male X chromosome. Cell Rep 22(12): 3227-3239. PubMed ID: 29562179

    Urban, J. A., Urban, J. M., Kuzu, G. and Larschan, E. N. (2017). The Drosophila CLAMP protein associates with diverse proteins on chromatin. PLoS One 12(12): e0189772. PubMed ID: 29281702

    Urban, J., Kuzu, G., Bowman, S., Scruggs, B., Henriques, T., Kingston, R., Adelman, K., Tolstorukov, M. and Larschan, E. (2017). Enhanced chromatin accessibility of the dosage compensated Drosophila male X-chromosome requires the CLAMP zinc finger protein. PLoS One 12(10): e0186855. PubMed ID: 29077765

    Kaye, E. G., Kurbidaeva, A., Wolle, D., Aoki, T., Schedl, P. and Larschan, E. (2017). Drosophila dosage compensation loci associate with a boundary forming insulator complex. Mol Cell Biol. [Epub ahead of print] PubMed ID: 28784719

    Urban, J. A., Doherty, C. A., Jordan, W. T., 3rd, Bliss, J. E., Feng, J., Soruco, M. M., Rieder, L. E., Tsiarli, M. A. and Larschan, E. N. (2016). The essential Drosophila CLAMP protein differentially regulates non-coding roX RNAs in male and females. Chromosome Res [Epub ahead of print]. PubMed ID: 27995349

    Kuzu, G., Kaye, E. G., Chery, J., Siggers, T., Yang, L., Dobson, J. R., Boor, S., Bliss, J., Liu, W., Jogl, G., Rohs, R., Singh, N. D., Bulyk, M. L., Tolstorukov, M. Y. and Larschan, E. (2016). Expansion of GA dinucleotide repeats increases the density of CLAMP binding sites on the X-Chromosome to promote Drosophila dosage compensation. PLoS Genet 12: e1006120. PubMed ID: 27414415

    Soruco, M. M. and Larschan, E. (2014). A new player in X identification: the CLAMP protein is a key factor in Drosophila dosage compensation. Chromosome Res. PubMed ID: 25102930

    Chery, J. and Larschan, E. (2014). X-marks the spot: X-chromosome identification during dosage compensation. Biochim Biophys Acta 1839: 234-240. PubMed ID: 24406325

  • Camilla Larsen MRC Centre for Developmental Neurobiology, King's College London
    Slater, G., Levy, P., Chan, K.L. and Larsen, C. (2015). A central neural pathway controlling odor tracking in Drosophila. J Neurosci 35: 1831-1848. PubMed ID: 25653345

    Levy, P. and Larsen, C. (2013). Odd-skipped labels a group of distinct neurons associated with the mushroom body and optic lobe in the adult Drosophila brain. J Comp Neurol. PubMed ID: 23749685

    Larsen, C., Shy, D., Spindler, S. R., Fung, S., Pereanu, W., Younossi-Hartenstein, A. and Hartenstein, V. (2009). Patterns of growth, axonal extension and axonal arborization of neuronal lineages in the developing Drosophila brain. Dev Biol 335: 289-304. PubMed ID: 19538956

  • Jan Larsson Department of Molecular Biology, Umea University, Sweden
    Faucillion, M. L., Johansson, A. M. and Larsson, J. (2022). Modulation of RNA stability regulates gene expression in two opposite ways: through buffering of RNA levels upon global perturbations and by supporting adapted differential expression. Nucleic Acids Res. PubMed ID: 35390159

    Ekhteraei-Tousi, S., Lewerentz, J. and Larsson, J. (2020). Painting of fourth and the X-linked 1.688 satellite in D. melanogaster is involved in chromosome-wide gene regulation. Cells 9(2). PubMed ID: 32019091

    Kim, M., Faucillion, M. L. and Larsson, J. (2018). RNA-on-X 1 and 2 in Drosophila melanogaster fulfill separate functions in dosage compensation. PLoS Genet 14(12): e1007842. PubMed ID: 30532158

    Kim, M., Ekhteraei-Tousi, S., Lewerentz, J. and Larsson, J. (2018). The X-linked 1.688 satellite in Drosophila melanogaster promotes specific targeting by Painting of fourth. Genetics 208(2): 623-632. PubMed ID: 29242291

    Figueiredo, M. L., Kim, M., Philip, P., Allgardsson, A., Stenberg, P. and Larsson, J. (2014). Non-coding roX RNAs Prevent the Binding of the MSL-complex to Heterochromatic Regions. PLoS Genet 10: e1004865. PubMed ID: 25501352

    Lundberg, L. E., Kim, M., Johansson, A. M., Faucillion, M. L., Josupeit, R. and Larsson, J. (2013). Targeting of Painting of Fourth to roX1 and roX2 Proximal Sites Suggests Evolutionary Links Between Dosage Compensation and the Regulation of the 4th Chromosome in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 23733888

    Lundberg, L. E., Stenberg, P., Larsson, J. (2013) HP1a, Su(var)3-9, SETDB1 and POF stimulate or repress gene expression depending on genomic position, gene length and expression pattern in Drosophila melanogaster. Nucleic Acids Res. PubMed ID: 23476027

    Crona, F., Dahlberg, O., Lundberg, L. E., Larsson, J. and Mannervik, M. (2013). Gene regulation by the lysine demethylase KDM4A in Drosophila. Dev Biol 373: 453-463. PubMed ID: 23195220

    Figueiredo, M. L., Philip, P., Stenberg, P. and Larsson, J. (2012). HP1a recruitment to promoters is independent of H3K9 methylation in Drosophila melanogaster. PLoS Genet 8: e1003061. PubMed ID: 23166515

    Johansson, A. M., Stenberg, P., Allgardsson, A. and Larsson, J. (2012). POF regulates the expression of genes on the fourth chromosome in Drosophila melanogaster by binding to nascent RNA. Mol Cell Biol 32: 2121-2134. PubMed ID: 22473994

  • Paul Lasko Department of Biology, McGill University, Montreal
    Chaouch, A., Berlandi, J., Chen, C. C. L., Frey, F., Badini, S., Harutyunyan, A. S., Chen, X., Krug, B., Hebert, S., Jeibmann, A., Lu, C., Kleinman, C. L., Hasselblatt, M., Lasko, P., Shirinian, M. and Jabado, N. (2021). Histone H3.3 K27M and K36M mutations de-repress transposable elements through perturbation of antagonistic chromatin marks. Mol Cell. PubMed ID: 34739871

    Dold, A., Han, H., Liu, N., Hildebrandt, A., Bruggemann, M., Ruckle, C., Hanel, H., Busch, A., Beli, P., Zarnack, K., Konig, J., Roignant, J. Y. and Lasko, P. (2020). Makorin 1 controls embryonic patterning by alleviating Bruno1-mediated repression of oskar translation. PLoS Genet 16(1): e1008581. PubMed ID: 31978041

    Kong, J., Han, H., Bergalet, J., Bouvrette, L. P. B., Hernandez, G., Moon, N. S., Vali, H., Lecuyer, E. and Lasko, P. (2019). A ribosomal protein S5 isoform is essential for oogenesis and interacts with distinct RNAs in Drosophila melanogaster. Sci Rep 9(1): 13779. PubMed ID: 31551467

    Berlandi, J., Chaouch, A., De Jay, N., Tegeder, I., Thiel, K., Shirinian, M., Kleinman, C. L., Jeibmann, A., Lasko, P., Jabado, N. and Hasselblatt, M. (2019). Identification of genes functionally involved in the detrimental effects of mutant histone H3.3-K27M in Drosophila melanogaster. Neuro Oncol. PubMed ID: 30715493

    Kowanda, M., Bergalet, J., Wieczorek, M., Brouhard, G., Lecuyer, E. and Lasko, P. (2016). Loss of function of the Drosophila Ninein-related centrosomal protein Bsg25D causes mitotic defects and impairs embryonic development. Biol Open [Epub ahead of print]. PubMed ID: 27422905

    Valzania, L., Ono, H., Ignesti, M., Cavaliere, V., Bernardi, F., Gamberi, C., Lasko, P. and Gargiulo, G. (2015). Drosophila 4EHP is essential for the larval-pupal transition and required in the prothoracic gland for ecdysone biosynthesis. Dev Biol. PubMed ID: 26721418

    Liu, N. and Lasko, P. (2015). Analysis of RNA interference lines identifies new functions of maternally-expressed genes involved in embryonic patterning in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 25834215

    Ghosh, S and Lasko, P. (2015). Loss-of-function analysis reveals distinct requirements of the translation initiation factors eIF4E, eIF4E-3, eIF4G and eIF4G2 in Drosophila spermatogenesis. PLoS One 10: e0122519. PubMed ID: 25849588

    Liu, N. and Lasko, P. (2015). Analysis of RNA interference lines identifies new functions of maternally-expressed genes involved in embryonic patterning in Drosophila melanogaster. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 25834215

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

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

    Srivastav, S. P., Rahman, R., Ma, Q., Pierre, J., Bandyopadhyay, S. and Lau, N. C. (2019). Har-P, a short P-element variant, weaponizes P-transposase to severely impair Drosophila development. Elife 8. PubMed ID: 31845649

    Kozeretska, I. A., Shulha, V. I., Serga, S. V., Rozhok, A. I., Protsenko, O. V. and Lau, N. C. (2018). A rapid change in P-element-induced hybrid dysgenesis status in Ukrainian populations of Drosophila melanogaster. Biol Lett 14(8). PubMed ID: 30135116

    Clark, J. P., Rahman, R., Yang, N., Yang, L. H. and Lau, N. C. (2017). Drosophila PAF1 Modulates PIWI/piRNA Silencing Capacity. Curr Biol 27(17): 2718-2726 e2714. PubMed ID: 28844648

  • Allen Laughon Laboratory of Genetics University of Wisconsin-Madison
    Chen, J., et al. (2012). Crossveinless d is a vitellogenin-like lipoprotein that binds BMPs and HSPGs, and is required for normal BMP signaling in the Drosophila wing. Development 139(12): 2170-6. PubMed ID: 22573617

  • Gilles Laurent Max Planck Institute for Brain Research, Frankfurt
    Cassenaer, S. and Laurent, G. (2012). Conditional modulation of spike-timing-dependent plasticity for olfactory learning. Nature 482: 47-52. PubMed ID: 22278062

    Papadopoulou, M., Cassenaer, S., Nowotny, T. and Laurent, G. (2011). Normalization for sparse encoding of odors by a wide-field interneuron. Science 332: 721-725. PubMed ID: 21551062

    Geffen, M. N., Broome, B. M., Laurent, G. and Meister, M. (2009). Neural encoding of rapidly fluctuating odors. Neuron 61: 570-586. PubMed ID: 19249277

  • Stefan Laurent Department of Biology II, Ludwig-Maximilian University Munich, Planegg, Germany,
    Kapopoulou, A., Kapun, M., Pieper, B., Pavlidis, P., Wilches, R., Duchen, P., Stephan, W. and Laurent, S. (2020). Demographic analyses of a new sample of haploid genomes from a Swedish population of Drosophila melanogaster. Sci Rep 10(1): 22415. PubMed ID: 33376238

    Arguello, J. R., Laurent, S. and Clark, A. G. (2019). Demographic history of the human commensal Drosophila melanogaster. Genome Biol Evol. PubMed ID: 30715331

    Kapopoulou, A., Pfeifer, S. P., Jensen, J. D. and Laurent, S. (2018). The demographic history of African Drosophila melanogaster. Genome Biol Evol. PubMed ID: 30169784

    Werzner, A., Pavlidis, P., Ometto, L., Stephan, W., Laurent, S. (2013) Selective Sweep in the Flotillin-2 Region of European Drosophila melanogaster. PLoS One 8: e56629. PubMed ID: 23437190

    Duchen, P., Zivkovic, D., Hutter, S., Stephan, W. and Laurent, S. (2013). Demographic inference reveals African and European admixture in the North American Drosophila melanogaster population. Genetics 193: 291-301. PubMed ID: 23150605

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

  • Sergey Alexandrovich Lavrov Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics of National Research Centre, Moscow
    Kolesnikova, T. D., Klenov, M. S., Nokhova, A. R., Lavrov, S. A., Pokholkova, G. V., Schubert, V., Maltseva, S. V., Cook, K. R., Dixon, M. J. and Zhimulev, I. F. (2022). A Spontaneous Inversion of the X Chromosome Heterochromatin Provides a Tool for Studying the Structure and Activity of the Nucleolus in Drosophila melanogaster. Cells 11(23). PubMed ID: 36497131

    Kogan, G. L., Mikhaleva, E. A., Olenkina, O. M., Ryazansky, S. S., Galzitskaya, O. V., Abramov, Y. A., Leinsoo, T. A., Akulenko, N. V., Lavrov, S. A. and Gvozdev, V. A. (2022). Extended disordered regions of ribosome-associated NAC proteins paralogs belong only to the germline in Drosophila melanogaster. Sci Rep 12(1): 11191. PubMed ID: 35778515

    Solodovnikov, A. A., Gvozdev, V. A. and Lavrov, S. A. (2020). High Level of Gene Transcription at the Embryonic Stage Leads to the Suppression of Heterochromatic Trans-Inactivation in Drosophila melanogaster Adults. Biochemistry (Mosc) 85(4): 472-479. PubMed ID: 32569554

  • Hakeem Lawal Delaware State University, Dover
    White, D., de Sousa Abreu, R. P., Blake, A., Murphy, J., Showell, S., Kitamoto, T. and Lawal, H. O. (2020). Deficits in the vesicular acetylcholine transporter alter lifespan and behavior in adult Drosophila melanogaster. Neurochem Int 137: 104744. PubMed ID: 32315665

    Showell, S. S., Martinez, Y., Gondolfo, S., Boppana, S. and Lawal, H. O. (2020). Overexpression of the vesicular acetylcholine transporter disrupts cognitive performance and causes age-dependent locomotion decline in Drosophila. Mol Cell Neurosci 105: 103483. PubMed ID: 32217162

    White, D., de Sousa Abreu, R. P., Blake, A., Murphy, J., Showell, S., Kitamoto, T. and Lawal, H. O. (2020). Deficits in the vesicular acetylcholine transporter alter lifespan and behavior in adult Drosophila melanogaster. Neurochem Int 137: 104744. PubMed ID: 32315665

    Martin, C. A., Barajas, A., Lawless, G., Lawal, H. O., Assani, K., Lumintang, Y. P., Nunez, V. and Krantz, D. E. (2014). Synergistic effects on dopamine cell death in a Drosophila model of chronic toxin exposure. Neurotoxicology 44: 344-351. PubMed ID: Peter Lawrence Department of Zoology, University of Cambridge
    Pietra, S., Ng, K., Lawrence, P. A. and Casal, J. (2020). Planar cell polarity in the larval epidermis of Drosophila and the role of microtubules. Open Biol 10(12): 200290. PubMed ID: 33295841

    Lawrence, P. A., Casal, J., Celis, J. F. and Morata, G. (2019). A refutation to 'A new A-P compartment boundary and organizer in holometabolous insect wings'. Sci Rep 9(1): 7049. PubMed ID: 31065001

    Casal, J., Ibanez-Jimenez, B. and Lawrence, P. A. (2018). Planar cell polarity: the prickle gene acts independently on both the Ds/Ft and the Stan/Fz systems. Development. PubMed ID: 30154173

    Saavedra, P., Brittle, A., Palacios, I.M., Strutt, D., Casal, J. and Lawrence, P.A. (2016). Planar cell polarity: the Dachsous/Fat system contributes differently to the embryonic and larval stages of Drosophila. Biol Open [Epub ahead of print]. PubMed ID: 26935392

    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

    Lawrence, P. A., Casal, J. (2013) The mechanisms of planar cell polarity, growth and the Hippo pathway: Some known unknowns. Dev Biol 377: 1-8. PubMed ID: 23592229

    Fabre, C. C., Hedwig, B., Conduit, G., Lawrence, P. A., Goodwin, S. F. and Casal, J. (2012). Substrate-borne vibratory communication during courtship in Drosophila melanogaster. Curr Biol 22: 2180-2185. PubMed ID: 23103187

    Struhl, G., Casal, J. and Lawrence, P. A. (2012). Dissecting the molecular bridges that mediate the function of Frizzled in planar cell polarity. Development 139: 3665-3674. PubMed ID: 22949620

    Krzemien, J., Fabre, C. C., Casal, J. and Lawrence, P. A. (2012). The muscle pattern of the Drosophila abdomen depends on a subdivision of the anterior compartment of each segment. Development 139: 75-83. PubMed ID: 22147953

    Lawrence, P. A. (2011). Planar cell polarity: fashioning solutions. Fly (Austin) 5: 126-128. PubMed ID: 21245663

  • Brian Lazzaro Department of Entomology, Cornell University
    Gupta, V., Frank, A. M., Matolka, N. and Lazzaro, B. P. (2022). Inherent constraints on a polyfunctional tissue lead to a reproduction-immunity tradeoff. BMC Biol 20(1): 127. PubMed ID: 35655304

    Gordon, K. E., Wolfner, M. F. and Lazzaro, B. P. (2022). A single mating is sufficient to induce persistent reduction of immune defense in mated female Drosophila melanogaster. J Insect Physiol 140: 104414. PubMed ID: 35728669

    Shahrestani, P., King, E., Ramezan, R., Phillips, M., Riddle, M., Thornburg, M., Greenspan, Z., Estrella, Y., Garcia, K., Chowdhury, P., Malarat, G., Zhu, M., Rottshaefer, S. M., Wraight, S., Griggs, M., Vandenberg, J., Long, A. D., Clark, A. G. and Lazzaro, B. P. (2021). The molecular architecture of Drosophila melanogaster defense against Beauveria bassiana explored through evolve and resequence and quantitative trait locus mapping. G3 (Bethesda). PubMed ID: 34534291

    Chambers, M. C., Jacobson, E., Khalil, S. and Lazzaro, B. P. (2019). Consequences of chronic bacterial infection in Drosophila melanogaster. PLoS One 14(10): e0224440. PubMed ID: 31648237

    Im, J. H. and Lazzaro, B. P. (2018). Population genetic analysis of autophagy and phagocytosis genes in Drosophila melanogaster and D. simulans. PLoS One 13(10): e0205024. PubMed ID: 30281656

    Duneau, D. F., Kondolf, H. C., Im, J. H., Ortiz, G. A., Chow, C., Fox, M. A., Eugenio, A. T., Revah, J., Buchon, N. and Lazzaro, B. P. (2017). The Toll pathway underlies host sexual dimorphism in resistance to both Gram-negative and Gram-positive bacteria in mated Drosophila. BMC Biol 15(1): 124. PubMed ID: 29268741

    Schwenke, R. A. and Lazzaro, B. P. (2017). Juvenile Hormone Suppresses Resistance to Infection in Mated Female Drosophila melanogaster. Curr Biol 27(4): 596-601. PubMed ID: 28190728

    Howick, V. M. and Lazzaro, B. P. (2017). The genetic architecture of defense as resistance to and tolerance of bacterial infection in Drosophila melanogaster. Mol Ecol [Epub ahead of print]. PubMed ID: 28099780

    Unckless, R.L., Howick, V.M. and Lazzaro, B.P. (2016). Convergent balancing selection on an antimicrobial peptide in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26776733

    Khalil, S., Jacobson, E., Chambers, M. C. and Lazzaro, B. P. (2015). Systemic bacterial infection and immune defense phenotypes in Drosophila melanogaster. J Vis Exp. PubMed ID: 25992475

    Unckless, R. L., Rottschaefer, S. M. and Lazzaro, B. P. (2015). The complex contributions of genetics and nutrition to immunity in Drosophila melanogaster. PLoS Genet 11: e1005030. PubMed ID: 25764027

  • Judy Leatherman Department of Biological Sciences, College of Natural and Health Sciences, University of Northern Colorado, Greeley, CO
    Mues, N., Hammer, K. and Leatherman, J. (2022). Pvr regulates cyst stem cell division in the Drosophila testis niche, and has functions distinct from Egfr. Cells Dev 173: 203822. PubMed ID: 36400422

    Johnson, B. and Leatherman, J. (2021). Merlin and expanded integrate cell signaling that regulates cyst stem cell proliferation in the Drosophila testis niche. Dev Biol 477: 133-144. PubMed ID: 34044021

    Leatherman, J. L. and Dinardo, S. (2010). Germline self-renewal requires cyst stem cells and stat regulates niche adhesion in Drosophila testes. Nat Cell Biol 12(8): 806-811. PubMed ID: 20622868

  • Eric Le Bourg Centre de Recherches sur la Cognition Animale, Toulouse
    Houssin, E., Pinot, M., Bellec, K. and Le Borgne, R. (2021). Par3 cooperates with Sanpodo for the assembly of Notch clusters following asymmetric division of Drosophila sensory organ precursor cells. Elife 10. PubMed ID: 34596529

    Bellec, K., Pinot, M., Gicquel, I. and Le Borgne, R. (2021). The Clathrin adaptor AP-1 and Stratum act in parallel pathways to control Notch activation in Drosophila sensory organ precursors cells. Development 148(1). PubMed ID: 33298463

    Esmangart de Bournonville, T. and Le Borgne, R. (2020). Interplay between Anakonda, Gliotactin, and M6 for Tricellular Junction Assembly and Anchoring of Septate Junctions in Drosophila Epithelium. Curr Biol. PubMed ID: 32857971

    Bellec, K., Gicquel, I. and Le Borgne, R. (2018). Stratum recruits Rab8 at Golgi exit sites to regulate the basolateral sorting of Notch and Sanpodo. Development 145(13). PubMed ID: 29967125

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

    Polesello, C. and Le Bourg, E. (2017). A mild cold stress that increases resistance to heat lowers FOXO translocation in Drosophila melanogaster. Biogerontology [Epub ahead of print]. PubMed ID: 28677014

    Le Bourg, E. (2013). Fasting can protect young and middle-aged Drosophila melanogaster flies against a severe cold stress. Biogerontology. PubMed ID: 23990216

    Le Bourg, E., Malod, K. and Massou, I. (2012). The NF-kappaB-like factor DIF could explain some positive effects of a mild stress on longevity, behavioral aging, and resistance to strong stresses in Drosophila melanogaster. Biogerontology 13: 445-455. PubMed ID: 22791143

    Le Bourg, E. (2012). Combined effects of two mild stresses (cold and hypergravity) on longevity, behavioral aging, and resistance to severe stresses in Drosophila melanogaster. Biogerontology 13: 313-328. PubMed ID: 22391643

  • Roland Le Borgne Institute of Genetics and Development of Rennes
    Teng, X., Qin, L., Le Borgne, R. and Toyama, Y. (2016). Remodeling of adhesion and modulation of mechanical tensile forces during apoptosis in Drosophila epithelium. Development. PubMed ID: 27888195

    Loyer, N., Kolotuev, I., Pinot, M. and Le Borgne, R. (2015). Drosophila E-cadherin is required for the maintenance of ring canals anchoring to mechanically withstand tissue growth. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26424451

    Cotton, M., Benhra, N., Le Borgne, R. (2013) Numb Inhibits the Recycling of Sanpodo in Drosophila Sensory Organ Precursor. Curr Biol. PubMed ID: 23523246

    Founounou, N., Loyer, N., Le Borgne, R. (2013) Septins Regulate the Contractility of the Actomyosin Ring to Enable Adherens Junction Remodeling during Cytokinesis of Epithelial Cells. Dev Cell 24: 242-255. PubMed ID: 23410939

    Le Bras, S., Rondanino, C., Kriegel-Taki, G., Dussert, A. and Le Borgne, R. (2012). Genetic identification of intracellular trafficking regulators involved in Notch-dependent binary cell fate acquisition following asymmetric cell division. J Cell Sci 125: 4886-4901. PubMed ID: 22825875

    Founounou, N. and Le Borgne, R. (2011). Tissue polarity: PCP inheritance ensured by selective mitotic endocytosis. Curr Biol 21: R690-692. PubMed ID: 21959155

    Burgess, J., Jauregui, M., Tan, J., Rollins, J., Lallet, S., Leventis, P. A., Boulianne, G. L., Chang, H. C., Le Borgne, R., Kramer, H. and Brill, J. A. (2011). AP-1 and clathrin are essential for secretory granule biogenesis in Drosophila. Mol Biol Cell 22: 2094-2105. PubMed ID: 21490149

  • Magalie Lecourtois Human Biology, Neuroscience, Universit de Rouen, Mont-Saint-Aignan
    Miguel, L., Frebourg, T., Campion, D. and Lecourtois, M. (2020). Moderate Overexpression of Tau in Drosophila Exacerbates Amyloid-beta-Induced Neuronal Phenotypes and Correlates with Tau Oligomerization. J Alzheimers Dis. PubMed ID: 32065789

    Feuillette, S., Charbonnier, C., Frebourg, T., Campion, D. and Lecourtois, M. (2020). A Connected Network of Interacting Proteins Is Involved in Human-Tau Toxicity in Drosophila. Front Neurosci 14: 68. PubMed ID: 32116515

    Pons, M., Prieto, S., Miguel, L., Frebourg, T., Campion, D., Sune, C. and Lecourtois, M. (2018). Identification of TCERG1 as a new genetic modulator of TDP-43 production in Drosophila. Acta Neuropathol Commun 6(1): 138. PubMed ID: 30541625

    Miguel, L., Avequin, T., Pons, M., Frebourg, T., Campion, D. and Lecourtois, M. (2018). FTLD/ALS-linked TDP-43 mutations do not alter TDP-43's ability to self-regulate its expression in Drosophila. Brain Res. PubMed ID: 29778779

    Feuillette, S., Delarue, M., Riou, G., Gaffuri, A. L., Wu, J., Lenkei, Z., Boyer, O., Frebourg, T., Campion, D. and Lecourtois, M. (2017). Neuron-to-Neuron Transfer of FUS in Drosophila Primary Neuronal Culture Is Enhanced by ALS-Associated Mutations. J Mol Neurosci. PubMed ID: 28429234

    Khalil, B., Cabirol-Pol, M. J., Miguel, L., Whitworth, A. J., Lecourtois, M. and Lievens, J. C. (2017). Enhancing Mitofusin/Marf ameliorates neuromuscular dysfunction in Drosophila models of TDP-43 proteinopathies. Neurobiol Aging 54: 71-83. PubMed ID: 28324764

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

  • Thomas Lecuit Development Biology Institute of Marseille Luminy
    Lavalou, J., Mao, Q., Harmansa, S., Kerridge, S., Lellouch, A. C., Philippe, J. M., Audebert, S., Camoin, L. and Lecuit, T. (2021). Formation of polarized contractile interfaces by self-organized Toll-8/Cirl GPCR asymmetry. Dev Cell. PubMed ID: 33932333

    Chandran, R., Kale, G., Philippe, J. M., Lecuit, T. and Mayor, S. (2021). Distinct actin-dependent nanoscale assemblies underlie the dynamic and hierarchical organization of E-cadherin. Curr Biol. PubMed ID: 33607036

    Palavalli, A., Tizon-Escamilla, N., Rupprecht, J. F. and Lecuit, T. (2020). Deterministic and Stochastic Rules of Branching Govern Dendrite Morphogenesis of Sensory Neurons. Curr Biol. PubMed ID: 33212017

    Dehapiot, B., Clément, R., Alegot, H., Gazso-Gerhat, G., Philippe, J. M. and Lecuit, T. (2020). Assembly of a persistent apical actin network by the formin Frl/Fmnl tunes epithelial cell deformability. Nat Cell Biol. PubMed ID: 32483386

    Garcia De Las Bayonas, A., Philippe, J. M., Lellouch, A. C. and Lecuit, T. (2019). Distinct RhoGEFs activate apical and junctional contractility under control of G proteins during epithelial morphogenesis. Curr Biol. PubMed ID: 31522942

    Bailles, A., Collinet, C., Philippe, J. M., Lenne, P. F., Munro, E. and Lecuit, T. (2019). Genetic induction and mechanochemical propagation of a morphogenetic wave. Nature 572(7770): 467-473. PubMed ID: 31413363

    Clement, R., Dehapiot, B., Collinet, C., Lecuit, T. and Lenne, P. F. (2017). Viscoelastic dissipation stabilizes cell shape changes during tissue morphogenesis. Curr Biol 27(20): 3132-3142. PubMed ID: 28988857

    Hampoelz, B., Mackmull, M. T., Machado, P., Ronchi, P., Bui, K. H., Schieber, N., Santarella-Mellwig, R., Necakov, A., Andres-Pons, A., Philippe, J. M., Lecuit, T., Schwab, Y. and Beck, M. (2016). Pre-assembled nuclear pores insert into the nuclear envelope during early development. Cell. PubMed ID: 27397507

    Kerridge, S., Munjal, A., Philippe, J. M., Jha, A., de Las Bayonas, A. G., Saurin, A. J. and Lecuit, T. (2016). Modular activation of Rho1 by GPCR signalling imparts polarized myosin II activation during morphogenesis. Nat Cell Biol. PubMed ID: 26780298

    Collinet, C., Rauzi, M., Lenne, P.F. and Lecuit, T. (2015). Local and tissue-scale forces drive oriented junction growth during tissue extension. Nat Cell Biol [Epub ahead of print]. PubMed ID: 26389664

    Munjal, A., Philippe, J. M., Munro, E. and Lecuit, T. (2015). A self-organized biomechanical network drives shape changes during tissue morphogenesis. Nature 524: 351-355. PubMed ID: 26214737

    Heemskerk, I., Lecuit, T. and LeGoff, L. (2014). Dynamic clonal analysis based on chronic in vivo imaging allows multiscale quantification of growth in the Drosophila wing disc. Development 141: 2339-2348. PubMed ID: 24866118

  • Eric Lecuyer RNA Biology Research Unit, Institut de recherches cliniques de Montreal
    Chin, A. and Lecuyer, E. (2021). Puromycin Labeling Coupled with Proximity Ligation Assays to Define Sites of mRNA Translation in Drosophila Embryos and Human Cells. Methods Mol Biol 2381: 267-284. PubMed ID: 34590282

    Chin, A. and Lecuyer, E. (2021). Puromycin Labeling Coupled with Proximity Ligation Assays to Define Sites of mRNA Translation in Drosophila Embryos and Human Cells. Methods Mol Biol 2381: 267-284. PubMed ID: 34590282

    Bergalet, J., Patel, D., Legendre, F., Lapointe, C., Benoit Bouvrette, L. P., Chin, A., Blanchette, M., Kwon, E. and Lecuyer, E. (2020). Inter-dependent Centrosomal Co-localization of the cen and ik2 cis-Natural Antisense mRNAs in Drosophila. Cell Rep 30(10): 3339-3352.e3336. PubMed ID: 32160541

    Benoit Bouvrette, L. P., Bovaird, S., Blanchette, M. and Lecuyer, E. (2019). oRNAment: a database of putative RNA binding protein target sites in the transcriptomes of model species. Nucleic Acids Res. PubMed ID: 31724725

    Lefebvre, F. A., Benoit Bouvrette, L. P., Bergalet, J. and Lecuyer, E. (2017). Biochemical fractionation of time-resolved Drosophila embryos reveals similar transcriptomic alterations in replication checkpoint and histone mRNA processing mutants. J Mol Biol [Epub ahead of print]. PubMed ID: 28167048

    Kowanda, M., Bergalet, J., Wieczorek, M., Brouhard, G., Lecuyer, E. and Lasko, P. (2016). Loss of function of the Drosophila Ninein-related centrosomal protein Bsg25D causes mitotic defects and impairs embryonic development. Biol Open [Epub ahead of print]. PubMed ID: 27422905

    Lefebvre, F. A., Benoit Bouvrette, L. P., Perras, L., Blanchet-Cohen, A., Garnier, D., Rak, J. and Lecuyer, E. (2016). Comparative transcriptomic analysis of human and Drosophila extracellular vesicles. Sci Rep 6: 27680. PubMed ID: 27282340

    Iampietro, C., Bergalet, J., Wang, X., Cody, N. A., Chin, A., Lefebvre, F. A., Douziech, M., Krause, H. M. and Lecuyer, E. (2014). Developmentally Regulated Elimination of Damaged Nuclei Involves a Chk2-Dependent Mechanism of mRNA Nuclear Retention. Dev Cell. PubMed ID: 24835465

    Legendre, F., Cody, N., Iampietro, C., Bergalet, J., Lefebvre, F. A., Moquin-Beaudry, G., Zhang, O., Wang, X. and Lecuyer, E. (2013). Whole Mount RNA Fluorescent in situ Hybridization of Drosophila Embryos. J Vis Exp. PubMed ID: 23407302

    Wang, E. T., Cody, N. A., Jog, S., Biancolella, M., Wang, T. T., Treacy, D. J., Luo, S., Schroth, G. P., Housman, D. E., Reddy, S., Lecuyer, E. and Burge, C. B. (2012). Transcriptome-wide regulation of pre-mRNA splicing and mRNA localization by muscleblind proteins. Cell 150: 710-724. PubMed ID: 22901804

    Lecuyer, E. (2011). High resolution fluorescent in situ hybridization in Drosophila. Methods Mol Biol 714: 31-47. PubMed ID: 21431733

  • Cheng-Yu Lee Life Sciences Institute, University of Michigan
    Rajan, A., Ostgaard, C. M. and Lee, C. Y. (2021). Regulation of Neural Stem Cell Competency and Commitment during Indirect Neurogenesis. Int J Mol Sci 22(23). PubMed ID: 34884676

    Michki, N. S., Li, Y., Sanjasaz, K., Zhao, Y., Shen, F. Y., Walker, L. A., Cao, W., Lee, C. Y. and Cai, D. (2021). The molecular landscape of neural differentiation in the developing Drosophila brain revealed by targeted scRNA-seq and multi-informatic analysis. Cell Rep 35(4): 109039. PubMed ID: 33909998

    Komori, H., Golden, K. L., Kobayashi, T., Kageyama, R. and Lee, C. Y. (2018). Multilayered gene control drives timely exit from the stem cell state in uncommitted progenitors during Drosophila asymmetric neural stem cell division. Genes Dev 32(23-24): 1550-1561. PubMed ID: 30463902

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

    Komori, H., Xiao, Q., Janssens, D. H., Dou, Y. and Lee, C. Y. (2014). Trithorax maintains the functional heterogeneity of neural stem cells through the transcription factor Buttonhead. Elife 3. PubMed ID: 25285447

    Janssens, D. H., Komori, H., Grbac, D., Chen, K., Koe, C. T., Wang, H. and Lee, C. Y. (2014). Earmuff restricts progenitor cell potential by attenuating the competence to respond to self-renewal factors. Development 141: 1036-1046. PubMed ID: 24550111

    Janssens, D. H. and Lee, C. Y. (2014). It takes two to tango, a dance between the cells of origin and cancer stem cells in the Drosophila larval brain. Semin Cell Dev Biol. PubMed ID: 24631354

    Kuang, C., Golden, K. L., Simon, C. R., Damrath, J., Buttitta, L., Gamble, C. E. and Lee, C. Y. (2014). A novel Fizzy/Cdc20-dependent mechanism suppresses necrosis in neural stem cells. Development. PubMed ID: 24598157

    Komori, H., Xiao, Q., McCartney, B. M. and Lee, C. Y. (2013). Brain tumor specifies intermediate progenitor cell identity by attenuating beta-catenin/Armadillo activity. Development. PubMed ID: 24257623

    Haenfler, J. M., Kuang, C. and Lee, C. Y. (2012). Cortical aPKC kinase activity distinguishes neural stem cells from progenitor cells by ensuring asymmetric segregation of Numb. Dev Biol 365: 219-228. PubMed ID: 22394487

    Xiao, Q., Komori, H. and Lee, C. Y. (2012). klumpfuss distinguishes stem cells from progenitor cells during asymmetric neuroblast division. Development 139: 2670-2680. PubMed ID: 22745313

    Weng, M., Komori, H. and Lee, C. Y. (2012). Identification of neural stem cells in the Drosophila larval brain. Methods Mol Biol 879: 39-46. PubMed ID: 22610552

  • Chi-Hon Lee NICHD, NIH, Bethesda
    Luo, J., Ting, C. Y., Li, Y., McQueen, P., Lin, T. Y., Hsu, C. P. and Lee, C. H. (2020). Antagonistic regulation by insulin-like peptide and activin ensures the elaboration of appropriate dendritic field sizes of amacrine neurons. Elife 9. PubMed ID: 32175842

    Song, B. M. and Lee, C. H. (2018). Toward a Mechanistic Understanding of Color Vision in Insects. Front Neural Circuits 12: 16. PubMed ID: 29527156

    Ting, C. Y., McQueen, P. G., Pandya, N., McCreedy, E. S., McAuliffe, M. and Lee, C. H. (2017). Analyzing Dendritic Morphology in Columns and Layers. J Vis Exp(121) [Epub ahead of print]. PubMed ID: 28362388

    Li, Y., Dharkar, P., Han, T. H., Serpe, M., Lee, C. H. and Mayer, M. L. (2016). Novel functional properties of Drosophila CNS glutamate receptors. Neuron 92(5): 1036-1048. PubMed ID: 27889096

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

    Lin, T. Y., Luo, J., Shinomiya, K., Ting, C. Y., Lu, Z., Meinertzhagen, I. A. and Lee, C. H. (2016). Mapping chromatic pathways in the Drosophila visual system. J Comp Neurol 524(2):213-27. PubMed ID: 26648045

    Wardill, T. J., List, O., Li, X., Dongre, S., McCulloch, M., Ting, C. Y., O'Kane, C. J., Tang, S., Lee, C. H., Hardie, R. C. and Juusola, M. (2012). Multiple spectral inputs improve motion discrimination in the Drosophila visual system. Science 336: 925-931. PubMed ID: 22605779

    Melnattur, K. V., Pursley, R., Lin, T. Y., Ting, C. Y., Smith, P. D., Pohida, T. and Lee, C. H. (2014). Multiple Redundant Medulla Projection Neurons Mediate Color Vision in Drosophila. J Neurogenet. PubMed ID: 24766346
    Shinomiya, K., Karuppudurai, T., Lin, T. Y., Lu, Z., Lee, C. H. and Meinertzhagen, I. A. (2014). Candidate Neural Substrates for Off-Edge Motion Detection in Drosophila. Curr Biol. PubMed ID: 24768048

    Karuppudurai, T., Lin, T. Y., Ting, C. Y., Pursley, R., Melnattur, K. V., Diao, F., White, B. H., Macpherson, L. J., Gallio, M., Pohida, T. and Lee, C. H. (2014). A Hard-Wired Glutamatergic Circuit Pools and Relays UV Signals to Mediate Spectral Preference in Drosophila. Neuron 81: 603-615. PubMed ID: 24507194

  • Daewoo Lee Biological Sciences, Ohio University, Athens
    Ismael, S., Sindi, G., Colvin, R. A. and Lee, D. (2021). Activity-dependent release of phosphorylated human tau from Drosophila neurons in primary culture. J Biol Chem: 101108. PubMed ID: 34473990

    Ganguly, A., Qi, C., Bajaj, J. and Lee, D. (2020). Serotonin receptor 5-HT7 in Drosophila mushroom body neurons mediates larval appetitive olfactory learning. Sci Rep 10(1): 21267. PubMed ID: 33277559

    Blosser, J. A., Podolsky, E. and Lee, D. (2020). L-DOPA-Induced Dyskinesia in a Genetic Drosophila Model of Parkinson's Disease. Exp Neurobiol 29(4): 273-284. PubMed ID: 32921640

    Wiemerslage, L., Ismael, S. and Lee, D. (2016). Early alterations of mitochondrial morphology in dopaminergic neurons from Parkinson's disease-like pathology and time-dependent neuroprotection with D2 receptor activation. Mitochondrion [Epub ahead of print]. PubMed ID: 27423787

    Lee, D. (2015). Global and local missions of cAMP signaling in neural plasticity, learning, and memory. Front Pharmacol 6: 161. PubMed ID: 26300775

    Wiemerslage, L. and Lee, D. (2014). Role of Drosophila calcium channel cacophony in dopaminergic neurodegeneration and neuroprotection. Neurosci Lett 584C: 342-346. PubMed ID: 25445363

    Qi, C. and Lee, D. (2014). Pre- and postsynaptic role of Dopamine D2 Receptor DD2R in Drosophila olfactory associative learning. Biology (Basel) 3: 831-845. PubMed ID: 25422852

    Varga, S. J., Qi, C., Podolsky, E. and Lee, D. (2014). A new Drosophila model to study the interaction between genetic and environmental factors in Parkinsons disease. Brain Res. PubMed ID: 25130663

    Wiemerslage, L., Schultz, B. J., Ganguly, A., Lee, D. (2013) Selective degeneration of dopaminergic neurons by MPP and its rescue by D2 autoreceptors in Drosophila primary culture. J Neurochem. PubMed ID: 23452092

    Ganguly, A., Lee, D. (2013) Suppression of inhibitory GABAergic transmission by cAMP signaling pathway: alterations in learning and memory mutants. Eur J Neurosci. PubMed ID: 23387411

    Darya, K., Ganguly, A. and Lee, D. (2009). Quantitative analysis of synaptic boutons in Drosophila primary neuronal cultures. Brain Res 1280: 1-12. PubMed ID: 19460362

  • Ethan Lee Cell and Developmental Biology, Vanderbilt
    Neitzel, L. R., Broadus, M. R., Zhang, N., Sawyer, L., Wallace, H. A., Merkle, J. A., Jodoin, J. N., Sitaram, P., Crispi, E. E., Rork, W., Lee, L. A., Pan, D., Gould, K. L., Page-McCaw, A. and Lee, E. (2018). Characterization of a cdc14 null allele in Drosophila melanogaster. Biol Open. PubMed ID: 29945873

    Saito-Diaz, K., Benchabane, H., Tiwari, A., Tian, A., Li, B., Thompson, J. J., Hyde, A. S., Sawyer, L. M., Jodoin, J. N., Santos, E., Lee, L. A., Coffey, R. J., Beauchamp, R. D., Williams, C. S., Kenworthy, A. K., Robbins, D. J., Ahmed, Y. and Lee, E. (2018). APC Inhibits Ligand-Independent Wnt Signaling by the Clathrin Endocytic Pathway. Dev Cell 44(5): 566-581.e568. PubMed ID: 29533772

    Sitaram, P., Anderson, M. A., Jodoin, J. N., Lee, E. and Lee, L. A. (2012). Regulation of dynein localization and centrosome positioning by Lis-1 and asunder during Drosophila spermatogenesis. Development 139: 2945-2954. PubMed ID: 22764052

    Hanson, A. J., Wallace, H. A., Freeman, T. J., Beauchamp, R. D., Lee, L. A. and Lee, E. (2012). XIAP monoubiquitylates Groucho/TLE to promote canonical Wnt signaling. Mol Cell 45: 619-628. PubMed ID: 22304967

    Jernigan, K. K., Cselenyi, C. S., Thorne, C. A., Hanson, A. J., Tahinci, E., Hajicek, N., Oldham, W. M., Lee, L. A., Hamm, H. E., Hepler, J. R., Kozasa, T., Linder, M. E. and Lee, E. (2010). Gbetagamma activates GSK3 to promote LRP6-mediated beta-catenin transcriptional activity. Sci Signal 3: ra37. PubMed ID: 20460648

  • Jun Hee Lee Molecular & Integrative Physiology, University of Michigan
    Jung, J. E., Lee, J. Y., Kim, I. R., Park, S. M., Kang, J. W., Kim, Y. H., Park, H. R. and Lee, J. H. (2020). MicroRNA-31 Regulates Expression of Wntless in Both Drosophila melanogaster and Human Oral Cancer Cells. Int J Mol Sci 21(19). PubMed ID: 33007980

    Park, S.M., Park, H.R. and Lee, J.H. (2017). MAPK3 at the autism-linked human 16p11.2 locus influences precise synaptic target selection at Drosophila larval neuromuscular junctions. Mol Cells [Epub ahead of print]. PubMed ID: 28196412

    Park, S. M., Littleton, J. T., Park, H. R. and Lee, J. H. (2016). Drosophila homolog of human KIF22 at the autism-linked 16p11.2 loci influences synaptic connectivity at larval neuromuscular junctions. Exp Neurobiol 25: 33-39. PubMed ID: 26924931

    Kim, M., Semple, I., Kim, B., Kiers, A., Nam, S., Park, H. W., Park, H., Ro, S. H., Kim, J. S., Juhasz, G. and Lee, J. H. (2015). Drosophila Gyf/GRB10 interacting GYF protein is an autophagy regulator that controls neuron and muscle homeostasis. Autophagy: 0. PubMed ID: 26086452

    Kim, M., Park, H. L., Park, H. W., Ro, S. H., Nam, S. G., Reed, J. M., Guan, J. L. and Lee, J. H. (2013). Drosophila Fip200 is an essential regulator of autophagy that attenuates both growth and aging. Autophagy 9. PubMed ID: 23819996

    Hwang, S., Song, S., Hong, Y. K., Choi, G., Suh, Y. S., Han, S. Y., Lee, M., Park, S. H., Lee, J. H., Lee, S., Bang, S. M., Jeong, Y., Chung, W. J., Lee, I. S., Jeong, G., Chung, J. and Cho, K. S. (2013). Drosophila DJ-1 decreases neural sensitivity to stress by negatively regulating Daxx-like protein through dFOXO. PLoS Genet 9: e1003412. PubMed ID: 23593018

    Park, S. H., Lee, S., Hong, Y. K., Hwang, S., Lee, J. H., Bang, S. M., Kim, Y. K., Koo, B. S., Lee, I. S. and Cho, K. S. (2013). Suppressive effects of SuHeXiang Wan on amyloid-beta42-induced extracellular signal-regulated kinase hyperactivation and glial cell proliferation in a transgenic Drosophila model of Alzheimer's disease. Biol Pharm Bull 36: 390-398. PubMed ID: 23238278

  • Laura Lee Cell and Developmental Biology, Vanderbilt
    Wallace, H. A., Merkle, J. A., Yu, M. C., Berg, T. G., Lee, E., Bosco, G. and Lee, L. A. (2014). TRIP/NOPO E3 ubiquitin ligase promotes ubiquitylation of DNA polymerase eta. Development. PubMed ID: 24553286

    Sitaram, P., Hainline, S. G. and Lee, L. A. (2014). Cytological analysis of spermatogenesis: live and fixed preparations of Drosophila testes. J Vis Exp. PubMed ID: 24473184

    Sitaram, P., Merkle, J. A., Lee, E. and Lee, L. A. (2013). Asunder is required for dynein localization and dorsal fate determination during Drosophila oogenesis. Dev Biol. PubMed ID: 24333177

    Sitaram, P., Anderson, M. A., Jodoin, J. N., Lee, E. and Lee, L. A. (2012). Regulation of dynein localization and centrosome positioning by Lis-1 and asunder during Drosophila spermatogenesis. Development 139: 2945-2954. PubMed ID: 22764052

    Hanson, A. J., Wallace, H. A., Freeman, T. J., Beauchamp, R. D., Lee, L. A. and Lee, E. (2012). XIAP monoubiquitylates Groucho/TLE to promote canonical Wnt signaling. Mol Cell 45: 619-628. PubMed ID: 22304967

    Jernigan, K. K., Cselenyi, C. S., Thorne, C. A., Hanson, A. J., Tahinci, E., Hajicek, N., Oldham, W. M., Lee, L. A., Hamm, H. E., Hepler, J. R., Kozasa, T., Linder, M. E. and Lee, E. (2010). Gbetagamma activates GSK3 to promote LRP6-mediated beta-catenin transcriptional activity. Sci Signal 3: ra37. PubMed ID: 20460648

  • Seungbok Lee Department of Cell and Developmental Biology, Dental Research Institute, Seoul National University
    Park, H. G., Kim, Y. D., Cho, E., Lu, T. Y., Yao, C. K., Lee, J. and Lee, S. (2022). Vav independently regulates synaptic growth and plasticity through distinct actin-based processes. J Cell Biol 221(10). PubMed ID: 35976098

    Cho, J. H., Jo, M. G., Kim, E. S., Lee, N. Y., Kim, S. H., Chung, C. G., Park, J. H. and Lee, S. B. (2022). CBP-Mediated Acetylation of Importin α Mediates Calcium-Dependent Nucleocytoplasmic Transport of Selective Proteins in Drosophila Neurons. Mol Cells 45(11): 855-867. PubMed ID: 36172977

    Kim, S., Kim, J., Park, S., Park, J. J. and Lee, S. (2021). Drosophila Graf regulates mushroom body beta-axon extension and olfactory long-term memory. Mol Brain 14(1): 73. PubMed ID: 33892766

    Kim, E. S., Chung, C. G., Park, J. H., Ko, B. S., Park, S. S., Kim, Y. H., Cha, I. J., Kim, J., Ha, C. M., Kim, H. J. and Lee, S. B. (2021). C9orf72-associated arginine-rich dipeptide repeats induce RNA-dependent nuclear accumulation of Staufen in neurons. Hum Mol Genet. PubMed ID: 33783499

    Kim, J., Kim, S., Nahm, M., Li, T. N., Lin, H. C., Kim, Y. D., Lee, J., Yao, C. K. and Lee, S. (2021). ALS2 regulates endosomal trafficking, postsynaptic development, and neuronal survival. J Cell Biol 220(5). PubMed ID: 33683284

    Cha, I. J., Lee, D., Park, S. S., Chung, C. G., Kim, S. Y., Jo, M. G., Kim, S. Y., Lee, B. H., Lee, Y. S. and Lee, S. B. (2020). Ataxin-2 Dysregulation Triggers a Compensatory Fragile X Mental Retardation Protein Decrease in Drosophila C4da Neurons. Mol Cells 43(10): 870-879. PubMed ID: 33115979

    Park, J. H., Chung, C. G., Seo, J., Lee, B. H., Lee, Y. S., Kweon, J. H. and Lee, S. B. (2020). C9orf72-Associated Arginine-Rich Dipeptide Repeat Proteins Reduce the Number of Golgi Outposts and Dendritic Branches in Drosophila Neurons. Mol Cells 43(9): 821-830. PubMed ID: 32975212

    Heo, K., Nahm, M., Lee, M. J., Kim, Y. E., Ki, C. S., Kim, S. H. and Lee, S. (2017). The Rap activator Gef26 regulates synaptic growth and neuronal survival via inhibition of BMP signaling. Mol Brain 10(1): 62. PubMed ID: 29282074

    Kim, S., Nahm, M., Kim, N., Kwon, Y., Kim, J., Choi, S., Choi, E. Y., Shim, J., Lee, C. and Lee, S. (2017). Graf regulates hematopoiesis through GEEC endocytosis of EGFR. Development 144(22): 4159-4172. PubMed ID: 28993397

    Chung, C. G., Kwon, M. J., Jeon, K. H., Hyeon, D. Y., Han, M. H., Park, J. H., Cha, I. J., Cho, J. H., Kim, K., Rho, S., Kim, G. R., Jeong, H., Lee, J. W., Kim, T., Kim, K., Kim, K. P., Ehlers, M. D., Hwang, D. and B. (2017). Golgi Outpost Synthesis Impaired by Toxic Polyglutamine Proteins Contributes to Dendritic Pathology in Neurons. Cell Rep 20(2): 356-369. PubMed ID: 28700938

  • Siu Fai Lee Macquarie University & CSIRO, Australia
    Sgro, C. M., van Heerwaarden, B., Kellermann, V., Wee, C. W., Hoffmann, A. A. and Lee, S. F. (2013). Complexity of the genetic basis of ageing in nature revealed by a clinal study of lifespan and methuselah, a gene for ageing, in Drosophila from eastern Australia. Mol Ecol 22: 3539-3551. PubMed ID: 23802551

    Lee, S. F., Eyre-Walker, Y. C., Rane, R. V., Reuter, C., Vinti, G., Rako, L., Partridge, L. and Hoffmann, A. A. (2013). Polymorphism in the neurofibromin gene, Nf1, is associated with antagonistic selection on wing size and development time in Drosophila melanogaster. Mol Ecol 22: 2716-2725. PubMed ID: 23506114

    Hoffmann, A. A., Blacket, M. J., McKechnie, S. W., Rako, L., Schiffer, M., Rane, R. V., Good, R. T., Robin, C. and Lee, S. F. (2012). A proline repeat polymorphism of the Frost gene of Drosophila melanogaster showing clinal variation but not associated with cold resistance. Insect Mol Biol 21: 437-445. PubMed ID: 22708613

    Lee, S. F., White, V. L., Weeks, A. R., Hoffmann, A. A. and Endersby, N. M. (2012). High-throughput PCR assays to monitor Wolbachia infection in the dengue mosquito (Aedes aegypti) and Drosophila simulans. Appl Environ Microbiol 78: 4740-4743. PubMed ID: 22522691

  • Sung Bae Lee Lab of Neurodegenerative diseases and Aging, Daegu Gyeongbuk Institute of Science & Technology, Korea
    Chung, C. G., Kwon, M. J., Jeon, K. H., Hyeon, D. Y., Han, M. H., Park, J. H., Cha, I. J., Cho, J. H., Kim, K., Rho, S., Kim, G. R., Jeong, H., Lee, J. W., Kim, T., Kim, K., Kim, K. P., Ehlers, M. D., Hwang, D. and Lee, S. B. (2017). Golgi Outpost Synthesis Impaired by Toxic Polyglutamine Proteins Contributes to Dendritic Pathology in Neurons. Cell Rep 20(2): 356-369. PubMed ID: 28700938

    Cha, I. J., Lee, J. H., Cho, K. S. and Lee, S. B. (2017). Drosophila tensin plays an essential role in cell migration and planar polarity formation during oogenesis by mediating integrin-dependent extracellular signals to actin organization. Biochem Biophys Res Commun. PubMed ID: 28161642

    Cho, J. H., Park, J. H., Chung, C. G., Shim, H. J., Jeon, K. H., Yu, S. W. and Lee, S. B. (2015). Parkin-mediated responses against infection and wound involve TSPO-VDAC complex in Drosophila. Biochem Biophys Res Commun 463(1-2): 1-6. PubMed ID: 25979357

    Wang, X., Kim, J. R., Lee, S. B., Kim, Y. J., Jung, M. Y., Kwon, H. W. and Ahn, Y. J. (2014). Effects of curcuminoids identified in rhizomes of Curcuma longa on BACE-1 inhibitory and behavioral activity and lifespan of Alzheimer's disease Drosophila models. BMC Complement Altern Med 14: 88. PubMed ID: 24597901

    Lee, S. B., Bagley, J. A., Lee, H. Y., Jan, L. Y. and Jan, Y. N. (2011). Pathogenic polyglutamine proteins cause dendrite defects associated with specific actin cytoskeletal alterations in Drosophila. Proc Natl Acad Sci U S A 108(40): 16795-16800. PubMed ID: 21930920

  • Tina Lee Department of Biological Sciences, Carnegie Mellon University, Pittsburgh
    Crosby, D. and Lee, T. H. (2022). Membrane fusion by Drosophila atlastin does not require GTP hydrolysis. Mol Biol Cell 33(14): br23. PubMed ID: 36129776

    Elwell, J. A., Lovato, T. L., Adams, M. M., Baca, E. M., Lee, T. and Cripps, R. M. (2015). The myogenic repressor gene Holes in muscles is a direct transcriptional target of Twist and Tinman in the Drosophila embryonic mesoderm. Dev Biol 400(2): 266-276. PubMed ID: 25704510

    Schriner, S. E., Lee, K., Truong, S., Salvadora, K. T., Maler, S., Nam, A., Lee, T. and Jafari, M. (2013). Extension of Drosophila lifespan by Rhodiola rosea through a mechanism independent from dietary restriction. PLoS One 8(5): e63886. PubMed ID: 23704949

  • Tzu-Min Lee Janelia Farm, HHMI
    Lee, Y. J., Yang, C. P., Miyares, R. L., Huang, Y. F., He, Y., Ren, Q., Chen, H. M., Kawase, T., Ito, M., Otsuna, H., Sugino, K., Aso, Y., Ito, K. and Lee, T. (2020). Conservation and divergence of related neuronal lineages in the Drosophila central brain. Elife 9. PubMed ID: 32255422

    Chen, H. M., Marques, J. G., Sugino, K., Wei, D., Miyares, R. L. and Lee, T. (2020). CAMIO: a transgenic CRISPR pipeline to create diverse targeted genome deletions in Drosophila. Nucleic Acids Res. PubMed ID: 32187363

    Liu, L. Y., Long, X., Yang, C. P., Miyares, R. L., Sugino, K., Singer, R. H. and Lee, T. (2019). Mamo decodes hierarchical temporal gradients into terminal neuronal fate. Elife 8. PubMed ID: 31545163

    Garcia-Marques, J., Yang, C. P., Espinosa-Medina, I., Mok, K., Koyama, M. and Lee, T. (2019). Unlimited genetic switches for cell-type-specific manipulation. Neuron. PubMed ID: 31395429

    Ren, Q., Awasaki, T., Wang, Y. C., Huang, Y. F. and Lee, T. (2018). Lineage-guided Notch-dependent gliogenesis by Drosophila multi-potent progenitors. Development. Pubmed ID: 29764857

    Ren, Q., Yang, C. P., Liu, Z., Sugino, K., Mok, K., He, Y., Ito, M., Nern, A., Otsuna, H. and Lee, T. (2017). Stem Cell-Intrinsic, Seven-up-Triggered Temporal Factor Gradients Diversify Intermediate Neural Progenitors. Curr Biol [Epub ahead of print]. PubMed ID: 28434858

    Lee, T. (2017). Wiring the Drosophila Brain with Individually Tailored Neural Lineages. Curr Biol 27(2): R77-R82. PubMed ID: 28118595

    Ren, Q., Awasaki, T., Huang, Y.F., Liu, Z. and Lee, T. (2016). Cell class-lineage analysis reveals sexually dimorphic lineage compositions in the Drosophila brain. Curr Biol [Epub ahead of print]. PubMed ID: 27618265

    Liu, Z., Yang, C. P., Sugino, K., Fu, C. C., Liu, L. Y., Yao, X., Lee, L. P. and Lee, T. (2015). Opposing intrinsic temporal gradients guide neural stem cell production of varied neuronal fates. Science 350: 317-320. PubMed ID: 26472907

    Lee, T. (2014). Generating mosaics for lineage analysis in flies. Wiley Interdiscip Rev Dev Biol 3: 69-81. PubMed ID: 24902835

    Chen, H. M., Huang, Y., Pfeiffer, B. D., Yao, X. and Lee, T. (2015). An enhanced gene targeting toolkit for Drosophila: Golic+. Genetics [Epub ahead of print]. PubMed ID: 25555988

    Awasaki, T., Kao, C. F., Lee, Y. J., Yang, C. P., Huang, Y., Pfeiffer, B. D., Luan, H., Jing, X., Huang, Y. F., He, Y., Schroeder, M. D., Kuzin, A., Brody, T., Zugates, C. T., Odenwald, W. F. and Lee, T. (2014). Making Drosophila lineage-restricted drivers via patterned recombination in neuroblasts. Nat Neurosci. PubMed ID: 24561995

  • Won-Jae Lee School of Biological Science, Seoul National University and National Creative Research Initiative Center for Symbiosystem
    Lee, K. A., Cho, K. C., Kim, B., Jang, I. H., Nam, K., Kwon, Y. E., Kim, M., Hyeon, D. Y., Hwang, D., Seol, J. H. and Lee, W. J. (2018). Inflammation-Modulated Metabolic Reprogramming Is Required for DUOX-Dependent Gut Immunity in Drosophila. Cell Host Microbe 23(3): 338-352.e335. PubMed ID: 29503179

    Lee, K. A., Kim, B., Bhin, J., Kim do, H., You, H., Kim, E. K., Kim, S. H., Ryu, J. H., Hwang, D. and Lee, W. J. (2015). Bacterial uracil modulates Drosophila DUOX-dependent gut Immunity via Hedgehog-induced signaling endosomes. Cell Host Microbe 17: 191-204. PubMed ID: 25639794

    Kim, S. H. and Lee, W. J. (2014). Role of DUOX in gut inflammation: lessons from model of gut-microbiota interactions. Front Cell Infect Microbiol 3: 116. PubMed ID: 24455491

    Lee, K. A., Kim, S. H., Kim, E. K., Ha, E. M., You, H., Kim, B., Kim, M. J., Kwon, Y., Ryu, J. H., Lee, W. J. (2013) Bacterial-derived uracil as a modulator of mucosal immunity and gut-microbe homeostasis in Drosophila. Cell 153: 797-811. PubMed ID: 23663779

    Nam, H. J., Jang, I. H., You, H., Lee, K. A. and Lee, W. J. (2012). Genetic evidence of a redox-dependent systemic wound response via Hayan protease-phenoloxidase system in Drosophila. EMBO J 31: 1253-1265. PubMed ID: 22227521

    Shin, S. C., Kim, S. H., You, H., Kim, B., Kim, A. C., Lee, K. A., Yoon, J. H., Ryu, J. H. and Lee, W. J. (2011). Drosophila microbiome modulates host developmental and metabolic homeostasis via insulin signaling. Science 334: 670-674. PubMed ID: 22053049

  • Youngseok Lee Departments of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, Korea
    Aryal, B., Dhakal, S., Shrestha, B., Sang, J., Nhuchhen Pradhan, R. and Lee, Y. (2022). Protocol for binary food choice assays using Drosophila melanogaster. STAR Protoc 3(2): 101410. PubMed ID: 35620079

    Aryal, B. and Lee, Y. (2022). Histamine avoidance through three gustatory receptors in Drosophila melanogaster. Insect Biochem Mol Biol 144: 103760. PubMed ID: 35346814

    Aryal, B., Dhakal, S., Shrestha, B. and Lee, Y. (2022). Molecular and neuronal mechanisms for amino acid taste perception in the Drosophila labellum. Curr Biol 32(6): 1376-1386.e1374. PubMed ID: 35176225

    Shrestha, B., Nhuchhen Pradhan, R., Nath, D. K. and Lee, Y. (2021). Cellular and molecular basis of IR3535 perception in Drosophila. Pest Manag Sci. PubMed ID: 34708523

    Dhakal, S., Sang, J., Aryal, B. and Lee, Y. (2021). Ionotropic receptors mediate nitrogenous waste avoidance in Drosophila melanogaster. Commun Biol 4(1): 1281. PubMed ID: 34773080

    Rimal, S., Sang, J., Dhakal, S. and Lee, Y. (2020). Cucurbitacin B Activates Bitter-Sensing Gustatory Receptor Neurons via Gustatory Receptor 33a in Drosophila melanogaster. Mol Cells. PubMed ID: 32451368

    Rimal, S. and Lee, Y. (2019). Molecular sensor of nicotine in taste of Drosophila melanogaster. Insect Biochem Mol Biol: 103178. PubMed ID: 31226410

    Poudel, S., Kim, Y., Gwak, J. S., Jeong, S. and Lee, Y. (2017). Gustatory receptor 22e is essential for sensing chloroquine and strychnine in Drosophila melanogaster. Insect Biochem Mol Biol 88: 30-36. PubMed ID: 28751111

    Lee, J. E., Kim, Y., Kim, K. H., Lee do, Y. and Lee, Y. (2016). Contribution of Drosophila TRPA1 to metabolism. PLoS One 11: e0152935. PubMed ID: 27055172

    Poudel, S. and Lee, Y. (2016). Gustatory Receptors Required for Avoiding the Toxic Compound Coumarin in Drosophila melanogaster. Mol Cells [Epub ahead of print]. PubMed ID: 26912085

    Lee, Y. (2013). Contribution of Drosophila TRPA1-Expressing Neurons to Circadian Locomotor Activity Patterns. PLoS One 8: e85189. PubMed ID: 24367706

  • Fritz-Olaf Lehmann Universität Rostock
    Bartussek, J. and Lehmann, F. O. (2016). Proprioceptive feedback determines visuomotor gain in Drosophila. R Soc Open Sci 3: 150562. PubMed ID: 26909184

    Lehmann, F. O., Skandalis, D. A. and Berthe, R. (2013). Calcium signalling indicates bilateral power balancing in the Drosophila flight muscle during manoeuvring flight. J R Soc Interface 10: 20121050. PubMed ID: 23486171

    Lehmann, F. O. and Cierotzki, V. (2010). Locomotor performance in the Drosophila brain mutant drop-dead. Comp Biochem Physiol A Mol Integr Physiol 156: 337-343. PubMed ID: 20045484

  • Michael Lehmann Department of Biological Sciences, University of Arkansas, Fayetteville.
    Hood, S. E., Kofler, X. V., Chen, Q., Scott, J., Ortega, J. and Lehmann, M. (2020). Nuclear translocation ability of Lipin differentially affects gene expression and survival in fed and fasting Drosophila. J Lipid Res. PubMed ID: 32989002

    Schmitt, S., Ugrankar, R., Greene, S.E., Prajapati, M. and Lehmann, M. (2015). Drosophila lipin interacts with insulin and TOR signaling pathways in the control of growth and lipid metabolism. J Cell Sci [Epub ahead of print]. PubMed ID: 26490996

    Schmitt, S., Ugrankar, R., Greene, S. E., Prajapati, M. and Lehmann, M. (2015). Drosophila lipin interacts with insulin and TOR signaling pathways in the control of growth and lipid metabolism. J Cell Sci. PubMed ID: 26490996

  • Ruth Lehmann The Skirball Institute of Biomolecular Medicine, New York University School of Medicine
    Lin, B., Luo, J. and Lehmann, R. (2022). An AMPK phosphoregulated RhoGEF feedback loop tunes cortical flow-driven amoeboid migration in vivo. Sci Adv 8(37): eabo0323. PubMed ID: 36103538

    Gebert, D., Neubert, L. K., Lloyd, C., Gui, J., Lehmann, R. and Teixeira, F. K. (2021). Large Drosophila germline piRNA clusters are evolutionarily labile and dispensable for transposon regulation. Mol Cell. PubMed ID: 34352205

    Slaidina, M., Gupta, S., Banisch, T. U. and Lehmann, R. (2021). A single-cell atlas reveals unanticipated cell type complexity in Drosophila ovaries. Genome Res. PubMed ID: 34389661

    Lin, B., Luo, J. and Lehmann, R. (2020). Collectively stabilizing and orienting posterior migratory forces disperses cell clusters in vivo. Nat Commun 11(1): 4477. PubMed ID: 32901019

    Slaidina, M., Banisch, T. U., Gupta, S. and Lehmann, R. (2020). A single-cell atlas of the developing Drosophila ovary identifies follicle stem cell progenitors. Genes Dev 34(3-4): 239-249. PubMed ID: 31919193

    Coux, R. X., Teixeira, F. K. and Lehmann, R. (2018). L(3)mbt and the LINT complex safeguard cellular identity in the Drosophila ovary. Development. PubMed ID: 29511022

    LeBlanc, M. G. and Lehmann, R. (2017). Domain-specific control of germ cell polarity and migration by multifunction Tre1 GPCR. J Cell Biol 216(9): 2945-2958. PubMed ID: 28687666

    Pae, J., Cinalli, R. M., Marzio, A., Pagano, M. and Lehmann, R. (2017). GCL and CUL3 control the switch between Cell lineages by mediating localized degradation of an RTK. Dev Cell 42(2): 130-142.e137. PubMed ID: 28743001

    LeBlanc, M. G. and Lehmann, R. (2017). Domain-specific control of germ cell polarity and migration by multifunction Tre1 GPCR. J Cell Biol [Epub ahead of print]. PubMed ID: 28687666

    Trcek, T., Lionnet, T., Shroff, H. and Lehmann, R. (2017). mRNA quantification using single-molecule FISH in Drosophila embryos. Nat Protoc 12(7): 1326-1348. PubMed ID: 28594816

  • Christian Lehner Institute of Molecular Life Sciences, University of Zurich
    Kabakci, Z., Yamada, H., Vernizzi, L., Gupta, S., Weber, J., Sun, M. S. and L (2022). Teflon promotes chromosomal recruitment of homolog conjunction proteins during Drosophila male meiosis. PLoS Genet 18(10): e1010469. PubMed ID: 36251690

    Kabakci, Z., Reichle, H. E., Lemke, B., Rousova, D., Gupta, S., Weber, J., Schleiffer, A., Weir, J. R. and Lehner, C. F. (2022). Homologous chromosomes are stably conjoined for Drosophila male meiosis I by SUM, a multimerized protein assembly with modules for DNA-binding and for separase-mediated dissociation co-opted from cohesin. PLoS Genet 18(12): e1010547. PubMed ID: 36480577

    Ghosh, S. and Lehner, C. F. (2022). Incorporation of CENP-A/CID into centromeres during early Drosophila embryogenesis does not require RNA polymerase II-mediated transcription. Chromosoma. PubMed ID: 35015118

    Bai, Y., Caussinus, E., Leo, S., Bosshardt, F., Myachina, F., Rot, G., Robinson, M. D. and Lehner, C. F. (2021). A cis-regulatory element promoting increased transcription at low temperature in cultured ectothermic Drosophila cells. BMC Genomics 22(1): 771. PubMed ID: 34711176

    Vernizzi, L. and Lehner, C. F. (2021). Bivalent individualization during chromosome territory formation in Drosophila spermatocytes by controlled condensin II protein activity and additional force generators. PLoS Genet 17(10): e1009870. PubMed ID: 34669718

    Weber, J., Kabakci, Z., Chaurasia, S., Brunner, E. and Lehner, C. F. (2020). Chromosome separation during Drosophila male meiosis I requires separase-mediated cleavage of the homolog conjunction protein UNO. PLoS Genet 16(10): e1008928. PubMed ID: 33001976

    Sun, M. S., Weber, J., Blattner, A. C., Chaurasia, S. and Lehner, C. F. (2019). MNM and SNM maintain but do not establish achiasmate homolog conjunction during Drosophila male meiosis. PLoS Genet 15(5): e1008162. PubMed ID: 31136586

    Blattner, A. C., Chaurasia, S., McKee, B. D. and Lehner, C. F. (2016). Separase is required for homolog and sister disjunction during Drosophila melanogaster male meiosis, but not for biorientation of sister centromeres. PLoS Genet 12: e1005996. PubMed ID: 27120695

    Blattner, A. C., Aguilar-Rodriguez, J., Kranzlin, M., Wagner, A. and Lehner, C. F. (2016). Drosophila Nnf1 paralogs are partially redundant for somatic and germ line kinetochore function. Chromosoma [Epub ahead of print]. PubMed ID: 26892014

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

    Urban, E., Nagarkar-Jaiswal, S., Lehner, C. F. and Heidmann, S. K. (2014). The Cohesin Subunit Rad21 Is Required for Synaptonemal Complex Maintenance, but Not Sister Chromatid Cohesion, during Drosophila Female Meiosis. PLoS Genet 10: e1004540. PubMed ID: 25101996

  • Elissa Lei NIDDK, National Institutes of Health, Bethesda
    Bag, I., Chen, Y., D'Orazio, K., Lopez, P., Wenzel, S., Takagi, Y. and Lei, E. P. (2022). Isha is a su(Hw) mRNA-binding protein required for gypsy insulator function. G3 (Bethesda). PubMed ID: 35708663

    Chen, S., Rosin, L. F., Pegoraro, G., Moshkovich, N., Murphy, P. J., Yu, G. and Lei, E. P. (2022). NURF301 contributes to gypsy chromatin insulator-mediated nuclear organization. Nucleic Acids Res 50(14): 7906-7924. PubMed ID: 35819192

    Chen, D., Brovkina, M., Matzat, L. H. and Lei, E. P. (2019). Shep RNA-Binding Capacity Is Required for Antagonism of gypsy Chromatin Insulator Activity. G3 (Bethesda). PubMed ID: 30630880

    Nazer, E., Dale, R. K., Palmer, C. and Lei, E. P. (2018). Argonaute2 attenuates active transcription by limiting RNA Polymerase II elongation in Drosophila melanogaster. Sci Rep 8(1): 15685. PubMed ID: 29529026

    Chen, D., Dale, R. K. and Lei, E. P. (2017). Shep regulates Drosophila neuronal remodeling by controlling transcription of its chromatin targets. Development [Epub ahead of print]. PubMed ID: 29158441

    Chinen, M. and Lei, E. P. (2017). Drosophila Argonaute2 turnover is regulated by the ubiquitin proteasome pathway. Biochem Biophys Res Commun [Epub ahead of print]. PubMed ID: 28087276

    Dale, R. K., Matzat, L. H. and Lei, E. P. (2014). metaseq: a Python package for integrative genome-wide analysis reveals relationships between chromatin insulators and associated nuclear mRNA. Nucleic Acids Res. PubMed ID: 25063299

    King, M. R., Matzat, L. H., Dale, R. K., Lim, S. J. and Lei, E. P. (2014). The RNA-binding protein Rumpelstiltskin antagonizes gypsy chromatin insulator function in a tissue-specific manner. J Cell Sci. PubMed ID: 24706949

    Nazer, E. and Lei, E. P. (2014). Modulation of chromatin modifying complexes by noncoding RNAs in trans. Curr Opin Genet Dev 25C: 68-73. PubMed ID: 24534715

  • Jeff Leips University of Maryland, Baltimore County
    Gabrawy, M. M., Khosravian, N., Morcos, G. S., Morozova, T. V., Jezek, M., Walston, J. D., Huang, W., Abadir, P. M. and Leips, J. (2022). Genome-Wide Analysis in Drosophila Reveals the Genetic Basis of Variation in Age-Specific Physical Performance and Response to ACE Inhibition. Genes (Basel) 13(1). PubMed ID: 35052483

    Durham, M. F., Magwire, M. M., Stone, E. A. and Leips, J. (2014). Genome-wide analysis in Drosophila reveals age-specific effects of SNPs on fitness traits. Nat Commun 5: 4338. PubMed ID: 25000897

    Hodges, T. K., Laskowski, K. L., Squadrito, G. L., De Luca, M., Leips, J. (2013) Defense Traits of Larval Drosophila Melanogaster Exhibit Genetically Based Trade-Offs against Different Species of Parasitoids. Evolution 67: 749-760. PubMed ID: 23461325

    Remolina, S. C., Chang, P. L., Leips, J., Nuzhdin, S. V. and Hughes, K. A. (2012). Genomic basis of aging and life-history evolution in Drosophila melanogaster. Evolution 66: 3390-3403. PubMed ID: 23106705

    Felix, T. M., Hughes, K. A., Stone, E. A., Drnevich, J. M. and Leips, J. (2012). Age-specific variation in immune response in Drosophila melanogaster has a genetic basis. Genetics 191: 989-1002. PubMed ID: 22554890

    De Luca, M., Klimentidis, Y. C., Casazza, K., Chambers, M. M., Cho, R., Harbison, S. T., Jumbo-Lucioni, P., Zhang, S., Leips, J. and Fernandez, J. R. (2010). A conserved role for syndecan family members in the regulation of whole-body energy metabolism. PLoS One 5: e11286. PubMed ID: 20585652

  • Bruno Lemaitre EPFL, Lausanne, Switzerland
    Hanson, M. A. and Lemaitre, B. (2022). Repeated truncation of a modular antimicrobial peptide gene for neural context. PLoS Genet 18(6): e1010259. PubMed ID: 35714143

    Hanson, M. A., Kondo, S. and Lemaitre, B. (2022). Drosophila immunity: the Drosocin gene encodes two host defence peptides with pathogen-specific roles. Proc Biol Sci 289(1977): 20220773. PubMed ID: 35730150

    Carboni, A. L., Hanson, M. A., Lindsay, S. A., Wasserman, S. A. and Lemaitre, B. (2021). Cecropins contribute to Drosophila host defense against a subset of fungal and Gram-negative bacterial infection. Genetics. PubMed ID: 34791204

    Petrignani, B., Rommelaere, S., Hakim-Mishnaevski, K., Masson, F., Ramond, E., Hilu-Dadia, R., Poidevin, M., Kondo, S., Kurant, E. and Lemaitre, B. (2021). A secreted factor NimrodB4 promotes the elimination of apoptotic corpses by phagocytes in Drosophila. EMBO Rep 22(9): e52262. PubMed ID: 34370384

    Hanson, M. A., Cohen, L. B., Marra, A., Iatsenko, I., Wasserman, S. A. and Lemaitre, B. (2021). The Drosophila Baramicin polypeptide gene protects against fungal infection. PLoS Pathog 17(8): e1009846. PubMed ID: 34432851

    Marra, A., Hanson, M. A., Kondo, S., Erkosar, B. and Lemaitre, B. (2021). Drosophila Antimicrobial Peptides and Lysozymes Regulate Gut Microbiota Composition and Abundance. mBio: e0082421. PubMed ID: 34253067

    Ramond, E., Dudzic, J. P. and Lemaitre, B. (2020). Comparative RNA-Seq analyses of Drosophila plasmatocytes reveal gene specific signatures in response to clean injury and septic injury. PLoS One 15(6): e0235294. PubMed ID: 32598400

    Li, X., Rommelaere, S., Kondo, S. and Lemaitre, B. (2020). Renal Purge of Hemolymphatic Lipids Prevents the Accumulation of ROS-Induced Inflammatory Oxidized Lipids and Protects Drosophila from Tissue Damage. Immunity 52(2): 374-387. PubMed ID: 32075729

    Iatsenko, I., Marra, A., Boquete, J. P., Pena, J. and Lemaitre, B. (2020). Iron sequestration by transferrin 1 mediates nutritional immunity in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 32188787

    Rommelaere, S., Boquete, J. P., Piton, J., Kondo, S. and Lemaitre, B. (2019). The exchangeable spolipoprotein Nplp2 sustains lipid flow and heat acclimation in Drosophila. Cell Rep 27(3): 886-899.e886. PubMed ID: 30995484

    Melcarne, C., Ramond, E., Dudzic, J., Bretscher, A. J., Kurucz, E., Ando, I. and Lemaitre, B. (2019). Two Nimrod receptors, NimC1 and Eater, synergistically contribute to bacterial phagocytosis in Drosophila melanogaster. Febs j. PubMed ID: 30993828

    Dudzic, J. P., Hanson, M. A., Iatsenko, I., Kondo, S. and Lemaitre, B. (2019). More Than Black or White: Melanization and Toll Share Regulatory Serine Proteases in Drosophila. Cell Rep 27(4): 1050-1061.e1053. PubMed ID: 31018123

    Iatsenko, I., Boquete, J. P. and Lemaitre, B. (2018). Microbiota-derived lactate activates production of reactive oxygen species by the intestinal NADPH Oxidase Nox and shortens Drosophila lifespan. Immunity 49(5): 929-942.e925. PubMed ID: 30446385

    Zhai, Z., Boquete, J. P. and Lemaitre, B. (2018). Cell-specific Imd-NF-kappaB responses enable simultaneous antibacterial immunity and intestinal epithelial cell shedding upon bacterial infection. Immunity 48(5): 897-910.e897. Pubmed ID: 29752064

  • Steffen Lemke Centre for Organismal Studies, Universität Heidelberg
    Urbansky, S., Gonzalez Avalos, P., Wosch, M. and Lemke, S. (2016). Folded gastrulation and T48 drive the evolution of coordinated mesoderm internalization in flies. Elife 5 [Epub ahead of print]. PubMed ID: 27685537

    Lemke, S., Antonopoulos, D. A., Meyer, F., Domanus, M. H., Schmidt-Ott, U. (2011). BMP signaling components in embryonic transcriptomes of the hover fly Episyrphus balteatus (Syrphidae). BMC Genomics.12:278.

  • Bernardo Lemos School of Public Health, Harvard University
    Wang, M. and Lemos, B. (2019). Ribosomal DNA harbors an evolutionarily conserved clock of biological aging. Genome Res. PubMed ID: 30765617']

    Wang, M. and Lemos, B. (2019). Ribosomal DNA harbors an evolutionarily conserved clock of biological aging. Genome Res. PubMed ID: 30765617

    Wang, M., Branco, A. T. and Lemos, B. (2017). The Y chromosome modulates splicing and sex-biased intron retention rates in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 29263027

    Branco, A. T., Brito, R. M. and Lemos, B. (2017). Sex-specific adaptation and genomic responses to Y chromosome presence in female reproductive and neural tissues. Proc Biol Sci 284(1869). PubMed ID: 29237855

    Branco, A.T., Schilling, L., Silkaitis, K., Dowling, D.K. and Lemos, B. (2016). Reproductive activity triggers accelerated male mortality and decreases lifespan: genetic and gene expression determinants in Drosophila. Heredity (Edinb) [Epub ahead of print]. PubMed ID: 27731328

    Francisco, F. O. and Lemos, B. (2014). How do y-chromosomes modulate genome-wide epigenetic States: genome folding, chromatin sinks, and gene expression. J Genomics 2: 94-103. PubMed ID: 25057325

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

    Branco, A. T., Tao, Y., Hartl, D. L. and Lemos, B. (2013). Natural variation of the Y chromosome suppresses sex ratio distortion and modulates testis-specific gene expression in Drosophila simulans. Heredity (Edinb). PubMed ID: 23591516

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

  • Pierre-françois Lenne Institut de Biologie du Développement de Marseille
    Toret, C. P., Shivakumar, P. C., Lenne, P. F. and Le Bivic, A. (2018). The elmo-mbc complex and rhogap19d couple Rho family GTPases during mesenchymal-to-epithelial-like transitions. Development. PubMed ID: 29437779

    Chan, E. H., Chavadimane Shivakumar, P., Clement, R., Laugier, E. and Lenne, P. F. (2017). Patterned cortical tension mediated by N-cadherin controls cell geometric order in the Drosophila eye. Elife 6. PubMed ID: 28537220

    Shivakumar, P. C. and Lenne, P. F. (2016). Laser Ablation to Probe the Epithelial Mechanics in Drosophila. Methods Mol Biol 1478: 241-251. PubMed ID: 27730586

    Markova, O., Senatore, S., Chardes, C. and Lenne, P. F. (2015). Calcium Spikes in Epithelium: study on Drosophila early embryos. Sci Rep 5: 11379. PubMed ID: 26198871

    Truong Quang, B. A. and Lenne, P. F. (2015). Superresolution measurements in vivo: imaging Drosophila embryo by photoactivated localization microscopy. Methods Cell Biol 125: 119-142. PubMed ID: Pierre Léopold Institute of Biology Valrose, Université Nice
    Vijendravarma, R. K., Narasimha, S., Steinfath, E., Clemens, J. and Leopold, P. (2022). Drosophila females have an acoustic preference for symmetric males. Proc Natl Acad Sci U S A 119(13): e2116136119. PubMed ID: 35312357

    Santabarbara-Ruiz, P. and Leopold, P. (2021). An Oatp transporter-mediated steroid sink promotes tumor-induced cachexia in Drosophila. Dev Cell 56(19): 2741-2751. PubMed ID: 34610327

    Arquier, N., Bjordal, M., Hammann, P., Kuhn, L. and Leopold, P. (2021). Brain adiponectin signaling controls peripheral insulin response in Drosophila. Nat Commun 12(1): 5633. PubMed ID: 34561451

    Boulan, L., Andersen, D., Colombani, J., Boone, E. and Leopold, P. (2019). Inter-Organ Growth Coordination Is Mediated by the Xrp1-Dilp8 Axis in Drosophila. Dev Cell. PubMed ID: 31006647

    Boone, E., Colombani, J., Andersen, D. S. and Leopold, P. (2016). The Hippo signalling pathway coordinates organ growth and limits developmental variability by controlling dilp8 expression. Nat Commun 7: 13505. PubMed ID: 27874005

    Delanoue, R., Meschi, E., Agrawal, N., Mauri, A., Tsatskis, Y., McNeill, H. and Léopold, P. (2016). Drosophila insulin release is triggered by adipose Stunted ligand to brain Methuselah receptor. Science 353: 1553-1556. PubMed ID: 27708106

    Agrawal, N., Delanoue, R., Mauri, A., Basco, D., Pasco, M., Thorens, B. and Leopold, P. (2016). The Drosophila TNF Eiger Is an Adipokine that Acts on Insulin-Producing Cells to Mediate Nutrient Response. Cell Metab 23: 675-684. PubMed ID: 27076079

    Andersen, D. S., Colombani, J., Palmerini, V., Chakrabandhu, K., Boone, E., Rothlisberger, M., Toggweiler, J., Basler, K., Mapelli, M., Hueber, A. O. and Leopold, P. (2015). The Drosophila TNF receptor Grindelwald couples loss of cell polarity and neoplastic growth. Nature. PubMed ID: 25874673

    Bjordal, M., Arquier, N., Kniazeff, J., Pin, J. P. and Leopold, P. (2014). Sensing of amino acids in a dopaminergic circuitry promotes rejection of an incomplete diet in Drosophila. Cell 156: 510-521. PubMed ID: 24485457

  • Maria Leptin European Molecular Biology Laboratory, Heidelberg
    Hafer, T. L., Patra, S., Tagami, D. and Kohwi, M. (2022). Enhancer of trithorax/polycomb, Corto, regulates timing of hunchback gene relocation and competence in Drosophila neuroblasts. Neural Dev 17(1): 3. PubMed ID: 35177098

    Rios-Barrera, L. D. and Leptin, M. (2022). An endosome-associated actin network involved in directed apical plasma membrane growth. J Cell Biol 221(3). PubMed ID: 35061016

    Bhide, S., Gombalova, D., Monke, G., Stegmaier, J., Zinchenko, V., Kreshuk, A., Belmonte, J. M. and Leptin, M. (2021). Mechanical competition alters the cellular interpretation of an endogenous genetic program. J Cell Biol 220(11). PubMed ID: 34449835

    Mathew, R., Rios-Barrera, L. D., Machado, P., Schwab, Y. and Leptin, M. (2020). Transcytosis via the late endocytic pathway as a cell morphogenetic mechanism. Embo j: e105332. PubMed ID: 32657472

    Best, B. T. and Leptin, M. (2020). Multiple Requirements for Rab GTPases in the Development of Drosophila Tracheal Dorsal Branches and Terminal Cells. G3 (Bethesda). PubMed ID: 31980432

    Kakanj, P., Moussian, B., Grönke, S., Bustos, V., Eming, S.A., Partridge, L. and Leptin, M. (2016). Insulin and TOR signal in parallel through FOXO and S6K to promote epithelial wound healing. Nat Commun 7: 12972. PubMed ID: 27713427

    Misra, M., Edmund, H., Ennis, D., Schlueter, M. A., Marot, J. E., Tambasco, J., Barlow, I., Sigurbjornsdottir, S., Mathew, R., Valles, A. M., Davis, I., Leptin, M. and Gavis, E. R. (2016). A Genome-Wide Screen for Dendritically Localized RNAs Identifies Genes Required for Dendrite Morphogenesis. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27260999

    Rauzi, M., Krzic, U., Saunders, T. E., Krajnc, M., Ziherl, P., Hufnagel, L. and Leptin, M. (2015). Embryo-scale tissue mechanics during Drosophila gastrulation movements. Nat Commun 6: 8677. PubMed ID: 26497898

    Sigurbjornsdottir, S., Mathew, R. and Leptin, M. (2014). Molecular mechanisms of de novo lumen formation. Nat Rev Mol Cell Biol. PubMed ID: 25186133

    Ukken, F. P., Aprill, I., JayaNandanan, N. and Leptin, M. (2014). Slik and the Receptor Tyrosine Kinase Breathless Mediate Localized Activation of Moesin in Terminal Tracheal Cells. PLoS One 9: e103323. PubMed ID: 25061859

    Schafer, G., Narasimha, M., Vogelsang, E. and Leptin, M. (2014). Cadherin switching during the formation and differentiation of the Drosophila mesoderm: implications for epithelial mesenchymal transitions. J Cell Sci. PubMed ID: 24496448

    Jayanandanan, N., Mathew, R. and Leptin, M. (2014). Guidance of subcellular tubulogenesis by actin under the control of a synaptotagmin-like protein and Moesin. Nat Commun 5: 3036. PubMed ID: 24413568

  • Dorothy Lerit Department of Cell Biology at Emory University School of Medicine, Atlanta, Georgia
    Fang, J. and Lerit, D. A. (2022). Orb-dependent polyadenylation contributes to PLP expression and centrosome scaffold assembly. Development 149(13). PubMed ID: 35661190

    Mehta, D. S., Zein-Sabatto, H., Ryder, P. V., Lee, J. and Lerit, D. A. (2022). Drosophila centrocortin is dispensable for centriole duplication but contributes to centrosome separation. G3 (Bethesda) 12(2). PubMed ID: 35100335

    Colonnetta, M. M., Lym, L. R., Wilkins, L., Kappes, G., Castro, E. A., Ryder, P. V., Schedl, P., Lerit, D. A. and Deshpande, G. (2021). Antagonism between germ cell-less and Torso receptor regulates transcriptional quiescence underlying germline/soma distinction. Elife 10. PubMed ID: 33459591

    Ryder, P. V., Fang, J. and Lerit, D. A. (2020). centrocortin RNA localization to centrosomes is regulated by FMRP and facilitates error-free mitosis. J Cell Biol 219(12). PubMed ID: 33196763

    Ryder, P. V. and Lerit, D. A. (2020). Quantitative analysis of subcellular distributions with an open-source, object-based tool. Biol Open 9(10). PubMed ID: 32973081

  • Anthea Letsou Department of Human Genetics, University of Utah, Salt Lake City
    Moulton, M. J., Humphreys, G. B., Kim, A. and Letsou, A. (2020). O-GlcNAcylation Dampens Dpp/BMP Signaling to Ensure Proper Drosophila Embryonic Development. Dev Cell 53(3): 330-343.e333. PubMed ID: 32369743

    Sivachenko, A., Gordon, H. B., Kimball, S. S., Gavin, E. J., Bonkowsky, J. L. and Letsou, A. (2016). Neurodegeneration in a Drosophila model of Adrenoleukodystrophy: the roles of the bubblegum and double bubble acyl-CoA synthetases. Dis Model Mech [Epub ahead of print]. PubMed ID: 26893370

    Humphreys, G. B., Jud, M. C., Monroe, K. M., Kimball, S. S., Higley, M., Shipley, D., Vrablik, M. C., Bates, K. L. and Letsou, A. (2013). Mummy, A UDP-N-Acetylglucosamine Pyrophosphorylase, modulates DPP signaling in the embryonic epidermis of Drosophila. Dev Biol. PubMed ID: 23796903

    Bates, K. L., Higley, M. and Letsou, A. (2008). Raw mediates antagonism of AP-1 activity in Drosophila. Genetics 178: 1989-2002. PubMed ID: 18430930

  • François Leulier Institut de Génomique Fonctionnelle de Lyon
    Consuegra, J., Grenier, T., Baa-Puyoulet, P., Rahioui, I., Akherraz, H., Gervais, H., Parisot, N., da Silva, P., Charles, H., Calevro, F. and Leulier, F. (2020). Drosophila-associated bacteria differentially shape the nutritional requirements of their host during juvenile growth. PLoS Biol 18(3): e3000681. PubMed ID: 32196485

    Ma, D., Bou-Sleiman, M., Joncour, P., Indelicato, C. E., Frochaux, M., Braman, V., Litovchenko, M., Storelli, G., Deplancke, B. and Leulier, F. (2019). Commensal gut bacteria buffer the impact of host genetic variants on Drosophila developmental traits under nutritional stress. iScience 19: 436-447. PubMed ID: 31422284

    Ma, D. and Leulier, F. (2018). The importance of being persistent: The first true resident gut symbiont in Drosophila. PLoS Biol 16(8): e2006945. PubMed ID: 30071013

    Tefit, M. A., Gillet, B., Joncour, P., Hughes, S. and Leulier, F. (2017). Stable association of a Drosophila-derived microbiota with its animal partner and the nutritional environment throughout a fly population's life cycle. J Insect Physiol. PubMed ID: 28916161

    Ma, D., Storelli, G., Mitchell, M. L. and Leulier, F. (2015). Studying host-microbiota mutualism in Drosophila: Harnessing the power of gnotobiotic flies. Biomed J [Epub ahead of print]. PubMed ID: 26068125

    Matos, R. C. and Leulier, F. (2014). Lactobacilli-Host mutualism: 'learning on the fly'. Microb Cell Fact 13 Suppl 1: S6. PubMed ID: 25186369

    Erkosar, B. and Leulier, F. (2014). Transient adult microbiota, gut homeostasis and longevity: Novel insights from the Drosophila model. FEBS Lett. PubMed ID: 24983497

    Wayland, M. T., Defaye, A., Rocha, J., Jayaram, S. A., Royet, J., Miguel-Aliaga, I., Leulier, F. and Cognigni, P. (2014). Spotting the differences: Probing host/microbiota interactions with a dedicated software tool for the analysis of faecal outputs in Drosophila. J Insect Physiol. PubMed ID: 24907675

    Erkosar Combe, B., Defaye, A., Bozonnet, N., Puthier, D., Royet, J. and Leulier, F. (2014). Drosophila microbiota modulates host metabolic gene expression via IMD/NF-kappaB signaling. PLoS One 9: e94729. PubMed ID: 24733183

  • Joel Levine University of Toronto, Mississauga
    Alwash, N., Allen, A. M., Sokolowski, M. B.. and Levine, J. D. (2021). The Drosophila melanogaster foraging gene affects social networks. J Neurogenet: 1-13. PubMed ID: 34121597

    Rooke, R., Rasool, A., Schneider, J. and Levine, J. D. (2020). Drosophila melanogaster behaviour changes in different social environments based on group size and density. Commun Biol 3(1): 304. PubMed ID: 32533063

    Jezovit, J. A., Rooke, R., Schneider, J. and Levine, J. D. (2020). Behavioral and environmental contributions to drosophilid social networks. Proc Natl Acad Sci U S A 117(21): 11573-11583. PubMed ID: 32404421

    Krupp, J. J., Nayal, K., Wong, A., Millar, J. G. and Levine, J. D. (2019). Desiccation resistance is an adaptive life-history trait dependent upon cuticular hydrocarbons, and influenced by mating status and temperature in D. melanogaster. J Insect Physiol: 103990. PubMed ID: 31830467

    Schneider, J., Murali, N., Taylor, G. W. and Levine, J. D. (2018). Can Drosophila melanogaster tell who's who? PLoS One 13(10): e0205043. PubMed ID: 30356241

    Gorter, J. A., Jagadeesh, S., Gahr, C., Boonekamp, J. J., Levine, J. D. and Billeter, J. C. (2016). The nutritional and hedonic value of food modulate sexual receptivity in Drosophila melanogaster females. Sci Rep 6: 19441. PubMed ID: 26777264

    Krupp, J. J. and Levine, J. D. (2014). Neural circuits: anatomy of a sexual behavior. Curr Biol 24: R327-329. PubMed ID: 24735858

    Bloch, G., Herzog, E. D., Levine, J. D. and Schwartz, W. J. (2013). Socially synchronized circadian oscillators. Proc Biol Sci 280: 20130035. PubMed ID: 23825203

    Krupp, J. J., Billeter, J. C., Wong, A., Choi, C., Nitabach, M. N. and Levine, J. D. (2013). Pigment-Dispersing Factor Modulates Pheromone Production in Clock Cells that Influence Mating in Drosophila. Neuron 79: 54-68. PubMed ID: 23849197

    Bloch, G., Herzog, E. D., Levine, J. D. and Schwartz, W. J. (2013). Socially synchronized circadian oscillators. Proc Biol Sci 280: 20130035. PubMed ID: 23825203

  • Mia Levine Department of Biology, University of Pennsylvania
    Brand, C. L. and Levine, M. T. (2022). Cross-species incompatibility between a DNA satellite and the Drosophila Spartan homolog poisons germline genome integrity. Curr Biol. PubMed ID: 35643081

    Lee, Y. C., Leek, C. and Levine, M. T. (2017). Recurrent Innovation at Genes Required for Telomere Integrity in Drosophila. Mol Biol Evol 34(2): 467-482. PubMed ID: 27836984

    >Levine, M. T., Vander Wende, H. M., Hsieh, E., Baker, E. P. and Malik, H. S. (2016). Recurrent Gene Duplication Diversifies Genome Defense Repertoire in Drosophila. Mol Biol Evol 33(7): 1641-1653. PubMed ID: 26979388

    Levine, M. T. and Malik, H. S. (2013). A rapidly evolving genomic toolkit for Drosophila heterochromatin. Fly (Austin) 7(3): 137-141. PubMed ID: 23519206

  • Michael Levine Department of Molecular Biology, Princeton
    Levo, M., Raimundo, J., Bing, X. Y., Sisco, Z., Batut, P. J., Ryabichko, S., Gregor, T. and Levine, M. S. (2022). Transcriptional coupling of distant regulatory genes in living embryos. Nature 605(7911): 754-760. PubMed ID: 35508662

    Batut, P. J., Bing, X. Y., Sisco, Z., Raimundo, J., Levo, M. and Levine, M. S. (2022). Genome organization controls transcriptional dynamics during development. Science 375(6580): 566-570. PubMed ID: 35113722

    Ing-Simmons, E., Vaid, R., Bing, X. Y., Levine, M., Mannervik, M. and Vaquerizas, J. M. (2021). Independence of chromatin conformation and gene regulation during Drosophila dorsoventral patterning. Nat Genet 53(4): 487-499. PubMed ID: 33795866

    Cofer, E. M., Raimundo, J., Tadych, A., Yamazaki, Y., Wong, A. K., Theesfeld, C. L., Levine, M. S. and Troyanskaya, O. G. (2021). Modeling transcriptional regulation of model species with deep learning. Genome Res. PubMed ID: 33888512

    Heist, T., Fukaya, T. and Levine, M. (2019). Large distances separate coregulated genes in living Drosophila embryos. Proc Natl Acad Sci U S A 116(30): 15062-15067. PubMed ID: 31285341

    Lim, B., Fukaya, T., Heist, T. and Levine, M. (2018). Temporal dynamics of pair-rule stripes in living Drosophila embryos. Proc Natl Acad Sci U S A 115(33): 8376-8381. PubMed ID: 30061421

    Lim, B., Heist, T., Levine, M. and Fukaya, T. (2018). Visualization of transvection in living Drosophila embryos. Mol Cell 70(2): 287-296. PubMed ID: 29606591

    Fukaya, T., Lim, B. and Levine, M. (2017). Rapid rates of Pol II elongation in the Drosophila embryo. Curr Biol 27(9): 1387-1391. PubMed ID: 28457866

    Rogers, W. A., Goyal, Y., Yamaya, K., Shvartsman, S. Y. and Levine, M. S. (2017). Uncoupling neurogenic gene networks in the Drosophila embryo. Genes Dev 31(7): 634-638. PubMed ID: 28428262

    Fukaya, T., Lim, B. and Levine, M. (2017). Rapid Rates of Pol II Elongation in the Drosophila Embryo. Curr Biol 27: 1387-1391. PubMed ID: 28457866

    Lim, B., Levine, M. and Yamakazi, Y. (2017). Transcriptional pre-patterning of Drosophila gastrulation. Curr Biol 27(2): 286-290. PubMed ID: 28089518

  • Edwin Levitan University of Pittsburgh
    Klose, M. K., Bruchez, M. P., Deitcher, D. L. and Levitan, E. S. (2021). Temporally and spatially partitioned neuropeptide release from individual clock neurons. Proc Natl Acad Sci U S A 118(17). PubMed ID: 33875606

    Klose, M. K., Bruchez, M. P., Deitcher, D. L. and Levitan, E. S. (2021). Temporally and spatially partitioned neuropeptide release from individual clock neurons. Proc Natl Acad Sci U S A 118(17). PubMed ID: 33875606

    Hsu, I. U., Linsley, J. W., Zhang, X., Varineau, J. E., Berkhoudt, D. A., Reid, L. E., Lum, M. C., Orzel, A. M., Leflein, A., Xu, H., Collins, C. A., Hume, R. I., Levitan, E. S. and Kuwada, J. Y. (2020). Stac protein regulates release of neuropeptides. Proc Natl Acad Sci U S A. PubMed ID: 33168737

    Bulgari, D., Deitcher, D. L., Schmidt, B. F., Carpenter, M. A., Szent-Gyorgyi, C., Bruchez, M. P. and Levitan, E. S. (2019). Activity-evoked and spontaneous opening of synaptic fusion pores. Proc Natl Acad Sci U S A 116(34): 17039-17044. PubMed ID: 31383765

    Tao, J., Bulgari, D., Berkhoudt, D. A., Calderon, M. J., Watkins, S. C., Fonseca, H., Sabeva, N., Deitcher, D. L. and Levitan, E. S. (2019). Ptp4E regulates vesicular packaging for monoamine-neuropeptide co-transmission. J Cell Sci. PubMed ID: 30837287

    Bulgari, D., Jha, A., Deitcher, D. L. and Levitan, E. S. (2018). Myopic (HD-PTP, PTPN23) selectively regulates synaptic neuropeptide release. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 29378961

    Tao, J., Bulgari, D., Deitcher, D. L. and Levitan, E. S. (2017). Limited distal organelles and synaptic function in extensive monoaminergic innervation. J Cell Sci. PubMed ID: 28600320

    Bulgari, D., Deitcher, D. L. and Levitan, E. S. (2017). Loss of Huntingtin stimulates capture of retrograde dense-core vesicles to increase synaptic neuropeptide stores. Eur J Cell Biol [Epub ahead of print]. PubMed ID: 28129919

    Cavolo, S. L., Bulgari, D., Deitcher, D. L. and Levitan, E. S. (2016). Activity induces Fmr1-sensitive synaptic capture of anterograde circulating neuropeptide vesicles. J Neurosci 36(46): 11781-11787. PubMed ID: 27852784

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

  • Irwin Levitan Department of Neuroscience, Thomas Jefferson University, Philadelphia
    Shahidullah, M., Le Marchand, S. J., Fei, H., Zhang, J., Pandey, U. B., Dalva, M. B., Pasinelli, P. and Levitan, I. B. (2013). Defects in Synapse Structure and Function Precede Motor Neuron Degeneration in Drosophila Models of FUS-Related ALS. J Neurosci 33: 19590-19598. PubMed ID: 24336723

    Jepson, J., Sheldon, A., Shahidullah, M., Fei, H., Koh, K. and Levitan, I. B. (2013). Cell-specific fine-tuning of neuronal excitability by differential expression of modulator protein isoforms. J Neurosci 33: 16767-16777. PubMed ID: 24133277

    Sheldon, A. L., Zhang, J., Fei, H. and Levitan, I. B. (2011). SLOB, a SLOWPOKE channel binding protein, regulates insulin pathway signaling and metabolism in Drosophila. PLoS One 6: e23343. PubMed ID: 21850269

  • Lindsay Lewellyn Department of Biology, Butler University, Indianapolis, Indiana
    Stark, K., Crowe, O. and Lewellyn, L. (2021). Precise levels of the Drosophila adaptor protein Dreadlocks maintain the size and stability of germline ring canals. J Cell Sci 134(8). PubMed ID: 33912915

    Thestrup, J., Tipold, M., Kindred, A., Stark, K., Curry, T. and Lewellyn, L. (2020). The Arp2/3 complex and the formin, Diaphanous, are both required to regulate the size of germline ring canals in the developing egg chamber. Dev Biol. PubMed ID: 31945342

    Kline, A., Curry, T. and Lewellyn, L. (2018). The Misshapen kinase regulates the size and stability of the germline ring canals in the Drosophila egg chamber. Dev Biol 440(2): 99-112. PubMed ID: 29753016

    Cheerambathur, D. K., Prevo, B., Hattersley, N., Lewellyn, L., Corbett, K. D., Oegema, K. and Desai, A. (2017). Dephosphorylation of the Ndc80 Tail Stabilizes Kinetochore-Microtubule Attachments via the Ska Complex. Dev Cell 41(4): 424-437 e424. PubMed ID: 28535376

    Cetera, M., Lewellyn, L. and Horne-Badovinac, S. (2016). Cultivation and Live Imaging of Drosophila Ovaries. Methods Mol Biol 1478: 215-226. PubMed ID: 27730584

    Fernandes, V. M., McCormack, K., Lewellyn, L. and Verheyen, E. M. (2014). Integrins regulate apical constriction via microtubule stabilization in the Drosophila eye disc epithelium. Cell Rep 9(6): 2043-2055. PubMed ID: 25533344

  • Mingfa Li School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai
    Xu, M., Xiang, Y., Liu, X., Bai, B., Chen, R., Liu, L. and Li, M. (2018). Long noncoding RNA SMRG regulates Drosophila macrochaetes by antagonizing scute through E(spl)mbeta. RNA Biol. PubMed ID: 30526271

    Goyal, Y., Levario, T. J., Mattingly, H. H., Holmes, S., Shvartsman, S. Y. and Lu, H. (2017). Parallel imaging of Drosophila embryos for quantitative analysis of genetic perturbations of the Ras pathway. Dis Model Mech. PubMed ID: 28495673

    He, J., Xuan, T., Xin, T., An, H., Wang, J., Zhao, G. and Li, M. (2014). Evidence for Chromatin-Remodeling Complex PBAP-Controlled Maintenance of the Drosophila Ovarian Germline Stem Cells. PLoS One 9: e103473. PubMed ID: 25068272

    Xin, T., Xuan, T., Tan, J., Li, M., Zhao, G. and Li, M. (2013). The Drosophila putative histone acetyltransferase Enok maintains female germline stem cells through regulating Bruno and the niche. Dev Biol. PubMed ID: 24120347

    Xuan, T., Xin, T., He, J., Tan, J., Gao, Y., Feng, S., He, L., Zhao, G., Li, M. (2013) dBre1/dSet1-dependent pathway for histone H3K4 trimethylation has essential roles in controlling the germline stem cell maintenance and germ cell differentiation in the Drosophila ovary. Dev Biol. PubMed ID: 23624310

    Li, Q., Feng, S., Yu, L., Zhao, G. and Li, M. (2011). Requirements of Lgl in cell differentiation and motility during Drosophila ovarian follicular epithelium morphogenesis. Fly (Austin) 5: 81-87. PubMed ID: 21245664

    Li, M., Belozerov, V. E. and Cai, H. N. (2010). Modulation of chromatin boundary activities by nucleosome-remodeling activities in Drosophila melanogaster. Mol Cell Biol 30: 1067-1076. PubMed ID: 19995906

  • Xin LIANG Life Sciences, Tsinghua University, China
    Liu, Z., Wu, M. H., Wang, Q. X., Lin, S. Z., Feng, X. Q., Li, B. and Liang, X. (2022). Drosophila mechanical nociceptors preferentially sense localized poking. Elife 11. PubMed ID: 36200757

    Zhai, J., Li, W., Liu, X., Wang, D., Zhang, D., Liu, Y., Liang, X. and Chen, Z. (2023). Tiny Drosophila intestinal stem cells, big power. Cell Biol Int 47(1): 3-14. PubMed ID: 36177490

    Zelhof, A. C., Mahato, S., Liang, X., Rylee, J., Bergh, E., Feder, L. E., Larsen, M. E., Britt, S. G. and Friedrich, M. (2020). The brachyceran de novo gene PIP82, a phosphorylation target of aPKC, is essential for proper formation and maintenance of the rhabdomeric photoreceptor apical domain in Drosophila. PLoS Genet 16(6): e1008890. PubMed ID: 32579558

    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 49(5): 731-747 e737. PubMed ID: 31006649

  • Faith Liebl Southern Illinois University, Edwardsville
    Latcheva, N. K., Delaney, T. L., Viveiros, J. M., Smith, R. A., Bernard, K. M., Harsin, B., Marenda, D. R. and Liebl, F. L. W. (2019). The CHD protein, Kismet, is important for the recycling of synaptic vesicles during endocytosis. Sci Rep 9(1): 19368. PubMed ID: 31852969

    Hussein, N. A., Delaney, T. L., Tounsel, B. L. and Liebl, F. L. (2016). The extracellular-regulated kinase effector Lk6 is required for Glutamate receptor localization at the Drosophila neuromuscular junction. J Exp Neurosci 10: 77-91. PubMed ID: 27199570

    Sturgeon, M., Davis, D., Albers, A., Beatty, D., Austin, R., Ferguson, M., Tounsel, B. and Liebl, F. L. (2015). The Notch ligand E3 ligase, Mind Bomb1, regulates glutamate receptor localization in Drosophila. Mol Cell Neurosci 70: 11-21. PubMed ID: 26596173

    Ghosh, R., Vegesna, S., Safi, R., Bao, H., Zhang, B., Marenda, D. R. and Liebl, F. L. (2014). Kismet positively regulates glutamate receptor localization and synaptic transmission at the Drosophila neuromuscular junction. PLoS One 9: e113494. PubMed ID: 25412171

    Liebl, F. L., McKeown, C., Yao, Y. and Hing, H. K. (2010). Mutations in Wnt2 alter presynaptic motor neuron morphology and presynaptic protein localization at the Drosophila neuromuscular junction. PLoS One 5: e12778. PubMed ID: 20856675

  • Petros Ligoxygakis Department of Biochemistry, University of Oxford
    Glittenberg, M., Kounatidis, I., Atilano, M. and Ligoxygakis, P. (2022). A genetic screen in Drosophila reveals the role of fucosylation in host susceptibility to Candida infection. Dis Model Mech. PubMed ID: 35142345

    Bahuguna, S., Atilano, M., Glittenberg, M., Lee, D., Arora, S., Wang, L., Zhou, J., Redhai, S., Boutros, M. and Ligoxygakis, P. (2022). Bacterial recognition by PGRP-SA and downstream signalling by Toll/DIF sustain commensal gut bacteria in Drosophila. PLoS Genet 18(1): e1009992. PubMed ID: 35007276

    Vaz, F., Kounatidis, I., Covas, G., Parton, R. M., Harkiolaki, M., Davis, I., Filipe, S. R. and Ligoxygakis, P. (2019). Accessibility to peptidoglycan is important for the recognition of gram-positive bacteria in Drosophila. Cell Rep 27(8): 2480-2492.e2486. PubMed ID: 31116990

    Wang, L., Sloan, M. A. and Ligoxygakis, P. (2019). Intestinal NF-kappaB and STAT signalling is important for uptake and clearance in a Drosophila-Herpetomonas interaction model. PLoS Genet 15(3): e1007931. PubMed ID: 30822306

    Atilano, M. L., Glittenberg, M., Monteiro, A., Copley, R. and Ligoxygakis, P. (2017). MicroRNAs that contribute to coordinating the immune response in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 28706002

    Kounatidis, I., Chtarbanova, S., Cao, Y., Hayne, M., Jayanth, D., Ganetzky, B. and Ligoxygakis, P. (2017). NF-κB Immunity in the Brain Determines Fly Lifespan in Healthy Aging and Age-Related Neurodegeneration. Cell Rep 19: 836-848. PubMed ID: 28445733

    Mistry, R., Kounatidis, I. and Ligoxygakis, P. (2017). Interaction Between Familial Transmission and a Constitutively Active Immune System Shapes Gut Microbiota in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 28413160

    Fernando, M. D., Kounatidis, I. and Ligoxygakis, P. (2014). Loss of Trabid, a New Negative Regulator of the Drosophila Immune-Deficiency Pathway at the Level of TAK1, Reduces Life Span. PLoS Genet 10: e1004117. PubMed ID: 24586180

    Wang, L., Kounatidis, I. and Ligoxygakis, P. (2014). Drosophila as a model to study the role of blood cells in inflammation, innate immunity and cancer. Front Cell Infect Microbiol 3: 113. PubMed ID: 24409421

    Ligoxygakis, P. (2013). Genetics of Immune Recognition and Response in Drosophila host defense. Adv Genet 83: 71-97. PubMed ID: 23890212

    Kounatidis, I. and Ligoxygakis, P. (2012). Drosophila as a model system to unravel the layers of innate immunity to infection. Open Biol 2: 120075. PubMed ID: 22724070

    Atilano, M. L., Yates, J., Glittenberg, M., Filipe, S. R. and Ligoxygakis, P. (2011). Wall teichoic acids of Staphylococcus aureus limit recognition by the drosophila peptidoglycan recognition protein-SA to promote pathogenicity. PLoS Pathog 7: e1002421. PubMed ID: 22144903

  • Kathryn S Lilley Cambridge Centre for Proteomics, Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, United Kingdom
    Fabre, B., Choteau, S. A., Duboe, C., Pichereaux, C., Montigny, A., Korona, D., Deery, M. J., Camus, M., Brun, C., Burlet-Schiltz, O., Russell, S., Combier, J. P., Lilley, K. S. and Plaza, S. (2022). In Depth Exploration of the Alternative Proteome of Drosophila melanogaster. Front Cell Dev Biol 10: 901351. PubMed ID: 35721519

    Korona, D., Dirnberger, B., Giachello, C. N. G., Queiroz, R. M. L., Popovic, R., Muller, K. H., Minde, D. P., Deery, M. J., Johnson, G., Firth, L. C., Earley, F. G., Russell, S. and Lilley, K. S. (2022). Drosophila nicotinic acetylcholine receptor subunits and their native interactions with insecticidal peptide toxins. Elife 11. PubMed ID: 35575460

    Fabre, B., Korona, D., Lees, J. G., Lazar, I., Livneh, I., Brunet, M., Orengo, C. A., Russell, S. and Lilley, K. S. (2019). Comparison of Drosophila melanogaster Embryo and Adult Proteome by SWATH-MS Reveals Differential Regulation of Protein Synthesis, Degradation Machinery, and Metabolism Modules. J Proteome Res 18(6): 2525-2534. PubMed ID: 31083952

  • Mary Lilly NICHD, NIH, Bethesda
    Yang, S., Zhang, Y., Ting, C. Y., Bettedi, L., Kim, K., Ghaniam, E. and Lilly, M. A. (2020). The Rag GTPase Regulates the Dynamic Behavior of TSC Downstream of Both Amino Acid and Growth Factor Restriction. Dev Cell. PubMed ID: 32898476

    Xi, J., Cai, J., Cheng, Y., Fu, Y., Wei, W., Zhang, Z., Zhuang, Z., Hao, Y., Lilly, M. A. and Wei, Y. (2019). The TORC1 inhibitor Nprl2 protects age-related digestive function in Drosophila. Aging (Albany NY) 11. PubMed ID: 31712450

    Wei, Y., Bettedi, L., Ting, C. Y., Kim, K., Zhang, Y., Cai, J. and Lilly, M. A. (2019). The GATOR complex regulates an essential response to meiotic double-stranded breaks in Drosophila. Elife 8. PubMed ID: 31650955

    Rotelli, M. D., Policastro, R. A., Bolling, A. M., Killion, A. W., Weinberg, A. J., Dixon, M. J., Zentner, G. E., Walczak, C. E., Lilly, M. A. and Calvi, B. R. (2019). A Cyclin A-Myb-MuvB-Aurora B network regulates the choice between mitotic cycles and polyploid endoreplication cycles. PLoS Genet 15(7): e1008253. PubMed ID: 31291240

    Wei, Y., Reveal, B., Cai, W. and Lilly, M.A. (2016). The GATOR1 complex regulates metabolic homeostasis and the response to nutrient stress in Drosophila melanogaster. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27672113

    Cai, W., Wei, Y., Jarnik, M., Reich, J. and Lilly, M. A. (2016). The GATOR2 component Wdr24 regulates TORC1 activity and lysosome function. PLoS Genet 12: e1006036. PubMed ID: 27166823

    Wei, Y., Reveal, B., Reich, J., Laursen, W. J., Senger, S., Akbar, T., Iida-Jones, T., Cai, W., Jarnik, M. and Lilly, M. A. (2014). TORC1 regulators Iml1/GATOR1 and GATOR2 control meiotic entry and oocyte development in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 25512509

    Wei, Y. and Lilly, M. A. (2014). The TORC1 inhibitors Nprl2 and Nprl3 mediate an adaptive response to amino-acid starvation in Drosophila. Cell Death Differ. PubMed ID: 24786828

    Senger, S., Csokmay, J., Akbar, T., Jones, T. I., Sengupta, P. and Lilly, M. A. (2011). The nucleoporin Seh1 forms a complex with Mio and serves an essential tissue-specific function in Drosophila oogenesis. Development 138: 2133-2142. PubMed ID: 21521741

  • Bomyi Lim Dept. of Cell & Developmental Biology, University of Pennsylvania, Philadelphia
    Deng, H., Jin, G. and Lim, B. (2022). Unveiling dynamic enhancer-promoter interactions in Drosophila melanogaster. Biochem Soc Trans. PubMed ID: 36350004

    Keller, S. H., Jena, S. G., Yamazaki, Y. and Lim, B. (2020). Regulation of spatiotemporal limits of developmental gene expression via enhancer grammar. Proc Natl Acad Sci U S A 117(26): 15096-15103. PubMed ID: 32541043

    Lim, B., Fukaya, T., Heist, T. and Levine, M. (2018). Temporal dynamics of pair-rule stripes in living Drosophila embryos. Proc Natl Acad Sci U S A 115(33): 8376-8381. PubMed ID: 30061421

    Lim, B., Heist, T., Levine, M. and Fukaya, T. (2018). Visualization of Transvection in Living Drosophila Embryos. Mol Cell 70(2): 287-296 e286. PubMed ID: 29606591

  • Chunghun Lim School of Life Sciences, UNIST, Ulsan, Republic of Korea
    Lee, H. and Lim, C. (2022). Circadian gating of light-induced arousal in Drosophila sleep. J Neurogenet: 1-11. PubMed ID: 36457164

    Jeong, J., Lee, J., Kim, J. H. and Lim, C. (2021). Metabolic flux from the Krebs cycle to glutamate transmission tunes a neural brake on seizure onset. PLoS Genet 17(10): e1009871. PubMed ID: 34714823

    Kim, J. H., Ki, Y., Lee, H., Hur, M. S., Baik, B., Hur, J. H., Nam, D. and Lim, C. (2020). The voltage-gated potassium channel Shaker promotes sleep via thermosensitive GABA transmission. Commun Biol 3(1): 174. PubMed ID: 32296133

    Ki, Y. and Lim, C. (2019). Sleep-promoting effects of threonine link amino acid metabolism in Drosophila neuron to GABAergic control of sleep drive. Elife 8. PubMed ID: 31313987

    Lee, J., Yoo, E., Lee, H., Park, K., Hur, J. H. and Lim, C. (2017). LSM12 and ME31B/DDX6 Define Distinct Modes of Posttranscriptional Regulation by ATAXIN-2 Protein Complex in Drosophila Circadian Pacemaker Neurons. Mol Cell 66(1): 129-140 e127. PubMed ID: 28388438

    Kim, M., Lee, H., Hur, J. H., Choe, J. and Lim, C. (2016). CRTC Potentiates Light-independent timeless Transcription to Sustain Circadian Rhythms in Drosophila. Sci Rep 6: 32113. PubMed ID: 27577611

    Lim, C., Gandhi, S., Biniossek, M. L., Feng, L., Schilling, O., Urban, S. and Chen, X. (2015). An Aminopeptidase in the Drosophila Testicular Niche Acts in Germline Stem Cell Maintenance and Spermatogonial Dedifferentiation. Cell Rep 13(2): 315-325. PubMed ID: 26440886

  • Kah-Leong Lim Neurodegeneration Research Laboratory, Singapore
    Hang, L., Wang, Z., Foo, A. S. C., Goh, G. W. Y., Choong, H. C., Thundyil, J., Xu, S., Lam, K. P. and Lim, K. L. (2021). Conditional disruption of AMP kinase in dopaminergic neurons promotes Parkinson's disease-associated phenotypes in vivo. Neurobiol Dis 161: 105560. PubMed ID: 34767944

    Sim, J. P. L., Ziyin, W., Basil, A. H., Lin, S., Chen, Z., Zhang, C., Zeng, L., Cai, Y. and Lim, K. L. (2019). Identification of PP2A and S6 kinase as modifiers of Leucine-rich repeat kinase-induced neurotoxicity. Neuromolecular Med. PubMed ID: 31664682

    Ng, C. H., Basil, A. H., Hang, L., Tan, R., Goh, K. L., O'Neill, S., Zhang, X., Yu, F. and Lim, K. L. (2017). Genetic or pharmacological activation of the Drosophila PGC-1alpha ortholog spargel rescues the disease phenotypes of genetic models of Parkinson's disease. Neurobiol Aging 55: 33-37. PubMed ID: 28407521

    Gallart-Palau, X., Ng, C. H., Ribera, J., Sze, S. K. and Lim, K. L. (2016). Drosophila expressing human SOD1 successfully recapitulates mitochondrial phenotypic features of familial amyotrophic lateral sclerosis. Neurosci Lett 624: 47-52. PubMed ID: 27163198

    Zhang, H., Wang, Y., Wong, J. J., Lim, K. L., Liou, Y. C., Wang, H. and Yu, F. (2014). Endocytic pathways downregulate the L1-type cell adhesion molecule neuroglian to promote dendrite pruning in Drosophila. Dev Cell 30(4): 463-478. PubMed ID: 25158855

  • Andrew Lin Department of Biomedical Science, University of Sheffield, U. K.,
    Abdelrahman, N. Y., Vasilaki, E. and Lin, A. C. (2021). Compensatory variability in network parameters enhances memory performance in the Drosophila mushroom body. Proc Natl Acad Sci U S A 118(49). PubMed ID: 34845010

    Amin, H., Apostolopoulou, A. A., Suarez-Grimalt, R., Vrontou, E. and Lin, A. C. (2020). Localized inhibition in the Drosophila mushroom body. Elife 9. PubMed ID: 32955437

    Apostolopoulou, A. A. and Lin, A. C. (2020). Mechanisms underlying homeostatic plasticity in the Drosophila mushroom body in vivo. Proc Natl Acad Sci U S A 117(28): 16606-16615. PubMed ID: 32601210

    Mittal, A. M., Gupta, D., Singh, A., Lin, A. C. and Gupta, N. (2020). Multiple network properties overcome random connectivity to enable stereotypic sensory responses. Nat Commun 11(1): 1023. PubMed ID: 32094345

    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

  • Haifan Lin Biological and Biomedical Sciences, Yale University
    Gonzalez, L. E., Tang, X. and Lin, H. (2021). Maternal Piwi Regulates Primordial Germ Cell Development to Ensure the Fertility of Female Progeny in Drosophila. Genetics. PubMed ID: 34142134

    Liu, N., Neuenkirchen, N., Zhong, M. and Lin, H. (2021). Genome-wide mapping of Piwi association with specific loci in Drosophila ovaries. G3 (Bethesda) 11(2). PubMed ID: 33609367

    Park, A. R., Liu, N., Neuenkirchen, N., Guo, Q. and Lin, H. (2018). The Role of Maternal HP1a in early Drosophila embryogenesis via regulation of maternal transcript production. Genetics. PubMed ID: 30442760

    Ku, H. Y., Gangaraju, V. K., Qi, H., Liu, N. and Lin, H. (2016). Tudor-SN interacts with Piwi antagonistically in regulating spermatogenesis but synergistically in silencing transposons in Drosophila. PLoS Genet 12: e1005813. PubMed ID: 26808625

    Peng, J.C., Valouev, A., Liu, N. and Lin, H. (2016). Piwi maintains germline stem cells and oogenesis in Drosophila through negative regulation of Polycomb group proteins. Nat Genet [Epub ahead of print]. PubMed ID: 26780607

    Gonzalez, J., Qi, H., Liu, N. and Lin, H. (2015). Piwi is a key regulator of both somatic and germline stem cells in the Drosophila testis. Cell Rep 12: 150-161. PubMed ID: 26119740

    Ross, R. J., Weiner, M. M. and Lin, H. (2014). PIWI proteins and PIWI-interacting RNAs in the soma. Nature 505: 353-359. PubMed ID: 24429634

    Mani, S. R., Megosh, H. and Lin, H. (2013). PIWI proteins are essential for early Drosophila embryogenesis. Dev Biol. PubMed ID: 24184635

    Yin, H. and Lin, H. (2014). Chromatin immunoprecipitation assay of piwi in Drosophila. Methods Mol Biol 1093: 1-11. PubMed ID: 24178552

    Peng, J. C., Lin, H. (2013) Beyond transposons: the epigenetic and somatic functions of the Piwi-piRNA mechanism. Curr Opin Cell Biol 25: 190-194. PubMed ID: 23465540

  • Suewei Lin Institute of Molecular Biology, Academia Sinica, Nankang, Taipei
    Senapati, B., Tsao, C. H., Juan, Y. A., Chiu, T. H., Wu, C. L., Waddell, S. and Lin, S. (2019). A neural mechanism for deprivation state-specific expression of relevant memories in Drosophila. Nat Neurosci. PubMed ID: 31659341

    Tsao, C. H., Chen, C. C., Lin, C. H., Yang, H. Y. and Lin, S. (2018). Drosophila mushroom bodies integrate hunger and satiety signals to control innate food-seeking behavior. Elife 7. PubMed ID: 29547121

    Felsenberg, J., Barnstedt, O., Cognigni, P., Lin, S. and Waddell, S. (2017). Re-evaluation of learned information in Drosophila. Nature 544(7649): 240-244. PubMed ID: 28379939

    Das, G., Lin, S. and Waddell, S. (2016). Remembering Components of Food in Drosophila. Front Integr Neurosci 10: 4. PubMed ID: 26924969

    Lin, S., Owald, D., Chandra, V., Talbot, C., Huetteroth, W. and Waddell, S. (2014). Neural correlates of water reward in thirsty Drosophila. Nat Neurosci 17(11): 1536-1542. PubMed ID: 25262493

  • Xinhua Lin Department of Pediatrics, University of Cincinnati
    Qi, Y., Liu, H., Zhang, K., Wu, Y., Shen, C. and Lin, X. (2022). Ihog proteins contribute to integrin-mediated focal adhesions. Sci China Life Sci. PubMed ID: 36103028

    Ray, A. and Li, X. (2022). A Notch-dependent transcriptional mechanism controls expression of temporal patterning factors in Drosophila medulla. Elife 11. PubMed ID: 36040415

    Feng, Y., Li, Z., Lv, L., Du, A., Lin, Z., Ye, X., Lin, Y. and Lin, X. (2018). Tankyrase regulates apoptosis by activating JNK signaling in Drosophila. Biochem Biophys Res Commun. PubMed ID: 29953853

    Ling, X., Huang, Q., Xu, Y., Jin, Y., Feng, Y., Shi, W., Ye, X., Lin, Y., Hou, L. and Lin, X. (2017). The deubiquitinating enzyme Usp5 regulates Notch and RTK signaling during Drosophila eye development. FEBS Lett [Epub ahead of print]. PubMed ID: 28140449

    Li, X., Wu, Y., Shen, C., Belenkaya, T. Y., Ray, L. and Lin, X. (2015). Drosophila p24 and Sec22 regulate Wingless trafficking in the early secretory pathway. Biochem Biophys Res Commun [Epub ahead of print]. PubMed ID: 26002470

    Ren, W., Zhang, Y., Li, M., Wu, L., Wang, G., Baeg, G.H., You, J., Li, Z. and Lin, X. (2015). Windpipe controls Drosophila intestinal homeostasis by regulating JAK/STAT pathway via promoting receptor endocytosis and lysosomal degradation PLoS Genet 11: e1005180. PubMed ID: 25923769

    Huang, Q., Tang, X., Wang, G., Fan, Y., Ray, L., Bergmann, A., Belenkaya, T. Y., Ling, X., Yan, D., Lin, Y., Ye, X., Shi, W., Zhou, X., Lu, F., Qu, J. and Lin, X. (2014). Ubr3 E3 ligase regulates apoptosis by controlling the activity of DIAP1 in Drosophila. Cell Death Differ. PubMed ID: 25146930

    Huang, X., Shi, L., Cao, J., He, F., Li, R., Zhang, Y., Miao, S., Jin, L., Qu, J., Li, Z. and Lin, X. (2014). The sterile 20-like kinase tao controls tissue homeostasis by regulating the hippo pathway in Drosophila adult midgut. J Genet Genomics 41: 429-438. PubMed ID: 25160975

    Guo, Y., Feng, Y., Li, Z. and Lin, X. (2014). Drosophila heparan sulfate 3-O sulfotransferase B Null Mutant Is Viable and Exhibits No Defects in Notch Signaling. J Genet Genomics 41: 369-378. PubMed ID: 25064676

    Guo, Y., Li, Z. and Lin, X. (2014). Hs3st-A and Hs3st-B Regulate Intestinal Homeostasis in Drosophila Adult Midgut. Cell Signal. PubMed ID: 25049075

  • Jennifer Lippincott-Schwartz NICHD, NIH, Bethesda
    Sulkowski, M.J., Han, T.H., Ott, C., Wang, Q., Verheyen, E.M., Lippincott-Schwartz, J. and Serpe, M. (2016). A novel, noncanonical BMP pathway modulates synapse maturation at the Drosophila neuromuscular junction. PLoS Genet 12: e1005810. PubMed ID: 26815659

    Rikhy, R., Mavrakis, M. and Lippincott-Schwartz, J. (2015). Dynamin regulates metaphase furrow formation and plasma membrane compartmentalization in the syncytial Drosophila embryo. Biol Open. PubMed ID: 25661871

    Daniels, B. R., Rikhy, R., Renz, M., Dobrowsky, T. M. and Lippincott-Schwartz, J. (2012). Multiscale diffusion in the mitotic Drosophila melanogaster syncytial blastoderm. Proc Natl Acad Sci U S A 109: 8588-8593. PubMed ID: 22592793

    Mitra, K., Rikhy, R., Lilly, M. and Lippincott-Schwartz, J. (2012). DRP1-dependent mitochondrial fission initiates follicle cell differentiation during Drosophila oogenesis. J Cell Biol 197: 487-497. PubMed ID: 22584906

  • Edwin Levitan
    Bulgari, D., Deitcher, D. L. and Levitan, E. S. (2017). Loss of Huntingtin stimulates capture of retrograde dense-core vesicles to increase synaptic neuropeptide stores. Eur J Cell Biol [Epub ahead of print]. PubMed ID: 28129919

    Cavolo, S. L., Bulgari, D., Deitcher, D. L. and Levitan, E. S. (2016). Activity induces Fmr1-sensitive synaptic capture of anterograde circulating neuropeptide vesicles. J Neurosci 36(46): 11781-11787. PubMed ID: 27852784

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

    Cavolo, S. L., Zhou, C., Ketcham, S. A., Suzuki, M. M., Ukalovic, K., Silverman, M. A., Schroer, T. A. and Levitan, E. S. (2015). Mycalolide B dissociates dynactin and abolishes retrograde axonal transport of dense-core vesicles. Mol Biol Cell. PubMed ID: 26023088

    Wong, M. Y., Cavolo, S. L. and Levitan, E. S. (2015). Synaptic neuropeptide release by Dynamin-dependent partial release from circulating besicles. Mol Biol Cell [Epub ahead of print]. PubMed ID: 25904335

    James, R. E., Hoover, K. M., Bulgari, D., McLaughlin, C. N., Wilson, C. G., Wharton, K. A., Levitan, E. S. and Broihier, H. T. (2014). Crimpy enables discrimination of presynaptic and postsynaptic pools of a BMP at the Drosophila neuromuscular junction. Dev Cell 31: 586-598. PubMed ID: 25453556

    Li, L., Tian, X., Zhu, M., Bulgari, D., Bohme, M. A., Goettfert, F., Wichmann, C., Sigrist, S. J., Levitan, E. S. and Wu, C. (2014). Drosophila Syd-1, liprin-alpha, and protein phosphatase 2A B' subunit Wrd function in a linear pathway to prevent ectopic accumulation of synaptic materials in distal axons. J Neurosci 34: 8474-8487. PubMed ID:24948803

  • Howard Lipshitz University of Toronto
    Cao, W. X., Karaiskakis, A., Lin, S., Angers, S. and Lipshitz, H. D. (2021). The F-box protein Bard (CG14317) targets the Smaug RNA-binding protein for destruction during the Drosophila maternal-to-zygotic transition. Genetic. s. PubMed ID: 34757425

    Mohr, S., Kenny, A., Lam, S. T. Y., Morgan, M. B., Smibert, C. A., Lipshitz, H. D. and Macdonald, P. M. (2021). Opposing roles for Egalitarian and Staufen in transport, anchoring and localization of oskar mRNA in the Drosophila oocyte. PLoS Genet 17(4): e1009500. PubMed ID: 33798193

    Cao, W. X., Kabelitz, S., Gupta, M., Yeung, E., Lin, S., Rammelt, C., Ihling, C., Pekovic, F., Low, T. C. H., Siddiqui, N. U., Cheng, M. H. K., Angers, S., Smibert, C. A., Wuhr, M., Wahle, E. and Lipshitz, H. D. (2020). Precise Temporal Regulation of Post-transcriptional Repressors Is Required for an Orderly Drosophila Maternal-to-Zygotic Transition. Cell Rep 31(12): 107783. PubMed ID: 32579915

    Laver, J. D., Ly, J., Winn, A. K., Karaiskakis, A., Lin, S., Nie, K., Benic, G., Jaberi-Lashkari, N., Cao, W. X., Khademi, A., Westwood, J. T., Sidhu, S. S., Morris, Q., Angers, S., Smibert, C. A. and Lipshitz, H. D. (2020). The RNA-Binding Protein Rasputin/G3BP Enhances the Stability and Translation of Its Target mRNAs. Cell Rep 30(10): 3353-3367.e3357. PubMed ID: 32160542

    Wang, M., Ly, M., Lugowski, A., Laver, J. D., Lipshitz, H. D., Smibert, C. A. and Rissland, O. S. (2017). ME31B globally represses maternal mRNAs by two distinct mechanisms during the Drosophila maternal-to-zygotic transition. Elife 6. PubMed ID: 28875934

    Luo, H., Li, X., Claycomb, J.M. and Lipshitz, H.D. (2016). The Smaug RNA-binding protein is essential for microRNA synthesis during the Drosophila maternal-to-zygotic transition. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27591754

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

    Na, H., Laver, J. D., Jeon, J., Singh, F., Ancevicius, K., Fan, Y., Cao, W. X., Nie, K., Yang, Z., Luo, H., Wang, M., Rissland, O., Westwood, J. T., Kim, P. M., Smibert, C. A., Lipshitz, H. D. and Sidhu, S. S. (2016). A high-throughput pipeline for the production of synthetic antibodies for analysis of ribonucleoprotein complexes. RNA. PubMed ID: 26847261

    Laver, J. D., Marsolais, A. J., Smibert, C. A. and Lipshitz, H. D. (2015). Regulation and Function of Maternal Gene Products During the Maternal-to-Zygotic Transition in Drosophila. Curr Top Dev Biol 113: 43-84. PubMed ID: 26358870

    Laver, J. D., Li, X., Ray, D., Cook, K. B., Hahn, N. A., Nabeel-Shah, S., Kekis, M., Luo, H., Marsolais, A. J., Fung, K. Y., Hughes, T. R., Westwood, J. T., Sidhu, S. S., Morris, Q., Lipshitz, H. D. and Smibert, C. A. (2015). Brain tumor is a sequence-specific RNA-binding protein that directs maternal mRNA clearance during the Drosophila maternal-to-zygotic transition. Genome Biol 16: 94. PubMed ID: 25962635

    Ming, L., Wilk, R., Reed, B. H. and Lipshitz, H. D. (2014). Drosophila Hindsight and mammalian RREB-1 are evolutionarily conserved DNA-binding transcriptional attenuators. Differentiation. PubMed ID: 24418439

  • Joe Lipsick Stanford School of Medicine
    Vorster, P. J., Goetsch, P., Wijeratne, T. U., Guiley, K. Z., Andrejka, L., Tripathi, S., Larson, B. J., Rubin, S. M., Strome, S. and Lipsick, J. S. (2020). A long lost key opens an ancient lock: Drosophila Myb causes a synthetic multivulval phenotype in nematodes. Biol Open. PubMed ID: 32295830

    Cheng, M. H., Andrejka, L., Vorster, P. J., Hinman, A. and Lipsick, J. S. (2017). The Drosophila LIN54 homolog Mip120 controls two aspects of oogenesis. Biol Open [Epub ahead of print]. PubMed ID: 28522430

    Grigorian, M., DeBruhl, H. and Lipsick, J.S. (2017). The role of variant histone H2AV in D. melanogaster larval hematopoiesis. Development [Epub ahead of print]. PubMed ID: 28242611

    Debruhl, H., Wen, H., Lipsick, J. S. (2013) The Drosophila Myb and RB/E2F2 complex regulates cytokinesis in a histone H2Av-dependent manner. Mol Cell Biol. PubMed ID: 23438598

    Lewis, P. W., Sahoo, D., Geng, C., Bell, M., Lipsick, J. S. and Botchan, M. R. (2012). Drosophila lin-52 acts in opposition to repressive components of the Myb-MuvB/dREAM complex. Mol Cell Biol 32: 3218-3227. PubMed ID: 22688510

    Sim, C. K., Perry, S., Tharadra, S. K., Lipsick, J. S. and Ray, A. (2012). Epigenetic regulation of olfactory receptor gene expression by the Myb-MuvB/dREAM complex. Genes Dev 26: 2483-2498. PubMed ID: 23105004

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

    Judd, J., Duarte, F. M. and Lis, J. T. (2021). Pioneer-like factor GAF cooperates with PBAP (SWI/SNF) and NURF (ISWI) to regulate transcription. Genes Dev 35(1-2): 147-156. PubMed ID: 33303640

    Ray, J., Munn, P. R., Vihervaara, A., Lewis, J. J., Ozer, A., Danko, C. G. and Lis, J. T. (2019). Chromatin conformation remains stable upon extensive transcriptional changes driven by heat shock. Proc Natl Acad Sci U S A. PubMed ID: 31506350

    Boija, A., Mahat, D. B., Zare, A., Holmqvist, P. H., Philip, P., Meyers, D. J., Cole, P. A., Lis, J. T., Stenberg, P. and Mannervik, M. (2017). CBP regulates recruitment and release of promoter-proximal RNA polymerase II. Mol Cell 68(3): 491-503.e495. PubMed ID: 29056321

    Duarte, F. M., Fuda, N. J., Mahat, D. B., Core, L. J., Guertin, M. J. and Lis, J. T. (2016). Transcription factors GAF and HSF act at distinct regulatory steps to modulate stress-induced gene activation. Genes Dev 30: 1731-1746. PubMed ID: 27492368

    Jonkers, I. and Lis, J. T. (2015). Getting up to speed with transcription elongation by RNA polymerase II. Nat Rev Mol Cell Biol 16: 167-177. PubMed ID: 25693130

    Marr, S. K., Lis, J. T., Treisman, J. E. and Marr, M. T. (2014). The Metazoan-Specific Mediator Subunit 26 (Med26) Is Essential For Viability And Is Found At Both Active Genes And Pericentric Heterochromatin In Drosophila. Mol Cell Biol. PubMed ID: 24820420

    Salamanca, H. H., Antonyak, M. A., Cerione, R. A., Shi, H. and Lis, J. T. (2014). Inhibiting heat shock factor 1 in human cancer cells with a potent RNA aptamer. PLoS One 9: e96330. PubMed ID: 24800749

    Kwak, H. and Lis, J. T. (2013). Control of transcriptional elongation. Annu Rev Genet 47: 483-508. PubMed ID: 24050178

    Buckley, M. S., Kwak, H., Zipfel, W. R. and Lis, J. T. (2014). Kinetics of promoter Pol II on Hsp70 reveal stable pausing and key insights into its regulation. Genes Dev 28: 14-19. PubMed ID: 24395245

    Pagano, J. M., Kwak, H., Waters, C. T., Sprouse, R. O., White, B. S., Ozer, A., Szeto, K., Shalloway, D., Craighead, H. G. and Lis, J. T. (2014). Defining NELF-E RNA Binding in HIV-1 and Promoter-Proximal Pause Regions. PLoS Genet 10: e1004090. PubMed ID: 24453987

  • Troy Littleton MIT Department of Biology Sauvola, C. W., Akbergenova, Y., Cunningham, K. L., Aponte-Santiago, N. A. and Littleton, J. T. (2021). The decoy SNARE Tomosyn sets tonic versus phasic release properties and is required for homeostatic synaptic plasticity. Elife 10. PubMed ID: 34713802

    Ormerod, K. G., Scibelli, A. E. and Littleton, J. T. (2021). Regulation of excitation-contraction coupling at the Drosophila neuromuscular junction. J Physiol. PubMed ID: 34788476

    Sakurai, A., Littleton, J. T., Kojima, H. and Yoshihara, M. (2021). Alteration in information flow through a pair of feeding command neurons underlies a form of Pavlovian conditioning in the Drosophila brain. Curr Biol. PubMed ID: 34352215

    Weiss, S., Clamon, L. C., Manoim, J. E., Ormerod, K. G., Parnas, M. and Littleton, J. T. (2021). Glial ER and GAP junction mediated Ca(2+) waves are crucial to maintain normal brain excitability. Glia. PubMed ID: 34528727

    Fujii, T., Sakurai, A., Littleton, J. T. and Yoshihara, M. (2021). Synaptotagmin 7 switches short-term synaptic plasticity from depression to facilitation by suppressing synaptic transmission. Sci Rep 11(1): 4059. PubMed ID: 33603074

    Aponte-Santiago, N. A., Ormerod, K. G., Akbergenova, Y. and Littleton, J. T. (2020). Synaptic plasticity induced by differential manipulation of tonic and phasic motoneurons in Drosophila. J Neurosci. PubMed ID: 32631939

    Guan, Z., Quinones-Frias, M. C., Akbergenova, Y. and Littleton, J. T. (2020). Drosophila Synaptotagmin 7 negatively regulates synaptic vesicle release and replenishment in a dosage-dependent manner. Elife 9. PubMed ID: 32343229

    Weiss, S., Melom, J. E., Ormerod, K. G., Zhang, Y. V. and Littleton, J. T. (2019). Glial Ca(2+) signaling links endocytosis to K(+) buffering around neuronal somas to regulate excitability. Elife 8. PubMed ID: 31025939

    Harris, K. P., Littleton, J. T. and Stewart, B. A. (2018). Postsynaptic Syntaxin 4 negatively regulates the efficiency of neurotransmitter release. J Neurogenet: 1-9. PubMed ID: 30175640

    Akbergenova, Y., Cunningham, K. L., Zhang, Y. V., Weiss, S. and Littleton, J. T. (2018). Characterization of developmental and molecular factors underlying release heterogeneity at Drosophila synapses. Elife 7. PubMed ID: 29989549
  • Ji-Long Liu Shanghai Tech University
    Zhou, Y., Liu, J. and Liu, J. L. (2022). Connecting Ras and CTP synthase in Drosophila. Exp Cell Res 416(1): 113155. PubMed ID: 35427600

    Liu, J., Zhang, Y., Zhou, Y., Wang, Q. Q., Ding, K., Zhao, S., Lu, P. and Liu, J. L. (2022). Cytoophidia coupling adipose architecture and metabolism. Cell Mol Life Sci 79(10): 534. PubMed ID: 36180607

    Zhou, Y., Liu, J. and Liu, J. L. (2022). Connecting Ras and CTP synthase in Drosophila. Exp Cell Res: 113155. PubMed ID: 35427600

    Zhou, Y., Liu, J., Zhang, Y. and Liu, J. L. (2021). Drosophila intestinal homeostasis requires CTP synthase. Exp Cell Res 408(1): 112838. PubMed ID: 34560103

    Zhang, Y., Liu, J. and Liu, J. L. (2020). The atlas of cytoophidia in Drosophila larvae. J Genet Genomics. PubMed ID: 32912804

    Wu, Z. and Liu, J. L. (2019). Cytoophidia respond to nutrient stress in Drosophila. Exp Cell Res. PubMed ID: 30768932

    Ghosh, S., Tibbit, C. and Liu, J. L. (2016). Effective knockdown of Drosophila long non-coding RNAs by CRISPR interference. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 26850642

    Bassett, A. R., Kong, L. and Liu, J. L. (2015). A genome-wide CRISPR library for high-throughput genetic screening in Drosophila cells. J Genet Genomics 42: 301-309. PubMed ID: 26165496

    Tastan, O. Y. and Liu, J. L. (2015). CTP synthase is required for optic lobe homeostasis in Drosophila. J Genet Genomics 42: 261-274. PubMed ID: 26059773

    Bassett, A. and Liu, J. L. (2014). CRISPR/Cas9 mediated genome engineering in Drosophila. Methods. PubMed ID: 24576617

    Bassett, A. R. and Liu, J. L. (2014). CRISPR/Cas9 and Genome Editing in Drosophila. J Genet Genomics 41: 7-19. PubMed ID: 24480743

  • Li Liu State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing
    Wang, M., Hu, Q., Lv, T., Wang, Y., Lan, Q., Xiang, R., Tu, Z., Wei, Y., Han, K., Shi, C., Guo, J., Liu, C., Yang, T., Du, W., An, Y., Cheng, M., Xu, J., Lu, H., Li, W., Zhang, S., Chen, A., Chen, W., Li, Y., Wang, X., Xu, X., Hu, Y. and Liu, L. (2022). High-resolution 3D spatiotemporal transcriptomic maps of developing Drosophila embryos and larvae. Dev Cell 57(10): 1271-1283.e1274. PubMed ID: 35512700

    Lei, Y., Liu, K., Hou, L., Ding, L., Li, Y. and Liu, L. (2017). Small chaperons and autophagy protected neurons from necrotic cell death. Sci Rep 7(1): 5650. PubMed ID: 28720827

    Ding, L., Lei, Y., Han, Y., Li, Y., Ji, X. and Liu, L. (2016). Vimar is a novel regulator of mitochondrial fission through Miro. PLoS Genet 12: e1006359. PubMed ID: 27716788

    Hou, L., Liu, K., Li, Y., Ma, S., Ji, X. and Liu, L. (2016). Necrotic pyknosis is a morphologically and biochemically distinct event from apoptotic pyknosis. J Cell Sci. PubMed ID: 27358477

    Peng, Q., Wang, Y., Li, M., Yuan, D., Xu, M., Li, C., Gong, Z., Jiao, R. and Liu, L. (2016). cGMP-dependent protein kinase encoded by foraging regulates motor axon guidance in Drosophila by suppressing Lola function. J Neurosci 36: 4635-4646. PubMed ID: 27098704

    Zhang, Y., Cai, R., Zhou, R., Li, Y. and Liu, L. (2015). Tousled-like kinase mediated a new type of cell death pathway in Drosophila. Cell Death Differ [Epub ahead of print]. PubMed ID: 26088162

    Li, M. and Liu, L. (2014). Neural functions of long noncoding RNAs in Drosophila. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 25223318

    Liu, K., Ding, L., Li, Y., Yang, H., Zhao, C., Lei, Y., Han, S., Tao, W., Miao, D., Steller, H., Welsh, M. J. and Liu, L. (2014). Neuronal necrosis is regulated by a conserved chromatin-modifying cascade. Proc Natl Acad Sci U S A. PubMed ID: 25201987

    Sun, F., Wang, Y., Zhou, Y., Van Swinderen, B., Gong, Z. and Liu, L. (2014). Identification of neurons responsible for feeding behavior in the Drosophila brain. Sci China Life Sci. PubMed ID: 24744088

    Li, M., Xu, M., Wen, S., Bai, B., Chen, R. and Liu, L. (2014). One Novel Long Noncoding RNA lnc10 in Drosophila. J Genet Genomics 41: 79-82. PubMed ID: 24576460

    Cheng, X., Jiang, H., Li, W., Lv, H., Gong, Z., Liu, L. (2013) Ten-a Affects the Fusion of Central Complex Primordia in Drosophila. PLoS One 8: e57129. PubMed ID: 23437330

  • Nan Liu Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences
    Wu, X., Niu, K., Wang, X., Zhao, J., Wang, H., Li, D., Wang, H., Miao, T., Yang, Y., Ma, H., Zhang, Y., Pan, L., Liu, R., Bai, H. and Liu, N. (2022). microRNA-252 and FoxO repress inflammaging by a dual inhibitory mechanism on Dawdle-mediated TGF-β pathway in Drosophila. Genetics 220(3). PubMed ID: 35100390

    Ma, Z., Wang, H., Cai, Y., Wang, H., Niu, K., Wu, X., Ma, H., Yang, Y., Tong, W., Liu, F., Liu, Z., Zhang, Y., Liu, R., Zhu, Z. J. and Liu, N. (2018). Epigenetic drift of H3K27me3 in aging links glycolysis to healthy longevity in Drosophila. Elife 7. PubMed ID: 29809154

    Wang, H., Ma, Z., Niu, K., Xiao, Y., Wu, X., Pan, C., Zhao, Y., Wang, K., Zhang, Y. and Liu, N. (2015). Antagonistic roles between Nibbler and Hen1 modulate piRNA 3' ends in Drosophila. Development. PubMed ID: 26718004

    Gonzalez, J., Qi, H., Liu, N. and Lin, H. (2015). Piwi Is a Key Regulator of Both Somatic and Germline Stem Cells in the Drosophila Testis. Cell Rep 12: 150-161. PubMed ID: 26119740

    Liu, N. and Lasko, P. (2015). Analysis of RNA Interference Lines Identifies New Functions of Maternally-Expressed Genes Involved in Embryonic Patterning in Drosophila melanogaster. G3 (Bethesda) 5: 1025-1034. PubMed ID: 25834215

    Hernandez, G., Miron, M., Han, H., Liu, N., Magescas, J., Tettweiler, G., Frank, F., Siddiqui, N., Sonenberg, N. and Lasko, P. (2013). Mextli is a novel eukaryotic translation initiation factor 4E-binding protein that promotes translation in Drosophila melanogaster. Mol Cell Biol 33: 2854-2864. PubMed ID: 23716590

  • Marta Llimargas Institut de Biologia Molecular de Barcelona
    Klussmann-Fricke, B. J., Martin-Bermudo, M. D. and Llimargas, M. (2022). The basement membrane controls size and integrity of the Drosophila tracheal tubes. Cell Rep 39(4): 110734. PubMed ID: 35476979

    Casani, S., Casanova, J. and Llimargas, M. (2020). Unravelling the distinct contribution of cell shape changes and cell intercalation to tissue morphogenesis: the case of the Drosophila trachea. Open Biol 10(11): 200329. PubMed ID: 33234070

    Olivares-Castineira, I. and Llimargas, M. (2018). Anisotropic Crb accumulation, modulated by Src42A, is coupled to polarised epithelial tube growth in Drosophila. PLoS Genet 14(11): e1007824. PubMed ID: 30475799

    Olivares-Castineira, I. and Llimargas, M. (2017). EGFR controls Drosophila tracheal tube elongation by intracellular trafficking regulation. PLoS Genet 13(7): e1006882. PubMed ID: 28678789

    Moussian, B., Letizia, A., Martinez-Corrales, G., Rotstein, B., Casali, A. and Llimargas, M. (2015). Deciphering the genetic programme triggering timely and spatially-regulated chitin deposition. PLoS Genet 11: e1004939. PubMed ID: 25617778
    Okenve-Ramos, P. and Llimargas, M. (2014). Fascin links Btl/FGFR signalling to the actin cytoskeleton during Drosophila tracheal morphogenesis. Development 141: 929-939. PubMed ID: 24496629

    Letizia, A. and Llimargas, M. (2012). Adherens junctions and cadherins in Drosophila development. Subcell Biochem 60: 251-277. PubMed ID: 22674075

    Rotstein, B., Molnar, D., Adryan, B. and Llimargas, M. (2011). Tramtrack is genetically upstream of genes controlling tracheal tube size in Drosophila. PLoS One 6: e28985. PubMed ID: 22216153

  • Ana Llopart Department of Biology, University of Iowa, Iowa City
    Llopart, A. (2018). Faster-X evolution of gene expression is driven by recessive adaptive cis-regulatory variation in Drosophila. Mol Ecol. PubMed ID: 29717553

    Beck, E. A. and Llopart, A. (2015). Widespread Positive Selection Drives Differentiation of Centromeric Proteins in the Drosophila melanogaster subgroup. Sci Rep 5: 17197. PubMed ID: 26603658

    Beck, E. A., Thompson, A. C., Sharbrough, J., Brud, E. and Llopart, A. (2015). Gene flow between Drosophila yakuba and Drosophila santomea in subunit V of cytochrome c oxidase: A potential case of cytonuclear cointrogression. Evolution 69: 1973-1986. PubMed ID: 26155926

    Llopart, A. (2015). Parallel faster-X evolution of gene expression and protein sequences in Drosophila: beyond differences in expression properties and protein interactions. PLoS One 10: e0116829. PubMed ID: 25789611

  • Tom Lloyd Department of Neuroscience, Johns Hopkins University
    Sung, H. and Lloyd, T. E. (2022). Defective axonal transport of endo-lysosomes and dense core vesicles in a Drosophila model of C9-ALS/FTD. Traffic 23(9): 430-441. PubMed ID: 35908282

    Dubey, S. K., Maulding, K., Sung, H. and Lloyd, T. E. (2022). Nucleoporins are degraded via upregulation of ESCRT-III/Vps4 complex in Drosophila models of C9-ALS/FTD. Cell Rep 40(12): 111379. PubMed ID: 36130523

    Cunningham, K. M., Maulding, K., Ruan, K., Senturk, M., Grima, J. C., Sung, H., Zuo, Z., Song, H., Gao, J., Dubey, S., Rothstein, J. D., Zhang, K., Bellen, H. J. and Lloyd, T. E. (2020). TFEB/Mitf links impaired nuclear import to autophagolysosomal dysfunction in C9-ALS. Elife 9. PubMed ID: 33300868

    Machamer, J. B., Woolums, B. M., Fuller, G. G. and Lloyd, T. E. (2018). FUS causes synaptic hyperexcitability in Drosophila dendritic arborization neurons. Brain Res. PubMed ID: 29625118

    Bharadwaj, R., Cunningham, K. M., Zhang, K. and Lloyd, T. E. (2015). FIG4 regulates lysosome membrane homeostasis independent of phosphatase function. Hum Mol Genet. PubMed ID: 26662798

    Zhang, K., Donnelly, C.J., Haeusler, A.R., Grima, J.C., Machamer, J.B., Steinwald, P., Daley, E.L., Miller, S.J., Cunningham, K.M., Vidensky, S., Gupta, S., Thomas, M.A., Hong, I., Chiu, S.L., Huganir, R.L., Ostrow, L.W., Matunis, M.J., Wang, J., Sattler, R., Lloyd, T.E. and Rothstein, J.D. (2015). The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature [Epub ahead of print]. PubMed ID: 26308891

    Machamer, J. B., Collins, S. E. and Lloyd, T. E. (2014). The ALS gene FUS regulates synaptic transmission at the Drosophila neuromuscular junction. Hum Mol Genet. PubMed ID: 24569165

    Xu, D., et al. (2013). Top3beta is an RNA topoisomerase that works with fragile X syndrome protein to promote synapse formation. Nat Neurosci 16: 1238-1247. PubMed ID: 23912945

    Bharadwaj, R., Roy, M., Ohyama, T., Sivan-Loukianova, E., Delannoy, M., Lloyd, T. E., Zlatic, M., Eberl, D. F. and Kolodkin, A. L. (2013). Cbl-associated protein regulates assembly and function of two tension-sensing structures in Drosophila. Development 140: 627-638. PubMed ID: 23293294

    Lloyd, T. E. (2012). Axonal transport disruption in peripheral nerve disease: From Jack's discoveries as a resident to recent contributions. J Peripher Nerv Syst 17 Suppl 3: 46-51. PubMed ID: 23279432

  • Greg Lnenicka Department of Biological Sciences, University of Albany
    He, T., Nitabach, M. N. and Lnenicka, G. A. (2018). Parvalbumin expression affects synaptic development and physiology at the Drosophila larval NMJ. J Neurogenet: 1-12. PubMed ID: 30175644

    Gertner, D. M., Desai, S. and Lnenicka, G. A. (2014). Synaptic excitation is regulated by the postsynaptic dSK channel at the Drosophila larval NMJ. J Neurophysiol. PubMed ID: 24671529

    Desai, S. A. and Lnenicka, G. A. (2011). Characterization of postsynaptic Ca2+ signals at the Drosophila larval NMJ. J Neurophysiol 106: 710-721. PubMed ID: 21593388

    He, T. and Lnenicka, G. A. (2011). Ca(2)+ buffering at a drosophila larval synaptic terminal. Synapse 65: 687-693. PubMed ID: 21218450

  • John Locke Biological Sciences, University of Alberta, Edmonton
    McCracken, A. and Locke, J. (2016). Mutations in ash1 and trx enhance P-element-dependent silencing in Drosophila melanogaster. Genome [Epub ahead of print]. PubMed ID: 27373142

    McCracken, A. and Locke, J. (2014). Mutations in CG8878, a Novel Putative Protein Kinase, Enhance P Element Dependent Silencing (PDS) and Position Effect Variegation (PEV) in Drosophila melanogaster. PLoS One 9: e71695. PubMed ID: 24614804

    Sameny, A., La, A., Hanna, S. and Locke, J. (2011). Point mutations in a Drosophila P element abolish both P element-dependent silencing (PDS) of a transgene and repressor functions. Chromosoma 120: 573-585. PubMed ID: 22009629

    Sameny, A. and Locke, J. (2011). The P-element-induced silencing effect of KP transposons is dose dependent in Drosophila melanogaster. Genome 54: 752-762. PubMed ID: 21888571

  • David W. Loehlin Department of Biology, Thompson Biology Lab, Williamstown, MA
    Dean, D. M., Deitcher, D. L., Paster, C. O., Xu, M. and Loehlin, D. W. (2022). "A fly appeared": sable, a classic Drosophila mutation, maps to Yippee, a gene affecting body color, wings, and bristles. G3 (Bethesda). PubMed ID: 35266526

    Dean, D. M., Deitcher, D. L., Paster, C. O., Xu, M. and Loehlin, D. W. (2022). "A fly appeared": sable, a classic Drosophila mutation, maps to Yippee, a gene affecting body color, wings, and bristles. G3 (Bethesda) 12(5). PubMed ID: 35266526

    Loehlin, D. W., Kim, J. Y. and Paster, C. O. (2022). A tandem duplication in Drosophila melanogaster shows enhanced expression beyond the gene copy number. Genetics 220(3). PubMed ID: 35100388

    Loehlin, D. W., Ames, J. R., Vaccaro, K. and Carroll, S. B. (2019). A major role for noncoding regulatory mutations in the evolution of enzyme activity. Proc Natl Acad Sci U S A 116(25): 12383-12389. PubMed ID: 31152141

  • Volker Loeschcke Department of Bioscience, Aarhus University, Denmark
    Amstrup Bay, A., Bæk, I., Loeschcke, V. and Givskov Sørensena, J. (2022). A functional study of the role of Turandot genes in Drosophila melanogaster: An emerging candidate mechanism for inducible heat tolerance. J Insect Physiol: 104456. PubMed ID: 36396076

    Navas, C. A., Agudelo-Cantero, G. A. and Loeschcke, V. (2022). Thermal boldness: Volunteer exploration of extreme temperatures in fruit flies. J Insect Physiol 136: 104330. PubMed ID: 34848182

    Manenti, T., Kjærsgaard, A., Schou, T. M., Pertoldi, C., Moghadam, N. N. and Loeschcke, V. (2021). Responses to Developmental Temperature Fluctuation in Life History Traits of Five Drosophila Species (Diptera: Drosophilidae) from Different Thermal Niches. Insects 12(10). PubMed ID: 34680694

    Manenti, T., Juul Sten, L. and Loeschcke, V. (2021). Daily increasing or decreasing photoperiod affects stress resistance and life history traits in four Drosophila species. J Insect Physiol: 104251. PubMed ID: 33971199

    Sorensen, J. G., Giribets, M. P., Tarrio, R., Rodriguez-Trelles, F., Schou, M. F. and Loeschcke, V. (2019). Expression of thermal tolerance genes in two Drosophila species with different acclimation capacities. J Therm Biol 84: 200-207. PubMed ID: 31466754

    Davies, L. R., Schou, M. F., Kristensen, T. N. and Loeschcke, V. (2019). Fluctuations in nutrient composition affect male reproductive output in Drosophila melanogaster. J Insect Physiol: 103940. PubMed ID: 31493390

    Mengel-From, J., Svane, A. M., Pertoldi, C., Kristensen, T. N., Loeschcke, V., Skytthe, A., Christensen, K., Lindahl-Jacobsen, R., Hjelmborg, J. and Christiansen, L. (2019). Advanced parental age at conception and sex affects mitochondrial DNA copy number in human and fruit flies. J Gerontol A Biol Sci Med Sci. PubMed ID: 30874797

    Michalak, P., Kang, L., Schou, M. F., Garner, H. R. and Loeschcke, V. (2018). Genomic signatures of experimental adaptive radiation in Drosophila. Mol Ecol. PubMed ID: 30375065

    Manenti, T., Cunha, T. R., Sorensen, J. G. and Loeschcke, V. (2018). How much starvation, desiccation and oxygen depletion can Drosophila melanogaster tolerate before its upper thermal limits are affected? J Insect Physiol 111: 1-7. PubMed ID: 30273554

    Davies, L. R., Schou, M. F., Kristensen, T. N. and Loeschcke, V. (2018). Linking developmental diet to adult foraging choice in Drosophila melanogaster. J Exp Biol [Epub ahead of print]. PubMed ID: 29666197

    Sorensen, J. G., Schou, M. F. and Loeschcke, V. (2017). Evolutionary adaptation to environmental stressors: A common response at the proteomic level. Evolution [Epub ahead of print]. PubMed ID: 28369831

    Michalak, P., Kang, L., Sarup, P. M., Schou, M. F. and Loeschcke, V. (2017). Nucleotide diversity inflation as a genome-wide response to experimental lifespan extension in Drosophila melanogaster. BMC Genomics 18(1): 84. PubMed ID: 28088192

    Sarup, P., Petersen, S. M., Nielsen, N. C., Loeschcke, V. and Malmendal, A. (2016). Mild heat treatments induce long-term changes in metabolites associated with energy metabolism in Drosophila melanogaster. Biogerontology. PubMed ID: 27511372

  • Mary Logan Oregon Health & Science University, Jungers Center for Neurosciences Research, Portland
    Ray, A., Speese, S. D. and Logan, M. A. (2017). Glial Draper rescues Abeta toxicity in a Drosophila model of Alzheimer's Disease. J Neurosci. PubMed ID: 29109235

    Winfree, L. M., Speese, S. D. and Logan, M. A. (2017). Protein phosphatase 4 coordinates glial membrane recruitment and phagocytic clearance of degenerating axons in Drosophila. Cell Death Dis 8(2): e2623. PubMed ID: 28230857

    Purice, M. D., Speese, S. D. and Logan, M. A. (2016). Delayed glial clearance of degenerating axons in aged Drosophila is due to reduced PI3K/Draper activity. Nat Commun 7: 12871. PubMed ID: 27647497

    Doherty, J., Logan, M. A., Tasdemir, O. E. and Freeman, M. R. (2009). Ensheathing glia function as phagocytes in the adult Drosophila brain. J Neurosci 29(15): 4768-4781. PubMed ID: 19369546

  • Ingrid Lohmann Centre for Organismal Studies, Heidelberg University
    Velten, J., Gao, X., Van Nierop, Y. S. P., Domsch, K., Agarwal, R., Bognar, L., Paulsen, M., Velten, L. and Lohmann, I. (2022). Single-cell RNA sequencing of motoneurons identifies regulators of synaptic wiring in Drosophila embryos. Mol Syst Biol 18(3): e10255. PubMed ID: 35225419

    Carnesecchi, J., Boumpas, P., van Nierop, Y. S. P., Domsch, K., Pinto, H. D., Borges Pinto, P. and Lohmann, I. (2022). The Hox transcription factor Ultrabithorax binds RNA and regulates co-transcriptional splicing through an interplay with RNA polymerase II. Nucleic Acids Res 50(2): 763-783. PubMed ID: 34931250

    Domsch, K., Schroder, J., Janeschik, M., Schaub, C. and Lohmann, I. (2021). The Hox Transcription Factor Ubx Ensures Somatic Myogenesis by Suppressing the Mesodermal Master Regulator Twist. Cell Rep 34(1): 108577. PubMed ID: 33406430

    Sorge, S., Theelke, J., Yildirim, K., Hertenstein, H., McMullen, E., Muller, S., Altburger, C., Schirmeier, S. and Lohmann, I. (2020). ATF4-Induced Warburg Metabolism Drives Over-Proliferation in Drosophila. Cell Rep 31(7): 107659. PubMed ID: 32433968

    Carnesecchi, J., Sigismondo, G., Domsch, K., Baader, C. E. P., Rafiee, M. R., Krijgsveld, J. and Lohmann, I. (2020). Multi-level and lineage-specific interactomes of the Hox transcription factor Ubx contribute to its functional specificity. Nat Commun 11(1): 1388. PubMed ID: 32170121

    Domsch, K., Carnesecchi, J., Disela, V., Friedrich, J., Trost, N., Ermakova, O., Polychronidou, M. and Lohmann, I. (2019). The Hox transcription factor Ubx stabilizes lineage commitment by suppressing cellular plasticity in Drosophila. Elife 8. PubMed ID: 31050646

    Tamirisa, S., Papagiannouli, F., Rempel, E., Ermakova, O., Trost, N., Zhou, J., Mundorf, J., Brunel, S., Ruhland, N., Boutros, M., Lohmann, J. U. and Lohmann, I. (2018). Decoding the regulatory logic of the Drosophila male stem cell system. Cell Rep 24(11): 3072-3086. PubMed ID: 30208329

    Friedrich, J., Sorge, S., Bujupi, F., Eichenlaub, M.P., Schulz, N.G., Wittbrodt, J. and Lohmann, I. (2016). Hox function is required for the development and maintenance of the Drosophila feeding motor unit. Cell Rep [Epub ahead of print]. PubMed ID: 26776518

    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. and Lohmann, I. (2015). Stage-specific control of stem cell niche architecture in the Drosophila testis by the posterior Hox gene Abd-B. Comput Struct Biotechnol J 13: 122-130. PubMed ID: 25750700

    Polychronidou, M. and Lohmann, I. (2014). Hox transcriptomics in Drosophila embryos. Methods Mol Biol 1196: 197-208. PubMed ID: 25151165

  • Anthony Long Ecology & Evolutionary Biology, University of California, Irvine
    Macdonald, S. J., Cloud-Richardson, K. M., Sims-West, D. J. and Long, A. D. (2022). Powerful, efficient QTL mapping in Drosophila melanogaster using bulked phenotyping and pooled sequencing. Genetics 220(3). PubMed ID: 35100395

    Chakraborty, M., Emerson, J. J., Macdonald, S. J. and Long, A. D. (2019). Structural variants exhibit widespread allelic heterogeneity and shape variation in complex traits. Nat Commun 10(1): 4872. PubMed ID: 31653862

    King, E. G., Kislukhin, G., Walters, K. N. and Long, A. D. (2014). Using Drosophila melanogaster To Identify Chemotherapy Toxicity Genes. Genetics 198: 31-43. PubMed ID: 25236447

    Burke, M. K., King, E. G., Shahrestani, P., Rose, M. R. and Long, A. D. (2013). Genome-wide association study of extreme longevity in Drosophila melanogaster. Genome Biol Evol. PubMed ID: 24259311

    Kislukhin, G., King, E. G., Walters, K. N., Macdonald, S. J. and Long, A. D. (2013). The Genetic Architecture of Methotrexate Toxicity is Similar in Drosophila melanogaster and Humans. G3 (Bethesda). PubMed ID: 23733889

    King, E. G., Macdonald, S. J. and Long, A. D. (2012). Properties and power of the Drosophila Synthetic Population Resource for the routine dissection of complex traits. Genetics 191: 935-949. PubMed ID: 22505626

    King, E. G., Merkes, C. M., McNeil, C. L., Hoofer, S. R., Sen, S., Broman, K. W., Long, A. D. and Macdonald, S. J. (2012). Genetic dissection of a model complex trait using the Drosophila Synthetic Population Resource. Genome Res 22: 1558-1566. PubMed ID: 22496517

    Kislukhin, G., Murphy, M. L., Jafari, M. and Long, A. D. (2012). Chemotherapy-induced toxicity is highly heritable in Drosophila melanogaster. Pharmacogenet Genomics 22: 285-289. PubMed ID: 22336958

  • Manyuan Long Department of Ecology & Evolution, University of Chicago
    Krinsky, B. H., Arthur, R. K., Xia, S., Sosa, D., Arsala, D., White, K. P. and Long, M. (2021). Rapid Cis-Trans Coevolution Driven by a Novel Gene Retroposed from a Eukaryotic Conserved CCR4-NOT Component in Drosophila. Genes (Basel) 13(1). PubMed ID: 35052398

    Ni, X., Zhang, Y. E., Negre, N., Chen, S., Long, M. and White, K. P. (2012). Adaptive Evolution and the Birth of CTCF Binding Sites in the Drosophila Genome. PLoS Biol 10: e1001420. PubMed ID: 23139640

    Vibranovski, M. D., Zhang, Y. E., Kemkemer, C., Vankuren, N. W., Lopes, H. F., Karr, T. L. and Long, M. (2012). Segmental dataset and whole body expression data do not support the hypothesis that non-random movement is an intrinsic property of Drosophila retrogenes. BMC Evol Biol 12: 169. PubMed ID: 22950647

    Chen, S., Spletter, M., Ni, X., White, K. P., Luo, L. and Long, M. (2012). Frequent recent origination of brain genes shaped the evolution of foraging behavior in Drosophila. Cell Rep 1: 118-132. PubMed ID: 22832161

  • Ben Longdon University of Exeter
    Duxbury, E. M., Day, J. P., Maria Vespasiani, D., Thuringer, Y., Tolosana, I., Smith, S. C., Tagliaferri, L., Kamacioglu, A., Lindsley, I., Love, L., Unckless, R. L., Jiggins, F. M. and Longdon, B. (2019). Host-pathogen coevolution increases genetic variation in susceptibility to infection. Elife 8. PubMed ID: 31038124

    Longdon, B., Day, J. P., Alves, J. M., Smith, S. C. L., Houslay, T. M., McGonigle, J. E., Tagliaferri, L. and Jiggins, F. M. (2018). Host shifts result in parallel genetic changes when viruses evolve in closely related species. PLoS Pathog 14(4): e1006951. PubMed ID: 29649296

    Longdon, B., Day, J. P., Schulz, N., Leftwich, P. T., de Jong, M. A., Breuker, C. J., Gibbs, M., Obbard, D. J., Wilfert, L., Smith, S. C., McGonigle, J. E., Houslay, T. M., Wright, L. I., Livraghi, L., Evans, L. C., Friend, L. A., Chapman, T., Vontas, J., Kambouraki, N. and Jiggins, F. M. (2017). Vertically transmitted rhabdoviruses are found across three insect families and have dynamic interactions with their hosts. Proc Biol Sci 284(1847). PubMed ID: 28100819

    Webster, C. L., Longdon, B., Lewis, S. H. and Obbard, D. J. (2016). Twenty-Five New Viruses Associated with the Drosophilidae (Diptera). Evol Bioinform Online 12(Suppl 2): 13-25. PubMed ID: 27375356

  • Michelle Longworth Lerner Research Institute, Cleveland Clinic
    Deutschman, E., Ward, J. R., Ho, A. L. K. T., Alban, T. J., Zhang, D., Willard, B., Lemieux, M. E., Lathia, J. D. and Longworth, M. S. (2018). Comparing and Contrasting the Effects of Drosophila Condensin II Subunit dCAP-D3 Overexpression and Depletion in Vivo. Genetics. PubMed ID: 30068527

    Klebanow, L. R., Peshel, E. C., Schuster, A. T., De, K., Sarvepalli, K., Lemieux, M. E., Lenoir, J. J., Moore, A. W., McDonald, J. A. and Longworth, M. S. (2016). Drosophila Condensin II subunit Chromosome-associated protein D3 regulates cell fate determination through non-cell-autonomous signaling. Development 143(15): 2791-2802. PubMed ID: 27317808

    Schuster, A. T., Sarvepalli, K., Murphy, E. A. and Longworth, M. S. (2013). Condensin II subunit dCAP-D3 restricts retrotransposon mobilization in Drosophila somatic cells. PLoS Genet 9(10): e1003879. PubMed ID: 24204294

  • Benjamin Loppin Biométrie et Biologie Evolutive, CNRS, Lyon
    Torres-Campana, D., Kimura, S., Orsi, G. A., Horard, B., Benoit, G. and Loppin, B. (2020). The Lid/KDM5 histone demethylase complex activates a critical effector of the oocyte-to-zygote transition. PLoS Genet 16(3): e1008543. PubMed ID: 32134927

    Horard, B., Sapey-Triomphe, L., Bonnefoy, E. and Loppin, B. (2018). ASF1 is required to load histones on the HIRA complex in preparation of paternal chromatin assembly at fertilization. Epigenetics Chromatin 11(1): 19. PubMed ID: 29751847

    Tirmarche, S., Kimura, S., Dubruille, R., Horard, B. and Loppin, B. (2016). Unlocking sperm chromatin at fertilization requires a dedicated egg thioredoxin in Drosophila. Nat Commun 7: 13539. PubMed ID: 27876811

    Kimura, S. and Loppin, B. (2016). The Drosophila chromosomal protein Mst77F is processed to generate an essential component of mature sperm chromatin. Open Biol 6. PubMed ID: 27810970

    Helleu, Q., Gerard, P. R., Dubruille, R., Ogereau, D., Prud'homme, B., Loppin, B. and Montchamp-Moreau, C. (2016). Rapid evolution of a Y-chromosome heterochromatin protein underlies sex chromosome meiotic drive. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26979956

    Dubruille, R. and Loppin, B. (2015). Protection of Drosophila chromosome ends with minimal telomere capping. J Cell Sci [Epub ahead of print]. PubMed ID: 25908850

    Kimura, S. and Loppin, B. (2015). Two bromodomain proteins functionally interact to recapitulate an essential BRDT-like function in Drosophila spermatocytes. Open Biol 5. PubMed ID: 25652540

    Horard, B. and Loppin, B. (2015). Histone storage and deposition in the early Drosophila embryo. Chromosoma [Epub ahead of print]. PubMed ID: 25563491

    Tirmarche, S., Kimura, S., Sapey-Triomphe, L., Sullivan, W., Landmann, F. and Loppin, B. (2014). Drosophila protamine-Like Mst35Ba and Mst35Bb are required for proper sperm nuclear morphology but are dispensable for male fertility. G3 (Bethesda). PubMed ID: 25236732

    Delabaere, L., Orsi, G. A., Sapey-Triomphe, L., Horard, B., Couble, P. and Loppin, B. (2014). The Spartan Ortholog Maternal Haploid Is Required for Paternal Chromosome Integrity in the Drosophila Zygote. Curr Biol. PubMed ID: 25242033

  • Tristan Long Laurier University, Waterloo, Ontario
    Khodaei, L. and Long, T. A. F. (2019). Kin recognition and co-operative foraging in Drosophila melanogaster larvae. J Evol Biol. PubMed ID: 31454451

    Filice, D. C. S. and Long, T. A. F. (2018). Genetic trade-offs between male reproductive traits in Drosophila melanogaster. Biol Lett 14(10). PubMed ID: 30333262

    Filice, D. C. S. and Long, T. A. F. (2017). Phenotypic plasticity in female mate choice behavior is mediated by an interaction of direct and indirect genetic effects in Drosophila melanogaster. Ecol Evol 7(10): 3542-3551. PubMed ID: 28515889

    Tennant, H. M., Sonser, E. E. and Long, T. A. (2014). Hemiclonal analysis of interacting phenotypes in male and female Drosophila melanogaster. BMC Evol Biol 14: 95. PubMed ID: 24884361

    Tennant, H. M., Sonser, E. E. and Long, T. A. (2013). Variation in male effects on female fecundity in Drosophila melanogaster. J Evol Biol. PubMed ID: 24372989

    Long, T. A., Rowe, L. and Agrawal, A. F. (2013). The effects of selective history and environmental heterogeneity on inbreeding depression in experimental populations of Drosophila melanogaster. Am Nat 181: 532-544. PubMed ID: 23535617

    Long, T. A., Pischedda, A. and Rice, W. R. (2010). Remating in Drosophila melanogaster: are indirect benefits condition dependent? Evolution 64: 2767-2774. PubMed ID: 20394654

  • Susan E. Lott Department of Evolution and Ecology, College of Biological Sciences, Center for Population Biology. UC Davis, California
    Feitzinger, A. A., Le, A., Thompson, A., Haseeb, M., Murugesan, M. K., Tang, A. M. and Lott, S. E. (2022). Natural variation in the maternal and zygotic mRNA complements of the early embryo in Drosophila melanogaster. BMC Genomics 23(1): 641. PubMed ID: 36076188

    Cartwright, E. L. and Lott, S. E. (2020). Evolved Differences in cis and trans Regulation Between the Maternal and Zygotic mRNA Complements in the Drosophila Embryo. Genetics. PubMed ID: 32928902

    Omura, C. S. and Lott, S. E. (2020). The conserved regulatory basis of mRNA contributions to the early Drosophila embryo differs between the maternal and zygotic genomes. PLoS Genet 16(3): e1008645. PubMed ID: 32226006

    >Kalay, G., Atallah, J., Sierra, N. C., Tang, A. M., Crofton, A. E., Murugesan, M. K., Wykoff-Clary, S. and Lott, S. E. (2019). Evolution of larval segment position across 12 Drosophila species. Evolution. PubMed ID: 31886902

    Atallah, J. and Lott, S. E. (2018). Evolution of maternal and zygotic mRNA complements in the early Drosophila embryo. PLoS Genet 14(12): e1007838. PubMed ID: 30557299

  • Matthieu Louis Centre for Genomic Regulation, Barcelona
    Tadres, D. and Louis, M. (2020). PiVR: An affordable and versatile closed-loop platform to study unrestrained sensorimotor behavior. PLoS Biol 18(7): e3000712. PubMed ID: 32663220

    Kim, D., Alvarez, M., Lechuga, L. M. and Louis, M. (2017). Species-specific modulation of food-search behavior by respiration and chemosensation in Drosophila larvae. Elife 6. PubMed ID: 28871963

    Schulze, A., Gomez-Marin, A., Rajendran, V. G., Lott, G., Musy, M., Ahammad, P., Deogade, A., Sharpe, J., Riedl, J., Jarriault, D., Trautman, E. T., Werner, C., Venkadesan, M., Druckmann, S., Jayaraman, V. and Louis, M. (2015). Dynamical feature extraction at the sensory periphery guides chemotaxis. Elife 4. PubMed ID: 26077825

    Tastekin, I., Riedl, J., Schilling-Kurz, V., Gomez-Marin, A., Truman, J. W. and Louis, M. (2015). Role of the Subesophageal Zone in Sensorimotor Control of Orientation in Drosophila Larva. Curr Biol. PubMed ID: 25959970

    Schleyer, M., Reid, S.F., Pamir, E., Saumweber, T., Paisios, E., Davies, A., Gerber, B. and Louis, M. (2015). The impact of odor-reward memory on chemotaxis in larval Drosophila. Learn Mem 22: 267-277. PubMed ID: 25887280

    Gomez-Marin, A. and Louis, M. (2014). Multilevel control of run orientation in Drosophila larval chemotaxis. Front Behav Neurosci 8: 38. PubMed ID: 24592220

    Riedl, J. and Louis, M. (2012). Behavioral neuroscience: Crawling is a no-brainer for fruit fly larvae. Curr Biol 22: R867-869. PubMed ID:
    Gomez-Marin, A., Partoune, N., Stephens, G. J., Louis, M. and Brembs, B. (2012). Automated tracking of animal posture and movement during exploration and sensory orientation behaviors. PLoS One 7: e41642. PubMed ID:
    22912674

    Gomez-Marin, A. and Louis, M. (2012). Active sensation during orientation behavior in the Drosophila larva: more sense than luck. Curr Opin Neurobiol 22: 208-215. PubMed ID: 22169055

  • Marie Louvet-Vallee Developmental Biology Laboratory, Sorbonne Universite, Paris
    Lacoste, J., Soula, H., Burg, A., Audibert, A., Darnat, P., Gho, M. and Louvet-Vallee, S. (2022). A neural progenitor mitotic wave is required for asynchronous axon outgrowth and morphology. Elife 11. PubMed ID: 35254258

    Lacoste, J., Soula, H., Burg, A., Audibert, A., Darnat, P., Gho, M. and Louvet-Vallee, S. (2022). A neural progenitor mitotic wave is required for asynchronous axon outgrowth and morphology. Elife 11. PubMed ID: 35254258

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

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

  • Bingwei Lu Stanford Institute for Neuro-Innovation & Translational Neurosciences
    Ojha, R., Tantray, I., Rimal, S., Mitra, S., Cheshier, S. and Lu, B. (2022). Regulation of reverse electron transfer at mitochondrial complex I by unconventional Notch action in cancer stem cells. Dev Cell 57(2): 260-276.e269. PubMed ID: 35077680

    Li, S., Wu, Z., Li, Y., Tantray, I., De Stefani, D., Mattarei, A., Krishnan, G., Gao, F. B., Vogel, H. and Lu, B. (2020). Altered MICOS Morphology and Mitochondrial Ion Homeostasis Contribute to Poly(GR) Toxicity Associated with C9-ALS/FTD. Cell Rep 32(5): 107989. PubMed ID: 32755582

    Wen, X., An, P., Li, H., Zhou, Z., Sun, Y., Wang, J., Ma, L. and Lu, B. (2020). Tau Accumulation via Reduced Autophagy Mediates GGGGCC Repeat Expansion-Induced Neurodegeneration in Drosophila Model of ALS. Neurosci Bull. PubMed ID: 32500377

    Lee, K. S., Huh, S., Lee, S., Wu, Z., Kim, A. K., Kang, H. Y. and Lu, B. (2018). Altered ER-mitochondria contact impacts mitochondria calcium homeostasis and contributes to neurodegeneration in vivo in disease models. Proc Natl Acad Sci U S A. PubMed ID: 30185553

    Wu, Y. C., Lee, K. S., Song, Y., Gehrke, S. and Lu, B. (2017). The bantam microRNA acts through Numb to exert cell growth control and feedback regulation of Notch in tumor-forming stem cells in the Drosophila brain. PLoS Genet 13(5): e1006785. PubMed ID: 28520736

    Yao, Y., Cui, X., Al-Ramahi, I., Sun, X., Li, B., Hou, J., Difiglia, M., Palacino, J., Wu, Z. Y., Ma, L., Botas, J. and Lu, B. (2015). A striatal-enriched intronic GPCR modulates huntingtin levels and toxicity. Elife 4. PubMed ID: 25738228

    Lee, K. S. and Lu, B. (2014). The myriad roles of Miro in the nervous system: axonal transport of mitochondria and beyond. Front Cell Neurosci 8: 330. PubMed ID: 25389385

    Lee, K. S., Wu, Z., Song, Y., Mitra, S. S., Feroze, A. H., Cheshier, S. H. and Lu, B. (2013). Roles of PINK1, mTORC2, and mitochondria in preserving brain tumor-forming stem cells in a noncanonical Notch signaling pathway. Genes Dev 27: 2642-2647. PubMed ID: 24352421

    Wu, Z., Sawada, T., Shiba, K., Liu, S., Kanao, T., Takahashi, R., Hattori, N., Imai, Y., Lu, B. (2013) Tricornered/NDR kinase signaling mediates PINK1-directed mitochondrial quality control and tissue maintenance. Genes Dev 27: 157-162. PubMed ID: 23348839

    Lee, S., Wang, J. W., Yu, W. and Lu, B. (2012). Phospho-dependent ubiquitination and degradation of PAR-1 regulates synaptic morphology and tau-mediated Abeta toxicity in Drosophila. Nat Commun 3: 1312. PubMed ID: 23271647

  • Zhe Lu Perelman School of Medicine, University of Pennsylvania, Philadelphia
    Xu, Y., Ramu, Y., Shin, H. G., Yamakaze, J., Lu, Z. (2013) Energetic role of the paddle motif in voltage gating of Shaker K channels. Nat Struct Mol Biol. PubMed ID: 23542156

    Takemura, S. Y., Karuppudurai, T., Ting, C. Y., Lu, Z., Lee, C. H. and Meinertzhagen, I. A. (2011). Cholinergic circuits integrate neighboring visual signals in a Drosophila motion detection pathway. Curr Biol 21: 2077-2084. PubMed ID: 22137471

    Rivera-Alba, M., Vitaladevuni, S. N., Mishchenko, Y., Lu, Z., Takemura, S. Y., Scheffer, L., Meinertzhagen, I. A., Chklovskii, D. B. and de Polavieja, G. G. (2011). Wiring economy and volume exclusion determine neuronal placement in the Drosophila brain. Curr Biol 21: 2000-2005. PubMed ID: 22119527

  • John Lucchesi Department of Biology, Emory University, Atlanta
    Kuroda, M. I., Hilfiker, A. and Lucchesi, J. C. (2016). Dosage compensation in Drosophila-a model for the coordinate regulation of transcription. Genetics 204: 435-450. PubMed ID: 27729494

    Cugusi, S., Li, Y., Jin, P. and Lucchesi, J.C. (2016). The Drosophila helicase MLE targets hairpin structures in genomic transcripts. PLoS Genet 12: e1005761. PubMed ID: 26752049

    Lucchesi, J. C. and Kuroda, M. I. (2015). Dosage Compensation in Drosophila. Cold Spring Harb Perspect Biol 7. PubMed ID: 25934013

    Cugusi, S., Kallappagoudar, S., Ling, H. and Lucchesi, J.C. (2015). The Drosophila helicase MLE is implicated in functions distinct from its role in dosage compensation. Mol Cell Proteomics [Epub ahead of print]. PubMed ID: 25776889

    Cugusi, S., Ramos, E., Ling, H., Yokoyama, R., Luk, K. M. and Lucchesi, J. C. (2013). Topoisomerase II plays a role in dosage compensation in Drosophila. Transcription 4. PubMed ID: 23989663

    Dunlap, D., Yokoyama, R., Ling, H., Sun, H. Y., McGill, K., Cugusi, S. and Lucchesi, J. C. (2012). Distinct contributions of MSL complex subunits to the transcriptional enhancement responsible for dosage compensation in Drosophila. Nucleic Acids Res 40: 11281-11291. PubMed ID: 23047951

    Bhadra, M. P., Horikoshi, N., Pushpavallipvalli, S. N., Sarkar, A., Bag, I., Krishnan, A., Lucchesi, J. C., Kumar, R., Yang, Q., Pandita, R. K., Singh, M., Bhadra, U., Eissenberg, J. C. and Pandita, T. K. (2012). The role of MOF in the ionizing radiation response is conserved in Drosophila melanogaster. Chromosoma 121: 79-90. PubMed ID: 22072291

    Morra, R., Yokoyama, R., Ling, H. and Lucchesi, J. C. (2011). Role of the ATPase/helicase maleless (MLE) in the assembly, targeting, spreading and function of the male-specific lethal (MSL) complex of Drosophila. Epigenetics Chromatin 4: 6. PubMed ID: 21486482

  • Martha J. Lundell Department of Biology, University of Texas San Diego
    Perez, E., Venkatanarayan, A. and Lundell, M. J. (2022). Hunchback prevents notch-induced apoptosis in the serotonergic lineage of Drosophila Melanogaster. Dev Biol. PubMed ID: 35381219

    Guntur, A. R., Venkatanarayan, A., Gangula, S. and Lundell, M. J. (2021). Zfh-2 facilitates Notch-induced apoptosis in the CNS and appendages of Drosophila melanogaster. Dev Biol 475: 65-79. PubMed ID: 33705738

    Ahantarig, A., Chadwell, L. V., Terrazas, I. B., Garcia, C. T., Nazarian, J. J., Lee, H. K., Lundell, M. J. and Cassill, J. A. (2009). Molecular characterization of Pegarn: a Drosophila homolog of UNC-51 kinase. Mol Biol Rep 36(6): 1311-1321. PubMed ID: 18636236

    Lee, H. K. and Lundell, M. J. (2007). Differentiation of the Drosophila serotonergic lineage depends on the regulation of Zfh-1 by Notch and Eagle. Mol Cell Neurosci 36(1): 47-58. PubMed ID: 17702602

  • William Ludington Carnegie Institution for Science, Department of Embryology, Baltimore, Maryland
    Dodge, R., Jones, E. W., Zhu, H., Obadia, B., Martinez, D. J., Wang, C., Aranda-Diaz, A., Aumiller, K., Liu, Z., Voltolini, M., Brodie, E. L., Huang, K. C., Carlson, J. M., Sivak, D. A., Spradling, A. C. and Ludington, W. B. (2023). A symbiotic physical niche in Drosophila melanogaster regulates stable association of a multi-species gut microbiota. Nat Commun 14(1): 1557. PubMed ID: 36944617

    Chandler, J. A., Innocent, L. V., Martinez, D. J., Huang, I. L., Yang, J. L., Eisen, M. B. and Ludington, W. B. (2022). Microbiome-by-ethanol interactions impact Drosophila melanogaster fitness, physiology, and behavior. iScience 25(4): 104000. PubMed ID: 35313693

    Jones, E. W., Carlson, J. M., Sivak, D. A. and Ludington, W. B. (2022). Stochastic microbiome assembly depends on context. Proc Natl Acad Sci U S A 119(7). PubMed ID: 35135881

    Ludington, W. B. and Ja, W. W. (2020). Drosophila as a model for the gut microbiome. PLoS Pathog 16(4): e1008398. PubMed ID: 32324814

    Aranda-Diaz, A., Obadia, B., Dodge, R., Thomsen, T., Hallberg, Z. F., Guvener, Z. T., Ludington, W. B. and Huang, K. C. (2020). Bacterial interspecies interactions modulate pH-mediated antibiotic tolerance. Elife 9. PubMed ID: 31995029

  • Erik Lundgren Department of Molecular Biology, Umea University, Sweden
    Andersson, K., Pokrzywa, M., Dacklin, I. and Lundgren, E. (2013). Inhibition of TTR Aggregation-Induced Cell Death - A New Role for Serum Amyloid P Component. PLoS One 8: e55766. PubMed ID: 23390551

    Noborn, F., O'Callaghan, P., Hermansson, E., Zhang, X., Ancsin, J. B., Damas, A. M., Dacklin, I., Presto, J., Johansson, J., Saraiva, M. J., Lundgren, E., Kisilevsky, R., Westermark, P. and Li, J. P. (2011). Heparan sulfate/heparin promotes transthyretin fibrillization through selective binding to a basic motif in the protein. Proc Natl Acad Sci U S A 108: 5584-5589. PubMed ID: 21422279

    Pokrzywa, M., Dacklin, I., Vestling, M., Hultmark, D., Lundgren, E. and Cantera, R. (2010). Uptake of aggregating transthyretin by fat body in a Drosophila model for TTR-associated amyloidosis. PLoS One 5: e14343. PubMed ID: 21179560

  • Hong Luo School of Life Sciences, Tsinghua University, Beijing
    Zhou, Y., Yang, Y., Huang, Y., Wang, H., Wang, S. and Luo, H. (2019). Broad promotes neuroepithelial stem cell differentiation in the Drosophila optic lobe. Genetics. PubMed ID: 31530575

    Chen, X., Quan, Y., Wang, H. and Luo, H. (2014). Trehalase regulates neuroepithelial stem cell maintenance and differentiation in the Drosophila optic lobe. PLoS One 9: e101433. PubMed ID: 25003205

    Wang, H., Chen, X., He, T., Zhou, Y. and Luo, H. (2013). Evidence for tissue-specific Jak/STAT target genes in Drosophila optic lobe development. Genetics 195: 1291-1306. PubMed ID: 24077308

    Zhou, L. and Luo, H. (2013). Replication protein a links cell cycle progression and the onset of neurogenesis in Drosophila optic lobe development. J Neurosci 33: 2873-2888. PubMed ID: 23407946

    Siddiqui, N. U., Li, X., Luo, H., Karaiskakis, A., Hou, H., Kislinger, T., Westwood, J. T., Morris, Q. and Lipshitz, H. D. (2012). Genome-wide analysis of the maternal-to-zygotic transition in Drosophila primordial germ cells. Genome Biol 13: R11. PubMed ID: 22348290

    Wang, W., Li, Y., Zhou, L., Yue, H. and Luo, H. (2011). Role of JAK/STAT signaling in neuroepithelial stem cell maintenance and proliferation in the Drosophila optic lobe. Biochem Biophys Res Commun 410: 714-720. PubMed ID: 21651897

    Wang, W., Liu, W., Wang, Y., Zhou, L., Tang, X. and Luo, H. (2011). Notch signaling regulates neuroepithelial stem cell maintenance and neuroblast formation in Drosophila optic lobe development. Dev Biol 350: 414-428. PubMed ID: 21146517

  • Liqun Luo HHMI, Department of Biology, Stanford University
    Xie, Q., Li, J., Li, H., Udeshi, N. D., Svinkina, T., Orlin, D., Kohani, S., Guajardo, R., Mani, D. R., Xu, C., Li, T., Han, S., Wei, W., Shuster, S. A., Luginbuhl, D. J., Quake, S. R., Murthy, S. E., Ting, A. Y., Carr, S. A. and Luo, L. (2022). Transcription factor Acj6 controls dendrite targeting via a combinatorial cell-surface code. Neuron. PubMed ID: 35613619

    McLaughlin, C. N., Qi, Y., Quake, S. R., Luo, L. and Li, H. (2022). Isolation and RNA sequencing of single nuclei from Drosophila tissues. STAR Protoc 3(2): 101417. PubMed ID: 35620068

    Li, T., Fu, T. M., Wong, K. K. L., Li, H., Xie, Q., Luginbuhl, D. J., Wagner, M. J., Betzig, E. and Luo, L. (2021). Cellular bases of olfactory circuit assembly revealed by systematic time-lapse imaging. Cell 184(20): 5107-5121. PubMed ID: 34551316

    Li, T. and Luo, L. (2021). An Explant System for Time-Lapse Imaging Studies of Olfactory Circuit Assembly in Drosophila. J Vis Exp(176). PubMed ID: 34723938

    McLaughlin, C. N., Brbic, M., Xie, Q., Li, T., Horns, F., Kolluru, S. S., Kebschull, J. M., Vacek, D., Xie, A., Li, J., Jones, R. C., Leskovec, J., Quake, S. R., Luo, L. and Li, H. (2021). Single-cell transcriptomes of developing and adult olfactory receptor neurons in Drosophila. Elife 10. PubMed ID: 33555999

    Xie, Q., Brbic, M., Horns, F., Kolluru, S. S., Jones, R. C., Li, J., Reddy, A. R., Xie, A., Kohani, S., Li, Z., McLaughlin, C. N., Li, T., Xu, C., Vacek, D., Luginbuhl, D. J., Leskovec, J., Quake, S. R., Luo, L. and Li, H. (2021). Temporal evolution of single-cell transcriptomes of Drosophila olfactory projection neurons. Elife 10. PubMed ID: 33427646

    Li, H., Li, T., Horns, F., Li, J., Xie, Q., Xu, C., Wu, B., Kebschull, J. M., McLaughlin, C. N., Kolluru, S. S., Jones, R. C., Vacek, D., Xie, A., Luginbuhl, D. J., Quake, S. R. and Luo, L. (2020). Single-Cell Transcriptomes Reveal Diverse Regulatory Strategies for Olfactory Receptor Expression and Axon Targeting. Curr Biol. PubMed ID: 32059767

    Li, J., Han, S., Li, H., Udeshi, N. D., Svinkina, T., Mani, D. R., Xu, C., Guajardo, R., Xie, Q., Li, T., Luginbuhl, D. J., Wu, B., McLaughlin, C. N., Xie, A., Kaewsapsak, P., Quake, S. R., Carr, S. A., Ting, A. Y. and Luo, L. (2020). Cell-Surface Proteomic Profiling in the Fly Brain Uncovers Wiring Regulators. Cell 180(2): 373-386.e315. PubMed ID: 31955847

    Xie, Q., Wu, B., Li, J., Xu, C., Li, H., Luginbuhl, D. J., Wang, X., Ward, A. and Luo, L. (2019). Transsynaptic Fish-lips signaling prevents misconnections between nonsynaptic partner olfactory neurons. Proc Natl Acad Sci U S A. PubMed ID: 31341080

    Guajardo, R., Luginbuhl, D. J., Han, S., Luo, L. and Li, J. (2019). Functional divergence of Plexin B structural motifs in distinct steps of Drosophila olfactory circuit assembly. Elife 8. PubMed ID: 31225795

    Li, J., Guajardo, R., Xu, C., Wu, B., Li, H., Li, T., Luginbuhl, D. J., Xie, X. and Luo, L. (2018). Stepwise wiring of the Drosophila olfactory map requires specific Plexin B levels. Elife 7. PubMed ID: 30136927

    Li, H., Horns, F., Wu, B., Xie, Q., Li, J., Li, T., Luginbuhl, D. J., Quake, S. R. and Luo, L. (2017). Classifying Drosophila olfactory projection neuron subtypes by single-cell RNA sequencing. Cell 171(5): 1206-1220 PubMed ID: 29149607

  • Oleh Lushchak Department of Biochemistry and Biotechnology, Natural Sciences Institute, Vasyl Stefanyk Precarpathian National University, Ukraine
    Semaniuk, U. V., Gospodaryov, D. V., Strilbytska, O. M., Kucharska, A. Z., Sokot-Letowska, A., Burdyliuk, N. I., Storey, K. B., Bayliak, M. M. and Lushchak, O. (2022). Chili-supplemented food decreases glutathione-S-transferase activity in Drosophila melanogaster females without a change in other parameters of antioxidant system. Redox Rep 27(1): 221-229. PubMed ID: 36200601

    Strilbytska, O., Strutynska, T., Semaniuk, U., Burdyliyk, N., Bubalo, V. and Lushchak, O. (2022). Dietary Sucrose Determines Stress Resistance, Oxidative Damages, and Antioxidant Defense System in Drosophila. Scientifica (Cairo) 2022: 7262342. PubMed ID: 35547569

    Strilbytska, O., Semaniuk, U., Bubalo, V., Storey, K. B. and Lushchak, O. (2022). Dietary Choice Reshapes Metabolism in Drosophila by Affecting Consumption of Macronutrients. Biomolecules 12(9). PubMed ID: 36139040

    Strilbytska, O. M., Semaniuk, U. V., Strutynska, T. R., Burdyliuk, N. I., Tsiumpala, S., Bubalo, V. and Lushchak, O. (2022). Herbicide Roundup shows toxic effects in nontarget organism Drosophila. Arch Insect Biochem Physiol: e21893. PubMed ID: 35388481

    Semaniuk, U., Gospodaryov, D., Mishchanyn, K., Storey, K. and Lushchak, O. (2021). Drosophila insulin-like peptides regulate concentration-dependent changes of appetite to different carbohydrates. Zoology (Jena) 146: 125927. PubMed ID: 33894679

    Strilbytska, O. M., Zayachkivska, A., Koliada, A., Galeotti, F., Volpi, N., Storey, K. B., Vaiserman, A. and Lushchak, O. (2020). Anise Hyssop Agastache foeniculum Increases Lifespan, Stress Resistance, and Metabolism by Affecting Free Radical Processes in Drosophila. Front Physiol 11: 596729. PubMed ID: 33391017

    Yurkevych, I. S., Gray, L. J., Gospodaryov, D. V., Burdylyuk, N. I., Storey, K. B., Simpson, S. J. and Lushchak, O. (2020). Development of fly tolerance to consuming a high-protein diet requires physiological, metabolic and transcriptional changes. Biogerontology. PubMed ID: 32468146

    Strilbytska, O. M., Semaniuk, U. V., Storey, K. B., Yurkevych, I. S. and Lushchak, O. (2020). Insulin Signaling in Intestinal Stem and Progenitor Cells as an Important Determinant of Physiological and Metabolic Traits in Drosophila. Cells 9(4). PubMed ID: 32225024

    Strilbytska, O. M., Storey, K. B. and Lushchak, O. V. (2020). TOR signaling inhibition in intestinal stem and progenitor cells affects physiology and metabolism in Drosophila. Comp Biochem Physiol B Biochem Mol Biol 243-244: 110424. PubMed ID: 32088257

    Koliada, A., Gavrilyuk, K., Burdylyuk, N., Strilbytska, O., Storey, K. B., Kuharskii, V., Lushchak, O. and Vaiserman, A. (2020). Mating status affects Drosophila lifespan, metabolism and antioxidant system. Comp Biochem Physiol A Mol Integr Physiol: 110716. PubMed ID: 32339661

    Strilbytska, O. M., Semaniuk, U. V., Storey, K. B., Yurkevych, I. S. and Lushchak, O. (2020). Insulin Signaling in Intestinal Stem and Progenitor Cells as an Important Determinant of Physiological and Metabolic Traits in Drosophila. Cells 9(4). PubMed ID: 32225024

    Semaniuk, U. V., Gospodaryov, D. V., Feden'ko, K. M., Yurkevych, I. S., Vaiserman, A. M., Storey, K. B., Simpson, S. J. and Lushchak, O. (2018). Insulin-like peptides regulate feeding preference and metabolism in Drosophila. Front Physiol 9: 1083. PubMed ID: 30197596

  • Volodymyr Lushchak Department of Biochemistry and Biotechnology, Precarpathian National University. Ukraine
    Bayliak, M. M., Demianchuk, O. I., Gospodaryov, D. V., Abrat, O. B., Lylyk, M. P., Storey, K. B. and Lushchak, V. I. (2020). Mutations in genes cnc or dKeap1 modulate stress resistance and metabolic processes in Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol 248: 110746. PubMed ID: 32579905

    Abrat, O. B., Storey, J. M., Storey, K. B. and Lushchak, V. I. (2018). High amylose starch consumption induces obesity in Drosophila melanogaster and metformin partially prevents accumulation of storage lipids and shortens lifespan of the insects. Comp Biochem Physiol A Mol Integr Physiol 215: 55-62. PubMed ID: 29054808

    Bayliak, M. M., Lylyk, M. P., Maniukh, O. V., Storey, J. M., Storey, K. B. and Lushchak, V. I. (2017). Dietary L-arginine accelerates pupation and promotes high protein levels but induces oxidative stress and reduces fecundity and life span in Drosophila melanogaster. J Comp Physiol B. PubMed ID: 28668996

    Bayliak, M. M., Lylyk, M. P., Shmihel, H. V., Sorochynska, O. M., Semchyshyn, O. I., Storey, J. M., Storey, K. B. and Lushchak, V. I. (2016). Dietary alpha-ketoglutarate promotes higher protein and lower triacylglyceride levels and induces oxidative stress in larvae and young adults but not in middle-aged Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol. PubMed ID: 27842224

    Rovenko, B. M., Kubrak, O. I., Gospodaryov, D. V., Perkhulyn, N. V., Yurkevych, I. S., Sanz, A., Lushchak, O. V. and Lushchak, V. I. (2015). High sucrose consumption promotes obesity whereas its low consumption induces oxidative stress in Drosophila melanogaster. J Insect Physiol 79: 42-54. PubMed ID: 26050918

    Rovenko, B. M., Kubrak, O. I., Gospodaryov, D. V., Yurkevych, I. S., Sanz, A., Lushchak, O. V. and Lushchak, V. I. (2015). Restriction of glucose and fructose causes mild oxidative stress independently of mitochondrial activity and reactive oxygen species in Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol 187: 27-39. PubMed ID: 25941153

    Perkhulyn, N. V., Rovenko, B. M., Zvarych, T. V., Lushchak, O. V., Storey, J. M., Storey, K. B. and Lushchak, V. I. (2015). Sodium chromate demonstrates some insulin-mimetic properties in the fruit fly Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 167: 74-80. PubMed ID: 25220772

    Rovenko, B. M., Perkhulyn, N. V., Gospodaryov, D. V., Sanz, A., Lushchak, O. V. and Lushchak, V. I. (2014). High consumption of fructose rather than glucose promotes a diet-induced obese phenotype in Drosophila melanogaster. Comp Biochem Physiol A Mol Integr Physiol 180C: 75-85. PubMed ID: 25461489

    Rovenko, B. M., Perkhulyn, N. V., Lushchak, O. V., Storey, J. M., Storey, K. B. and Lushchak, V. I. (2014). Molybdate partly mimics insulin-promoted metabolic effects in Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 165: 76-82. PubMed ID: 24952334

    Lozinsky, O. V., Lushchak, O. V. and Lushchak, V. I. (2013). 2,4-dinitrophenol partially alleviates ferrocyanide-induced toxicity in Drosophila melanogaster. Arch Insect Biochem Physiol 84: 157-173. PubMed ID: 24123210

  • Stefan Luschnig University of Münster
    Camelo, C., Korte, A., Jacobs, T. and Luschnig, S. (2022). Tracheal tube fusion in Drosophila involves release of extracellular vesicles from multivesicular bodies. J Cell Sci. PubMed ID: 35019140

    Isasti-Sanchez, J., Munz-Zeise, F., Lancino, M. and Luschnig, S. (2021). Transient opening of tricellular vertices controls paracellular transport through the follicle epithelium during Drosophila oogenesis. Dev Cell 56(8): 1083-1099. PubMed ID: 33831351

    Wittek, A., Hollmann, M., Schleutker, R. and Luschnig, S. (2020). The Transmembrane Proteins M6 and Anakonda Cooperate to Initiate Tricellular Junction Assembly in Epithelia of Drosophila. Curr Biol. PubMed ID: 32857972

    Sauerwald, J., Backer, W., Matzat, T., Schnorrer, F. and Luschnig, S. (2019). Matrix metalloproteinase 1 modulates invasive behavior of tracheal branches during entry into Drosophila flight muscles. Elife 8. PubMed ID: 31577228

    Sauerwald, J., Soneson, C., Robinson, M. D. and Luschnig, S. (2017). Faithful mRNA splicing depends on the Prp19 complex subunit faint sausage and is required for tracheal branching morphogenesis in Drosophila. Development [Epub ahead of print]. PubMed ID: 28087625

    Misra, T., Baccino-Calace, M., Meyenhofer, F., Rodriguez-Crespo, D., Akarsu, H., Armenta-Calderon, R., Gorr, T. A., Frei, C., Cantera, R., Egger, B. and Luschnig, S. (2016). A genetically encoded biosensor for visualizing hypoxia responses in vivo. Biol Open. PubMed ID: 28011628

    Caviglia, S., Brankatschk, M., Fischer, E. J., Eaton, S. and Luschnig, S. (2016). Staccato/Unc-13-4 controls secretory lysosome-mediated lumen fusion during epithelial tube anastomosis. Nat Cell Biol 18: 727-739. PubMed ID: 27323327

    De Lella Ezcurra, A. L., Bertolin, A. P., Kim, K., Katz, M. J., Gandara, L., Misra, T., Luschnig, S., Perrimon, N., Melani, M. and Wappner, P. (2016). miR-190 Enhances HIF-Dependent Responses to Hypoxia in Drosophila by Inhibiting the Prolyl-4-hydroxylase Fatiga. PLoS Genet 12: e1006073. PubMed ID: 27223464

    Byri, S., Misra, T., Syed, Z.A., Bätz, T., Shah, J., Boril, L., Glashauser, J., Aegerter-Wilmsen, T., Matzat, T., Moussian, B., Uv, A. and Luschnig, S. (2015). The triple-repeat protein Anakonda controls epithelial tricellular junction formation in Drosophila. Dev Cell [Epub ahead of print]. PubMed ID: 25982676

    Caviglia, S. and Luschnig, S. (2014). Tube fusion: Making connections in branched tubular networks. Semin Cell Dev Biol. PubMed ID: 24662893

  • Frank Lyko German Cancer Research Center, Heidelberg
    Raddatz, G., Guzzardo, P. M., Olova, N., Fantappie, M. R., Rampp, M., Schaefer, M., Reik, W., Hannon, G. J. and Lyko, F. (2013). Dnmt2-dependent methylomes lack defined DNA methylation patterns. Proc Natl Acad Sci U S A. PubMed ID: 23641003

    Durdevic, Z., Hanna, K., Gold, B., Pollex, T., Cherry, S., Lyko, F., Schaefer, M. (2013) Efficient RNA virus control in Drosophila requires the RNA methyltransferase Dnmt2. EMBO Rep. PubMed ID: 23370384

    Schaefer, M. and Lyko, F. (2010). Lack of evidence for DNA methylation of Invader4 retroelements in Drosophila and implications for Dnmt2-mediated epigenetic regulation. Nat Genet 42: 920-921; author reply 921. PubMed ID: 20980983

  • Lisa Lyons Florida State University, Department of Biological Science, Tallahassee
    De Nobrega, A. K., Noakes, E. J., Storch, N. A., Mellers, A. P. and Lyons, L. C. (2022). Sleep Modulates Alcohol Toxicity in Drosophila. Int J Mol Sci 23(20). PubMed ID: 36292943

    De Nobrega, A. K., Luz, K. V., Lyons, K. N. and Lyons, L. C. (2020). Investigating Alcohol Behavior and Physiology Using Drosophila melanogaster. Methods Mol Biol 2138: 135-158. PubMed ID: 32219744

    De Nobrega, A. K. and Lyons, L. C. (2017). Drosophila: An Emergent Model for Delineating Interactions between the Circadian Clock and Drugs of Abuse. Neural Plast 2017: 4723836. PubMed ID: 29391952

    De Nobrega, A. K., Mellers, A. P. and Lyons, L. C. (2017). Aging and circadian dysfunction increase alcohol sensitivity and exacerbate mortality in Drosophila melanogaster. Exp Gerontol 97: 49-59. PubMed ID: 28750752

  • Ping-Chiang Lyu Institute of Bioinformatics and Structural Biology & Department of Life Sciences, National Tsing Hua University
    Cheng, Y. Y., Huang, Y. F., Lin, H. H., Chang, W. W. and Lyu, P. C. (2019). The ligand-mediated affinity of brain-type fatty acid-binding protein for membranes determines the directionality of lipophilic cargo transport. Biochim Biophys Acta Mol Cell Biol Lipids 1864(12): 158506. PubMed ID: 31404652

    Tseng, T. S., Cheng, C. S., Hsu, S. T., Shih, M. F., He, P. L., Lyu, P. C. (2013) Residue-Specific Annotation of Disorder-to-Order Transition and Cathepsin Inhibition of a Propeptide-Like Crammer from D. melanogaster. PLoS One 8: e54187. PubMed ID: 23349821

    Chan, H. T., Lee, T. R., Huang, S. H., Lee, H. Y., Sang, T. K., Chan, H. L. and Lyu, P. C. (2012). Proteomic analysis of a drosophila IBMPFD model reveals potential pathogenic mechanisms. Mol Biosyst 8: 1730-1741. PubMed ID: 22481368

    Tseng, T. S., Cheng, C. S., Chen, D. J., Shih, M. F., Liu, Y. N., Hsu, S. T. and Lyu, P. C. (2012). A molten globule-to-ordered structure transition of Drosophila melanogaster crammer is required for its ability to inhibit cathepsin. Biochem J 442: 563-572. PubMed ID: 22150223


  • Return to The Interactive Fly