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


  • Fabrizio Gabbiani Department of Neuroscience, Baylor College of Medicine, Dallas
    Dewell, R. B. and Gabbiani, F. (2012). Escape behavior: linking neural computation to action. Curr Biol 22: R152-153. PubMed ID: 22401892

    Fotowat, H. and Gabbiani, F. (2011). Collision detection as a model for sensory-motor integration. Annu Rev Neurosci 34: 1-19. PubMed ID: 21391815

    Fotowat, H., Harrison, R. R. and Gabbiani, F. (2011). Multiplexing of motor information in the discharge of a collision detecting neuron during escape behaviors. Neuron 69: 147-158. PubMed ID: 21220105

  • Martina Galikova Department of Zoology, Stockholm University
    Klepsatel, P. and Galikova, M. (2022). Developmental temperature affects thermal dependence of locomotor activity in Drosophila. J Therm Biol 103: 103153. PubMed ID: 35027204

    Klepsatel, P., Girish, T. N. and Galikova, M. (2020). Acclimation temperature affects thermal reaction norms for energy reserves in Drosophila. Sci Rep 10(1): 21681. PubMed ID: 33303846

    Klepsatel, P., Knoblochova, D., Girish, T. N., Dircksen, H. and Galikova, M. (2020). The influence of developmental diet on reproduction and metabolism in Drosophila. BMC Evol Biol 20(1): 93. PubMed ID: 32727355

    Klepsatel, P., Girish, T. N., Dircksen, H. and Galikova, M. (2019). Reproductive fitness of Drosophila is maximised by optimal developmental temperature. J Exp Biol 222(Pt 10). PubMed ID: 31064855

    Klepsatel, P., Wildridge, D. and Galikova, M. (2019). Temperature induces changes in Drosophila energy stores. Sci Rep 9(1): 5239. PubMed ID: 30918312

  • Ibo Galindo Centro de Investigación Príncipe Felipe, Valencia, Spain
    Tapia, A., Palomino-Schatzlein, M., Roca, M., Lahoz, A., Pineda-Lucena, A., Lopez Del Amo, V. and Galindo, M. I. (2021). Mild Muscle Mitochondrial Fusion Distress Extends Drosophila Lifespan through an Early and Systemic Metabolome Reorganization. Int J Mol Sci 22(22). PubMed ID: 34830014

    Tapia, A., Giachello, C. N., Palomino-Schatzlein, M., Baines, R. A. and Galindo, M. I. (2021). Generation and Characterization of the Drosophila melanogaster paralytic Gene Knock-Out as a Model for Dravet Syndrome. Life (Basel) 11(11). PubMed ID: 34833136

    Calpena, E., Lopez Del Amo, V., Chakraborty, M., Llamusi, B., Artero, R., Espinos, C. and Galindo, M. I. (2018). The Drosophila junctophilin gene is functionally equivalent to its four mammalian counterparts and is a modifier of a Huntingtin poly-Q expansion and the Notch pathway. Dis Model Mech 11(1). PubMed ID: 29208631

    Lopez Del Amo, V., Palomino-Schatzlein, M., Seco-Cervera, M., Garcia-Gimenez, J. L., Pallardo, F. V., Pineda-Lucena, A. and Galindo, M. I. (2017). A Drosophila model of GDAP1 function reveals the involvement of insulin signalling in the mitochondria-dependent neuromuscular degeneration. Biochim Biophys Acta 1863(3): 801-809. PubMed ID: 28065847

    Calpena, E., Palau, F., Espinos, C. and Galindo, M. I. (2015). Evolutionary History of the Smyd Gene Family in Metazoans: A Framework to Identify the Orthologs of Human Smyd Genes in Drosophila and Other Animal Species. PLoS One 10(7): e0134106. PubMed ID: 26230726

    Lopez Del Amo, V., Seco-Cervera, M., Garcia-Gimenez, J. L., Whitworth, A. J., Pallardo, F. V. and Galindo, M. I. (2015). Mitochondrial defects and neuromuscular degeneration caused by altered expression of Drosophila Gdap1: implications for the Charcot-Marie-Tooth neuropathy. Hum Mol Genet 24(1): 21-36. PubMed ID: 25122658

  • Giovanni Galizia Universität Konstanz Fachbereich Biologie
    Szyszka, P. and Galizia, C. G. (2018). The Role of the Sucrose-Responsive IR60b Neuron for Drosophila melanogaster: A Hypothesis. Chem Senses. PubMed ID: 29546407

    Martinelli, E., Ludke, A., Adamo, P., Strauch, M., Di Natale, C. and Galizia, C. G. (2017). Normalizing brain activity across individuals using functional reference mapping. Sci Rep 7(1): 17128. PubMed ID: 29214995

    Ignatious Raja, J. S., Katanayeva, N., Katanaev, V. L. and Galizia, C. G. (2014). Role of G subgroup of G proteins in olfactory signaling of Drosophila melanogaster. Eur J Neurosci. PubMed ID: 24443946

    Rein, J., Mustard, J. A., Strauch, M., Smith, B. H. and Galizia, C. G. (2013). Octopamine modulates activity of neural networks in the honey bee antennal lobe. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 199: 947-962. PubMed ID: 23681219

    Munch, D., Schmeichel, B., Silbering, A. F. and Galizia, C. G. (2013). Weaker ligands can dominate an odor blend due to syntopic interactions. Chem Senses 38: 293-304. PubMed ID: 23315042

  • Michael Galko M. D. Anderson Center, Houston
    Wang, Y., Lopez-Bellido, R., Huo, X., Kavelaars, A. and Galko, M. J. (2023). The insulin receptor regulates the persistence of mechanical nociceptive sensitization in flies and mice. Biol Open 12(6). PubMed ID: 37259940

    Tsai, C. R., Wang, Y., Jacobson, A., Sankoorikkal, N., Chirinos, J. D., Burra, S., Makthal, N., Kumaraswami, M. and Galko, M. J. (2021). Pvr and distinct downstream signaling factors are required for hemocyte spreading and epidermal wound closure at Drosophila larval wound sites. G3 (Bethesda). PubMed ID: 34751396

    Wang, Y., Burra, S. and Galko, M. J. (2021). Drosophila larval epidermal cells only exhibit epidermal aging when they persist to the adult stage. J Exp Biol. PubMed ID: 33795421

    Tsai, C. R. and Galko, M. J. (2019). Casein kinase 1alpha decreases beta-catenin levels at adherens junctions to facilitate wound closure in Drosophila larvae. Development. PubMed ID: 31511254

    Tsai, C. R., Wang, Y. and Galko, M. J. (2018). Crawling wounded: molecular genetic insights into wound healing from Drosophila larvae. Int J Dev Biol 62(6-7-8): 479-489. PubMed ID: 29938760

    Im, S. H., Patel, A. A., Cox, D. N. and Galko, M. J. (2018). Drosophila Insulin receptor regulates the persistence of injury-induced nociceptive sensitization. Dis Model Mech 11(5). PubMed ID: 29752280

    Tsai, C. R., Anderson, A. E., Burra, S., Jo, J. and Galko, M. J. (2017). Yorkie regulates epidermal wound healing in Drosophila larvae independently of cell proliferation and apoptosis. Dev Biol. PubMed ID: 28514643

    Jo, J., Im, S. H., Babcock, D. T., Iyer, S. C., Gunawan, F., Cox, D. N. and Galko, M. J. (2017). Drosophila caspase activity is required independently of apoptosis to produce active TNF/Eiger during nociceptive sensitization. Cell Death Dis 8(5): e2786. PubMed ID: 28492538

    Wang, Y., Antunes, M., Anderson, A. E., Kadrmas, J. L., Jacinto, A. and Galko, M. J. (2015). Integrin adhesions suppress syncytium formation in the Drosophila larval epidermis. Curr Biol 25: 2215-2227. PubMed ID: 26255846

    Scherfer, C., Han, V. C., Wang, Y., Anderson, A. E. and Galko, M. J. (2013). Autophagy drives epidermal deterioration in a Drosophila model of tissue aging. Aging (Albany NY). PubMed ID: 23599123

  • Joseph Gall Deparment of Embryology, Carnegie Institute for Science, Baltimore
    Deryusheva, S. and Gall, J. G. (2019). Small, smaller, smallest: Minimal structural requirements for a fully functional box C/D modification guide RNA. Biomolecules 9(9). PubMed ID: 31500270

    Deryusheva, S. and Gall, J. G. (2017). Orchestrated positioning of post-transcriptional modifications at the branch point recognition region of U2 snRNA. RNA [Epub ahead of print] PubMed ID: 28974555

    Craddock, E. M., Gall, J. G. and Jonas, M. (2016). Hawaiian Drosophila genomes: size variation and evolutionary expansions. Genetica [Epub ahead of print]. PubMed ID: 26790663

    Deryusheva, S. and Gall, J. G. (2013). Novel small Cajal-body-specific RNAs identified in Drosophila: probing guide RNA function. RNA. PubMed ID: 24149844

    Gall, J. G. (2012). Are lampbrush chromosomes unique to meiotic cells? Chromosome Res 20: 905-909. PubMed ID: 23263880

    Singer, A. B. and Gall, J. G. (2011). An inducible nuclear body in the Drosophila germinal vesicle. Nucleus 2: 403-409. PubMed ID: 21941118

    Nizami, Z. F., Deryusheva, S. and Gall, J. G. (2010). Cajal bodies and histone locus bodies in Drosophila and Xenopus. Cold Spring Harb Symp Quant Biol 75: 313-320. PubMed ID: 21047905

  • Marco Gallio Department of Neurobiology, Northwestern University, Evanston, IL
    Alpert, M. H., Gil, H., Para, A. and Gallio, M. (2022). A thermometer circuit for hot temperature adjusts Drosophila behavior to persistent heat. Curr Biol. PubMed ID: 35981537

    Simoes, J. M., Levy, J. I., Zaharieva, E. E., Vinson, L. T., Zhao, P., Alpert, M. H., Kath, W. L., Para, A. and Gallio, M. (2021). Robustness and plasticity in Drosophila heat avoidance. Nat Commun 12(1): 2044. PubMed ID: 33824330

    Alpert, M. H., Frank, D. D., Kaspi, E., Flourakis, M., Zaharieva, E. E., Allada, R., Para, A. and Gallio, M. (2020). A Circuit Encoding Absolute Cold Temperature in Drosophila. Curr Biol. PubMed ID: 32442464

    Frank, D. D., Enjin, A., Jouandet, G. C., Zaharieva, E. E., Para, A., Stensmyr, M. C. and Gallio, M. (2017). Early Integration of Temperature and Humidity Stimuli in the Drosophila Brain. Curr Biol 27(15): 2381-2388 e2384. PubMed ID: 28736172

    Enjin, A., Zaharieva, E. E., Frank, D. D., Mansourian, S., Suh, G. S., Gallio, M. and Stensmyr, M. C. (2016). Humidity Sensing in Drosophila. Curr Biol 26(10): 1352-1358. PubMed ID: 27161501

    Frank, D. D., Jouandet, G. C., Kearney, P. J., Macpherson, L. J. and Gallio, M. (2015). Temperature representation in the Drosophila brain. Nature 519(7543): 358-361. PubMed ID: 25739506

    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(3): 603-615. PubMed ID: 24507194

    Gallio, M., Ofstad, T. A., Macpherson, L. J., Wang, J. W. and Zuker, C. S. (2011). The coding of temperature in the Drosophila brain. Cell 144(4): 614-624. PubMed ID: 21335241

    Wang, S., Tsarouhas, V., Xylourgidis, N., Sabri, N., Tiklova, K., Nautiyal, N., Gallio, M. and Samakovlis, C. (2009). The tyrosine kinase Stitcher activates Grainy head and epidermal wound healing in Drosophila. Nat Cell Biol 11(7): 890-895. PubMed ID: 19525935

  • Maria Cristina Gambetta Center for Integrative Genomics, University of Lausanne, Switzerland
    Mohana, G., Dorier, J., Li, X., Mouginot, M., Smith, R. C., Malek, H., Leleu, M., Rodriguez, D., Khadka, J., Rosa, P., Cousin, P., Iseli, C., Restrepo, S., Guex, N., McCabe, B. D., Jankowski, A., Levine, M. S. and Gambetta, M. C. (2023). Chromosome-level organization of the regulatory genome in the Drosophila nervous system. Cell. PubMed ID: 37536338

    Kaushal, A., Dorier, J., Wang, B., Mohana, G., Taschner, M., Cousin, P., Waridel, P., Iseli, C., Semenova, A., Restrepo, S., Guex, N., Aiden, E. L. and Gambetta, M. C. (2022). Essential role of Cp190 in physical and regulatory boundary formation. Sci Adv 8(19): eabl8834. PubMed ID: 35559678

    Kaushal, A., Mohana, G., Dorier, J., Ozdemir, I., Omer, A., Cousin, P., Semenova, A., Taschner, M., Dergai, O., Marzetta, F., Iseli, C., Eliaz, Y., Weisz, D., Shamim, M. S., Guex, N., Lieberman Aiden, E. and Gambetta, M. C. (2021). CTCF loss has limited effects on global genome architecture in Drosophila despite critical regulatory functions. Nat Commun 12(1): 1011. PubMed ID: 33579945

    Ozdemir, I. and Gambetta, M. C. (2019). The Role of Insulation in Patterning Gene Expression. Genes (Basel) 10(10). PubMed ID: 31569427

    Gambetta, M. C. and Furlong, E. E. M. (2018). The Insulator Protein CTCF Is Required for Correct Hox Gene Expression, but Not for Embryonic Development in Drosophila. Genetics 210(1): 129-136. PubMed ID: 30021792

  • Barry Ganetzky Laboratory of Genetics, University of Wisconsin, Madison
    Katzenberger, R. J., Ganetzky, B. and Wassarman, D. A. (2023). Lissencephaly-1 mutations enhance traumatic brain injury outcomes in Drosophila. Genetics. PubMed ID: 36683334

    Scharenbrock, A. R., Katzenberger, R. J., Fischer, M. C., Ganetzky, B. and Wassarman, D. A. (2021). Beta-blockers reduce intestinal permeability and early mortality following traumatic brain injury in Drosophila. MicroPubl Biol 2021. PubMed ID: 34723144

    Blommer, J., Fischer, M. C., Olszewski, A. R., Katzenberger, R. J., Ganetzky, B. and Wassarman, D. A. (2021). Ketogenic diet reduces early mortality following traumatic brain injury in Drosophila via the PPARγ ortholog Eip75B. PLoS One 16(10): e0258873. PubMed ID: 34699541'

    Lika, J., Katzenberger, R. J., Ganetzky, B. and Wassarman, D. A. (2021). Effects of blunt force injuries in third-instar Drosophila larvae persist through metamorphosis and reduce adult lifespan. MicroPubl Biol 2021. PubMed ID: 34278243

    Swanson, L. C., Trujillo, E. A., Thiede, G. H., Katzenberger, R. J., Shishkova, E., Coon, J. J., Ganetzky, B. and Wassarman, D. A. (2020). Survival Following Traumatic Brain Injury in Drosophila Is Increased by Heterozygosity for a Mutation of the NF-κB Innate Immune Response Transcription Factor Relish. Genetics. PubMed ID: 33109529

    Swanson, L. C., Rimkus, S. A., Ganetzky, B. and Wassarman, D. A. (2020). Loss of the Antimicrobial Peptide Mechnikowin Protects Against Traumatic Brain Injury Outcomes in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 32631949

    Perry, S., Goel, P., Tran, N. L., Pinales, C., Buser, C., Miller, D. L., Ganetzky, B. and Dickman, D. (2020). Developmental arrest of Drosophila larvae elicits presynaptic depression and enables prolonged studies of neurodegeneration. Development. PubMed ID: 32345746

    Loewen, C., Boekhoff-Falk, G., Ganetzky, B. and Chtarbanova, S. (2018). A novel mutation in brain tumor causes both neural over-proliferation and neurodegeneration in adult Drosophila. G3 (Bethesda). PubMed ID: 30126833

    Cunningham, P. C., Waldeck, K., Ganetzky, B. and Babcock, D. T. (2018). Neurodegeneration and locomotor dysfunction in Drosophila scarlet mutants. J Cell Sci. PubMed ID: 30154211

    Olufs, Z. P. G., Loewen, C. A., Ganetzky, B., Wassarman, D. A. and Perouansky, M. (2018). Genetic variability affects absolute and relative potencies and kinetics of the anesthetics isoflurane and sevoflurane in Drosophila melanogaster. Sci Rep 8(1): 2348. PubMed ID: 29402974

    Loewen, C. A. and Ganetzky, B. (2018). Mito-nuclear Interactions Affecting Lifespan and Neurodegeneration in a Drosophila Model of Leigh Syndrome. Genetics. PubMed ID: 29496745

    Kounatidis, I., Chtarbanova, S., Cao, Y., Hayne, M., Jayanth, D., Ganetzky, B. and Ligoxygakis, P. (2017). NF-kappaB immunity in the brain determines fly lifespan in healthy aging and age-related neurodegeneration. Cell Rep 19(4): 836-848. PubMed ID: 28445733

  • Geoffrey Ganter University of New England, Biddeford, Maine
    Hale, C., Moulton, J. K., Otis, Y. and Ganter, G. (2022). ARMADILLO REGULATES NOCICEPTIVE SENSITIVITY IN THE ABSENCE OF INJURY. Mol Pain: 17448069221111155. PubMed ID: 35712882

    McParland, A., Moulton, J., Brann, C., Hale, C., Otis, Y. and Ganter, G. (2021). The brinker repressor system regulates injury-induced nociceptive sensitization in Drosophila melanogaster. Mol Pain 17: 17448069211037401. PubMed ID: 34399634

    Gjelsvik, K. J., Follansbee, T. L. and Ganter, G. K. (2018). Bone Morphogenetic Protein Glass Bottom Boat (BMP5/6/7/8) and its receptor Wishful Thinking (BMPRII) are required for injury-induced allodynia in Drosophila. Mol Pain 14: 1744806918802703. PubMed ID: 30259786

    Follansbee, T. L., Gjelsvik, K. J., Brann, C. L., McParland, A. L., Longhurst, C. A., Galko, M. J. and Ganter, G. K. (2017). Drosophila Nociceptive Sensitization Requires BMP Signaling via the Canonical SMAD Pathway. J Neurosci 37(35): 8524-8533. PubMed ID: 28855331

  • Ranjan Ganguly University of Tennessee, Knoxville
    Gimenez, L. E., Ghildyal, P., Fischer, K. E., Hu, H., Ja, W. W., Eaton, B. A., Wu, Y., Austad, S. N. and Ranjan, R. (2013). Modulation of methuselah expression targeted to Drosophila insulin-producing cells extends life and enhances oxidative stress resistance. Aging Cell 12: 121-129. PubMed ID: 23121290

    Morra, R., Kuruganti, S., Lam, V., Lucchesi, J. C. and Ganguly, R. (2010). Functional analysis of the cis-acting elements responsible for the induction of the Cyp6a8 and Cyp6g1 genes of Drosophila melanogaster by DDT, phenobarbital and caffeine. Insect Mol Biol 19: 121-130. PubMed ID: 20002224

  • Fen-Biao Gao Department of Neurology, University of Massachusetts Medical School, Worcester
    Lee, S., Jun, Y. W., Linares, G. R., Butler, B., Yuva-Adyemir, Y., Moore, J., Krishnan, G., Ruiz-Juarez, B., Santana, M., Pons, M., Silverman, N., Weng, Z., Ichida, J. K. and Gao, F. B. (2023). Downregulation of Hsp90 and the antimicrobial peptide Mtk suppresses poly(GR)-induced neurotoxicity in C9ORF72-ALS/FTD. Neuron. PubMed ID: 36931278

    Jun, Y. W., Lee, S., Ban, B. K., Lee, J. A. and Gao, F. B. (2023). Non-muscle MYH10/myosin IIB recruits ESCRT-III to participate in autophagosome closure to maintain neuronal homeostasis. Autophagy: 1-17. PubMed ID: 36849436

    Chen, Y., Krishnan, G., Parsi, S., Pons, M., Nikolaki, V., Cao, L., Xu, Z. and Gao, F. B. (2022). The enhanced association between mutant CHMP2B and spastin is a novel pathological link between frontotemporal dementia and hereditary spastic paraplegias. Acta Neuropathol Commun 10(1): 169. PubMed ID: 36414997

    Lu, Y., West, R. J. H., Pons, M., Sweeney, S. T. and Gao, F. B. (2020). Ik2/TBK1 and Hook/Dynein, an adaptor complex for early endosome transport, are genetic modifiers of FTD-associated mutant CHMP2B toxicity in Drosophila. Sci Rep 10(1): 14221. PubMed ID: 32848189

    Tran, H., Almeida, S., Moore, J., Gendron, T. F., Chalasani, U., Lu, Y., Du, X., Nickerson, J. A., Petrucelli, L., Weng, Z. and Gao, F. B. (2015). Differential toxicity of nuclear RNA foci versus dipeptide repeat proteins in a Drosophila model of C9ORF72 FTD/ALS. Neuron 87: 1207-1214. PubMed ID: 26402604

    Yang, D., Abdallah, A., Li, Z., Lu, Y., Almeida, S. and Gao, F. B. (2015). FTD/ALS-associated poly(GR) protein impairs the Notch pathway and is recruited by poly(GA) into cytoplasmic inclusions. Acta Neuropathol. PubMed ID: 26031661

    West, R. J., Lu, Y., Marie, B., Gao, F. B. and Sweeney, S. T. (2015). Rab8, POSH, and TAK1 regulate synaptic growth in a Drosophila model of frontotemporal dementia. J Cell Biol 208: 931-947. PubMed ID: 25800055

    Cheruiyot, A., Lee, J. A., Gao, F. B. and Ahmad, S. T. (2014). Expression of mutant CHMP2B, an ESCRT-III component involved in frontotemporal dementia, causes eye deformities due to Notch misregulation in Drosophila. FASEB J 28: 667-675. PubMed ID: 24158394

    Li, Z., Lu, Y., Xu, X. L. and Gao, F. B. (2013). The FTD/ALS-associated RNA-binding protein TDP-43 regulates the robustness of neuronal specification through microRNA-9a in Drosophila. Hum Mol Genet 22: 218-225. PubMed ID: 23042786

  • Hernan G Garcia Physical Biology of the Embryo, University of California at Berkeley
    Alamos, S., Reimer, A., Westrum, C., Turner, M. A., Talledo, P., Zhao, J., Luu, E. and Garcia, H. G. (2023). Minimal synthetic enhancers reveal control of the probability of transcriptional engagement and its timing by a morphogen gradient. Cell Syst. PubMed ID: 36696901

    Kim, Y. J., Rhee, K., Liu, J., Jeammet, S., Turner, M. A., Small, S. J. and Garcia, H. G. (2022). Predictive modeling reveals that higher-order cooperativity drives transcriptional repression in a synthetic developmental enhancer. Elife 11. PubMed ID: 36503705

    Zhao J., Perkins, M. L., Norstad, M. and Garcia, H. G. (2023). A bistable autoregulatory module in the developing embryo commits cells to binary expression fates. Curr Biol33(14):2851-2864 e2811. PubMed ID: 36696901

    Kim Y. J., Rhee, K., Liu, J., Jeammet, S., Turner, M. A., Small, S. J., Garcia, H. G. (2022). Predictive modeling reveals that higher-order cooperativity drives transcriptional repression in a synthetic developmental enhancer. Elife11. PubMed ID: 34100718

  • Guanjun Gao School of Life Sciences, Tsinghua University, Beijing
    Zhang, X., Wu, X., Peng, J., Sun, A., Guo, Y., Fu, P. and Gao, G. (2022). Cis- and trans-regulation by histone H4 basic patch R17/R19 in metazoan development. Open Biol 12(11): 220066. PubMed ID: 36382370

    Wu, M., Zhang, X., Wei, W., Long, L., An, S. and Gao, G. (2020). CRISPR/Cas9 mediated genetic resource for unknown kinase and phosphatase genes in Drosophila. Sci Rep 10(1): 7383. PubMed ID: 32355295

    Zhang, W., Zhang, X., Xue, Z., Li, Y., Ma, Q., Ren, X., Zhang, J., Yang, S., Yang, L., Wu, M., Ren, M., Xi, R., Wu, Z., Liu, J. L., Matunis, E., Dai, J. and Gao, G. (2018). Probing the function of metazoan histones with a systematic library of H3 and H4 mutants. Dev Cell. PubMed ID: 30595536

    Wen, K., Yang, L., Xiong, T., Di, C., Ma, D., Wu, M., Xue, Z., Zhang, X., Long, L., Zhang, W., Zhang, J., Bi, X., Dai, J., Zhang, Q., Lu, Z. J. and Gao, G. (2016). Critical roles of long noncoding RNAs in Drosophila spermatogenesis. Genome Res 26: 1233-1244. PubMed ID: 27516619

    Xue, Z., Wu, M., Wen, K., Ren, M., Long, L., Zhang, X. and Gao, G. (2014). CRISPR/Cas9 Mediates Efficient Conditional Mutagenesis in Drosophila. G3 (Bethesda). PubMed ID: 25193494

    Gao, G., Chen, L. and Huang, C. (2014). Anti-cancer Drug Discovery: Update and Comparisons in Yeast, Drosophila, and Zebrafish. Curr Mol Pharmacol. PubMed ID: 24993385

    Xue, Z., Ren, M., Wu, M., Dai, J., Rong, Y. S. and Gao, G. (2014). Efficient gene knock-out and knock-in with transgenic Cas9 in Drosophila. G3 (Bethesda) 4: 925-929. PubMed ID: 24657904

    >Gao, G., Vibranovski, M. D., Zhang, L., Li, Z., Liu, M., Zhang, Y. E., Li, X., Zhang, W., Fan, Q., VanKuren, N. W., Long, M. and Wei, L. (2014). A long-term demasculinization of X-linked intergenic noncoding RNAs in Drosophila melanogaster. Genome Res 24: 629-638. PubMed ID: 24407956

    Yu, Z., Ren, M., Wang, Z., Zhang, B., Rong, Y. S., Jiao, R. and Gao, G. (2013). Highly efficient genome modifications mediated by CRISPR/Cas9 in Drosophila. Genetics 195: 289-291. PubMed ID: 23833182

  • Quentin Gaudry University of Maryland, College Park
    Schenk, J. E. and Gaudry, Q. (2023). Nonspiking Interneurons in the Drosophila Antennal Lobe Exhibit Spatially Restricted Activity. eNeuro 10(1). PubMed ID: 36650069

    Suzuki, Y., Schenk, J. E., Tan, H. and Gaudry, Q. (2020). A Population of Interneurons Signals Changes in the Basal Concentration of Serotonin and Mediates Gain Control in the Drosophila Antennal Lobe. Curr Biol. PubMed ID: 32142699

    Zhang, X., Coates, K., Dacks, A., Gunay, C., Lauritzen, J. S., Li, F., Calle-Schuler, S. A., Bock, D. and Gaudry, Q. (2019). Local synaptic inputs support opposing, network-specific odor representations in a widely projecting modulatory neuron. Elife 8. PubMed ID: 31264962

    Gaudry, Q. (2018). Serotonergic Modulation of Olfaction in Rodents and Insects. Yale J Biol Med 91(1): 23-32. PubMed ID: 29599654

    Zhang, X. and Gaudry, Q. (2018). Examining Monosynaptic Connections in Drosophila Using Tetrodotoxin Resistant Sodium Channels. J Vis Exp(132). PubMed ID: 29553525

  • Luis Garcia-Alonso Developmental Neurobiology, The Instituto de Neurociencias de Alicante, Spain
    Velasquez, E., Gomez-Sanchez, J. A., Donier, E., Grijota-Martinez, C., Cabedo, H. and Garcia-Alonso, L. (2022). Fasciclin 2 engages EGFR in an auto-stimulatory loop to promote imaginal disc cell proliferation in Drosophila. PLoS Genet 18(6): e1010224. PubMed ID: 35666718

    Donier, E., Gomez-Sanchez, J. A., Grijota-Martinez, C., Lakoma, J., Baars, S., Garcia-Alonso, L. and Cabedo, H. (2012). L1CAM binds ErbB receptors through Ig-like domains coupling cell adhesion and neuregulin signalling. PLoS One 7(7): e40674. PubMed ID: 22815787

    Nagaraj, K., Kristiansen, L. V., Skrzynski, A., Castiella, C., Garcia-Alonso, L. and Hortsch, M. (2009). Pathogenic human L1-CAM mutations reduce the adhesion-dependent activation of EGFR. Hum Mol Genet 18(20): 3822-3831. PubMed ID: 19617634

  • Rafael Garesse Instituto de Investigaciones Biomedicas "Alberto Sols", Madrid
    Fernandez-Moreno, M. A., Hernandez, R., Adan, C., Roberti, M., Bruni, F., Polosa, P. L., Cantatore, P., Matsushima, Y., Kaguni, L. S. and Garesse, R. (2013). Drosophila nuclear factor DREF regulates the expression of the mitochondrial DNA helicase and mitochondrial transcription factor B2 but not the mitochondrial translation factor B1. Biochim Biophys Acta. PubMed ID: 23916463

    Martinez-Azorin, F., Calleja, M., Hernandez-Sierra, R., Farr, C. L., Kaguni, L. S. and Garesse, R. (2013). Muscle-specific overexpression of the catalytic subunit of DNA polymerase gamma induces pupal lethality in Drosophila melanogaster. Arch Insect Biochem Physiol 83: 127-137. PubMed ID: 23729397

    Sanchez-Martinez, A., Calleja, M., Peralta, S., Matsushima, Y., Hernandez-Sierra, R., Whitworth, A. J., Kaguni, L. S. and Garesse, R. (2012). Modeling pathogenic mutations of human twinkle in Drosophila suggests an apoptosis role in response to mitochondrial defects. PLoS One 7: e43954. PubMed ID: 22952820

  • Daniel Garrigan Department of Biology, University of Rochester
    Vedanayagam, J. P. and Garrigan, D. (2015). The effects of natural selection across molecular pathways in Drosophila melanogaster. BMC Evol Biol 15: 203. PubMed ID: 26391223

    Brand, C. L., Kingan, S. B., Wu, L. and Garrigan, D. (2013). A selective sweep across species boundaries in Drosophila. Mol Biol Evol. PubMed ID: 23827876

    Garrigan, D., Kingan, S. B., Geneva, A. J., Andolfatto, P., Clark, A. G., Thornton, K. R. and Presgraves, D. C. (2012). Genome sequencing reveals complex speciation in the Drosophila simulans clade. Genome Res 22: 1499-1511. PubMed ID: 22534282

    Kingan, S. B., Garrigan, D. and Hartl, D. L. (2010). Recurrent selection on the Winters sex-ratio genes in Drosophila simulans. Genetics 184: 253-265. PubMed ID: 19897749

  • Paul Garrity Brandeis University
    Laursen, W. J., Busby, R., Sarkissian, T., Chang, E. C. and Garrity, P. A. (2023). DMKPs provide a generalizable strategy for studying genes required for reproduction or viability in non-traditional model organisms. Genetics. PubMed ID: 37036394

    Marin, E. C., Buld, L., Theiss, M., Sarkissian, T., Roberts, R. J. V., Turnbull, R., Tamimi, I. F. M., Pleijzier, M. W., Laursen, W. J., Drummond, N., Schlegel, P., Bates, A. S., Li, F., Landgraf, M., Costa, M., Bock, D. D., Garrity, P. A. and Jefferis, G. (2020). Connectomics Analysis Reveals First-, Second-, and Third-Order Thermosensory and Hygrosensory Neurons in the Adult Drosophila Brain. Curr Biol. PubMed ID: 32619476

    Greppi, C., Laursen, W. J., Budelli, G., Chang, E. C., Daniels, A. M., van Giesen, L., Smidler, A. L., Catteruccia, F. and Garrity, P. A. (2020). Mosquito heat seeking is driven by an ancestral cooling receptor. Science 367(6478): 681-684. PubMed ID: 32029627

    Budelli, G., Ni, L., Berciu, C., van Giesen, L., Knecht, Z. A., Chang, E. C., Kaminski, B., Silbering, A. F., Samuel, A., Klein, M., Benton, R., Nicastro, D. and Garrity, P. A. (2019). Ionotropic Receptors Specify the Morphogenesis of Phasic Sensors Controlling Rapid Thermal Preference in Drosophila. Neuron. PubMed ID: 30654923

    Knecht, Z. A., Silbering, A. F., Cruz, J., Yang, L., Croset, V., Benton, R. and Garrity, P. A. (2017). Ionotropic Receptor-dependent moist and dry cells control hygrosensation in Drosophila. Elife 6. PubMed ID: 28621663

    Knecht, Z. A., Silbering, A. F., Ni, L., Klein, M., Budelli, G., Bell, R., Abuin, L., Ferrer, A. J., Samuel, A. D., Benton, R. and Garrity, P. A. (2016). Distinct combinations of variant ionotropic glutamate receptors mediate thermosensation and hygrosensation in Drosophila. Elife 5. PubMed ID: 27656904

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

    Barbagallo, B. and Garrity, P. A. (2015). Temperature sensation in Drosophila. Curr Opin Neurobiol 34C: 8-13. PubMed ID: 25616212

    Klein, M., Afonso, B., Vonner, A. J., Hernandez-Nunez, L., Berck, M., Tabone, C. J., Kane, E. A., Pieribone, V. A., Nitabach, M. N., Cardona, A., Zlatic, M., Sprecher, S. G., Gershow, M., Garrity, P. A. and Samuel, A. D. (2014). Sensory determinants of behavioral dynamics in Drosophila thermotaxis. Proc Natl Acad Sci U S A. PubMed ID: 25550513

    Ni, L., Bronk, P., Chang, E. C., Lowell, A. M., Flam, J. O., Panzano, V. C., Theobald, D. L., Griffith, L. C. and Garrity, P. A. (2013). A gustatory receptor paralogue controls rapid warmth avoidance in Drosophila. Nature. PubMed ID: 23925112

  • Maurizio Gatti Istituto di Biologia e Patologia Molecolari, Rome
    Wan, B., Poirie, M. and Gatti, J. L. (2020). Parasitoid wasp venom vesicles (venosomes) enter Drosophila melanogaster lamellocytes through a flotillin/lipid raft-dependent endocytic pathway. Virulence 11(1): 1512-1521. PubMed ID: 33135553

    Maccallini, P., Bavasso, F., Scatolini, L., Bucciarelli, E., Noviello, G., Lisi, V., Palumbo, V., D'Angeli, S., Cacchione, S., Cenci, G., Ciapponi, L., Wakefield, J. G., Gatti, M. and Raffa, G. D. (2020). Intimate functional interactions between TGS1 and the Smn complex revealed by an analysis of the Drosophila eye development. PLoS Genet 16(5): e1008815. PubMed ID: 32453722

    Pavlova, G. A., Popova, J. V., Andreyeva, E. N., Yarinich, L. A., Lebedev, M. O., Razuvaeva, A. V., Dubatolova, T. D., Oshchepkova, A. L., Pellacani, C., Somma, M. P., Pindyurin, A. V. and Gatti, M. (2019). RNAi-mediated depletion of the NSL complex subunits leads to abnormal chromosome segregation and defective centrosome duplication in Drosophila mitosis. PLoS Genet 15(9): e1008371. PubMed ID: 31527906

    Pavlova, G. A., Razuvaeva, A. V., Popova, J. V., Andreyeva, E. N., Yarinich, L. A., Lebedev, M. O., Pellacani, C., Bonaccorsi, S., Somma, M. P., Gatti, M. and Pindyurin, A. V. (2019). The role of Patronin in Drosophila mitosis. BMC Mol Cell Biol 20(Suppl 1): 7. PubMed ID: 31284878

    Pellacani, C., Bucciarelli, E., Renda, F., Hayward, D., Palena, A., Chen, J., Bonaccorsi, S., Wakefield, J. G., Gatti, M. and Somma, M. P. (2018). Splicing factors Sf3A2 and Prp31 have direct roles in mitotic chromosome segregation. Elife 7. PubMed ID: 30475206

    Renda, F., Pellacani, C., Strunov, A., Bucciarelli, E., Naim, V., Bosso, G., Kiseleva, E., Bonaccorsi, S., Sharp, D. J., Khodjakov, A., Gatti, M. and Somma, M. P. (2017). The Drosophila orthologue of the INT6 onco-protein regulates mitotic microtubule growth and kinetochore structure. PLoS Genet 13(5): e1006784. PubMed ID: 28505193

    Merigliano, C., Marzio, A., Renda, F., Somma, M.P., Gatti, M. and Verni, F. (2016). A role for the Twins protein phosphatase (PP2A-B55) in the maintenance of Drosophila genome integrity. Genetics [Epub ahead of print]. PubMed ID: 28040742

    Cicconi, A., Micheli, E., Verni, F., Jackson, A., Gradilla, A. C., Cipressa, F., Raimondo, D., Bosso, G., Wakefield, J. G., Ciapponi, L., Cenci, G., Gatti, M., Cacchione, S. and Raffa, G. D. (2016). The Drosophila telomere-capping protein Verrocchio binds single-stranded DNA and protects telomeres from DNA damage response. Nucleic Acids Res. PubMed ID: 27940556

    Marzullo, M. and Gatti, M. (2016). Telomere fusion in Drosophila: The role of subtelomeric chromatin. Fly (Austin): [Epub ahead of print] PubMed ID: 26786804

    Marzullo, M. and Gatti, M. (2016). Telomere fusion in Drosophila: The role of subtelomeric chromatin. Fly (Austin) [Epub ahead of print]. PubMed ID: 26786804

  • Urlike Gaul Department of Biochemistry, Gene Center of the LMU, Munich
    Punzi, G., Ursini, G., Chen, Q., Radulescu, E., Tao, R., Huu Qi, Z., Jung, C., Bandilla, P., Ludwig, C., Heron, M., Sophie Kiesel, A., Museridze, M., Philippou-Massier, J., Nikolov, M., Renna Max Schnepf, A., Unnerstall, U., Ceolin, S., Mühlig, B., Gompel, N., Soeding, J. and Gaul, U. (2022). Large-scale analysis of Drosophila core promoter function using synthetic promoters. Mol Syst Biol 18(2): e9816. PubMed ID: 35156763

    Li, X., Fetter, R., Schwabe, T., Jung, C., Liu, L., Steller, H. and Gaul, U. (2021). The cAMP effector PKA mediates Moody GPCR signaling in Drosophila blood-brain barrier formation and maturation. Elife 10. PubMed ID: 34382936

    Schnepf, M., von Reutern, M., Ludwig, C., Jung, C. and Gaul, U. (2020). Transcription Factor Binding Affinities and DNA Shape Readout. iScience 23(11): 101694. PubMed ID: 33163946

    Ceolin, S., Hanf, M., Bozek, M., Storti, A. E., Gompel, N., Unnerstall, U., Jung, C. and Gaul, U. (2020). A sensitive mNeonGreen reporter system to measure transcriptional dynamics in Drosophila development. Commun Biol 3(1): 663. PubMed ID: 33184447

    Bozek, M., Cortini, R., Storti, A. E., Unnerstall, U., Gaul, U. and Gompel, N. (2019). ATAC-seq reveals regional differences in enhancer accessibility during the establishment of spatial coordinates in the Drosophila blastoderm. Genome Res. PubMed ID: 30962180

    Schwabe, T., Li, X. and Gaul, U. (2017). Dynamic analysis of the mesenchymal-epithelial transition of blood-brain barrier forming glia in Drosophila. Biol Open. PubMed ID: 28108476

    Deligiannaki, M., Casper, A. L., Jung, C. and Gaul, U. (2015). Pasiflora proteins are novel core components of the septate junction. Development 142: 3046-3057. PubMed ID: 26329602

    Bottcher, R., Hollmann, M., Merk, K., Nitschko, V., Obermaier, C., Philippou-Massier, J., Wieland, I., Gaul, U. and Forstemann, K. (2014). Efficient chromosomal gene modification with CRISPR/cas9 and PCR-based homologous recombination donors in cultured Drosophila cells. Nucleic Acids Res 42: e89. PubMed ID: 24748663

    Schroeder, M. D., Greer, C. and Gaul, U. (2011). How to make stripes: deciphering the transition from non-periodic to periodic patterns in Drosophila segmentation. Development 138: 3067-3078. PubMed ID: 21693522

  • Elizabeth Gavis Department of Molecular Biology, Princeton University
    Hakes, A. C. and Gavis, E. R. (2023). Plasticity of Drosophila germ granules during germ cell development. PLoS Biol 21(4): e3002069. PubMed ID: 37053289

    Siddiqui, N. U., Karaiskakis, A., Goldman, A. L., Eagle, W. V. I., Smibert, C. A., Gavis, E. R. and Lipshitz, H. D. (2023). Smaug regulates germ plasm synthesis and primordial germ cell number in Drosophila embryos by repressing the oskar and bruno 1 mRNAs. bioRxiv. PubMed ID: 36909513

    Breznak, S. M., Peng, Y., Deng, L., Kotb, N. M., Flamholz, Z., Rapisarda, I. T., Martin, E. T., LaBarge, K. A., Fabris, D., Gavis, E. R. and Rangan, P. (2023). H/ACA snRNP-dependent ribosome biogenesis regulates translation of polyglutamine proteins. Sci Adv 9(25): eade5492. PubMed ID: 37343092

    Eichler, C. E., Li, H., Grunberg, M. E. and Gavis, E. R. (2023). Localization of oskar mRNA by agglomeration in ribonucleoprotein granules. PLoS Genet 19(8): e1010877. PubMed ID: 37624861

    Li, H. and Gavis, E. R. (2022). Drosophila FMRP controls miR-276-mediated regulation of nejire mRNA for space-filling dendrite development. G3 (Bethesda) 12(11). PubMed ID: 36102801

    Li, H. and Gavis, E. R. (2022). The Drosophila fragile X mental retardation protein modulates the neuronal cytoskeleton to limit dendritic arborization. Development 149(10). PubMed ID: 35502752

    Peng, Y. and Gavis, E. R. (2022). The Drosophila hnRNP F/H homolog Glorund recruits dFMRP to inhibit nanos translation elongation. Nucleic Acids Res. PubMed ID: 35699205

    Valentino, M., Ortega, B. M., Ulrich, B., Doyle, D. A., Farnum, E. D., Joiner, D. A., Gavis, E. R. and Niepielko, M. G. (2022). Computational modeling offers new insight into Drosophila germ granule development. Biophys J 121(8): 1465-1482. PubMed ID: 35288123

    Li, H. and Gavis, E. R. (2022). Drosophila FMRP controls miR-276-mediated regulation of nejire mRNA for space filling dendrite development. G3 (Bethesda). PubMed ID: 36102801

    Valentino, M., Ortega, B. M., Ulrich, B., Doyle, D. A., Farnum, E. D., Joiner, D. A., Gavis, E. R. and Niepielko, M. G. (2022). Computational modeling offers new insight into Drosophila germ granule development. Biophys J. PubMed ID: 35288123Doherty, C. A., Diegmiller, R., Kapasiawala, M., Gavis, E. R. and Shvartsman, S. Y. (2021). Coupled oscillators coordinate collective germline growth. Dev Cell. PubMed ID: 33689691

  • Nick Gay Department of Biochemistry, University of Cambridge
    Lewis, M., Arnot, C. J., Beeston, H., McCoy, A., Ashcroft, A. E., Gay, N. J. and Gangloff, M. (2013). Cytokine Spatzle binds to the Drosophila immunoreceptor Toll with a neurotrophin-like specificity and couples receptor activation. Proc Natl Acad Sci U S A. PubMed ID: 24282309

    McIlroy, G., Foldi, I., Aurikko, J., Wentzell, J. S., Lim, M. A., Fenton, J. C., Gay, N. J. and Hidalgo, A. (2013). Toll-6 and Toll-7 function as neurotrophin receptors in the Drosophila melanogaster CNS. Nat Neurosci 16: 1248-1256. PubMed ID: 23892553

    Arnot, C. J., Gay, N. J. and Gangloff, M. (2010). Molecular mechanism that induces activation of Spatzle, the ligand for the Drosophila Toll receptor. J Biol Chem 285: 19502-19509. PubMed ID: 20378549

  • Wanzhong Ge Department Women's Hospital, School of Medicine, Zhejiang University
    Guo T., Miao, C., Liu, Z., Duan, J., Ma, Y., Zhang, X., Yang, W., Xue, M., Deng, Q., Guo, P., Xi, Y., Yang, X., Huang, X., Ge, W. (2023). Impaired dNKAP function drives genome instability and tumorigenic growth in Drosophila epithelia. J Mol Cell Biol. PubMed ID: 37543696

    Fan W., Jin, X., Xu, M., Xi, Y., Lu, W., Yang, X., Guan, M. X., Ge, W. (2021). FARS2 deficiency in Drosophila reveals the developmental delay and seizure manifested by aberrant mitochondrial tRNA metabolism. Nucleic Acids Res49(22):13108-13121. PubMed ID: 32913227

  • Fatima Gebauer Centre for Genomic Regulation, Barcelona
    Nadimpalli, H. P., Guitart, T., Coll, O. and Gebauer, F. (2022). Ataxin-2, Twenty-four, and Dicer-2 are components of a noncanonical cytoplasmic polyadenylation complex. Life Sci Alliance 5(12). PubMed ID: 36114004

    Szostak, E., Garcia-Beyaert, M., Guitart, T., Graindorge, A., Coll, O. and Gebauer, F. (2018). Hrp48 and eIF3d contribute to msl-2 mRNA translational repression. Nucleic Acids Res. PubMed ID: 29635389

    Coll, O., Guitart, T., Villalba, A., Papin, C., Simonelig, M. and Gebauer, F. (2018). Dicer-2 promotes mRNA activation through cytoplasmic polyadenylation. Rna. PubMed ID: 29317541

    Moschall, R., Strauss, D., Garcia-Bayert, M., Gebauer, F. and Medenbach, J. (2017). Drosophila Sister of Sex-lethal is a repressor of translation. RNA. PubMed ID: 29089381

    Hennig, J., Militti, C., Popowicz, G. M., Wang, I., Sonntag, M., Geerlof, A., Gabel, F., Gebauer, F., Sattler, M. (2014) Structural basis for the assembly of the Sxl-Unr translation regulatory complex. Nature. PubMed ID: 25209665

    Militti, C., Maenner, S., Becker, P. B., Gebauer, F. (2014) UNR facilitates the interaction of MLE with the lncRNA roX2 during Drosophila dosage compensation. Nat Commun 5: 4762. PubMed ID: 25158899

    Coll, O., Villalba, A. and Gebauer, F. (2014). Cytoplasmic polyadenylation assays. Methods Mol Biol 1125: 53-63. PubMed ID: 24590779

    Graindorge, A., Carre, C. and Gebauer, F. (2013). Sex-lethal promotes nuclear retention of msl2 mRNA via interactions with the STAR protein HOW. Genes Dev 27: 1421-1433. PubMed ID: 23788626

    Coll, O., Villalba, A., Bussotti, G., Notredame, C. and Gebauer, F. (2010). A novel, noncanonical mechanism of cytoplasmic polyadenylation operates in Drosophila embryogenesis. Genes Dev 24: 129-134. PubMed ID: 20080951

  • Brian Gebelein Developmental Biology, University of Cincinnati
    Gagliani, E. K., Gutzwiller, L. M., Kuang, Y., Odaka, Y., Hoffmeister, P., Hauff, S., Turkiewicz, A., Harding-Theobald, E., Dolph, P. J., Borggrefe, T., Oswald, F., Gebelein, B. and Kovall, R. A. (2022). A Drosophila Su(H) model of Adams-Oliver Syndrome reveals cofactor titration as a mechanism underlying developmental defects. PLoS Genet 18(8): e1010335. PubMed ID: 35951645

    Kuang, Y., Pyo, A., Eafergan, N., Cain, B., Gutzwiller, L. M., Axelrod, O., Gagliani, E. K., Weirauch, M. T., Kopan, R., Kovall, R. A., Sprinzak, D. and Gebelein, B. (2021). Enhancers with cooperative Notch binding sites are more resistant to regulation by the Hairless co-repressor. PLoS Genet 17(9): e1009039. PubMed ID: 34559800

    Salomone, J., Qin, S., Fufa, T. D., Cain, B., Farrow, E., Guan, B., Hufnagel, R. B., Nakafuku, M., Lim, H. W., Campbell, K. and Gebelein, B. (2021). Conserved Gsx2/Ind homeodomain monomer versus homodimer DNA binding defines regulatory outcomes in flies and mice. Genes Dev 35(1-2): 157-174. PubMed ID: 33334823

    Kuang, Y., Golan, O., Preusse, K., Cain, B., Christensen, C. J., Salomone, J., Campbell, I., Okwubido-Williams, F. V., Hass, M. R., Yuan, Z., Eafergan, N., Moberg, K. H., Kovall, R. A., Kopan, R., Sprinzak, D. and Gebelein, B. (2020). Enhancer architecture sensitizes cell-specific responses to Notch gene dose via a bind and discard mechanism. Elife 9. PubMed ID: 32297857

    Zandvakili, A., Uhl, J. D., Campbell, I., Salomone, J., Song, Y. C. and Gebelein, B. (2018). The cis-regulatory logic underlying abdominal Hox-mediated repression versus activation of regulatory elements in Drosophila. Dev Biol. PubMed ID: 30468713

    Zandvakili, A., Campbell, I., Gutzwiller, L. M., Weirauch, M. T. and Gebelein, B. (2018). Degenerate Pax2 and Senseless binding motifs improve detection of low-affinity sites required for enhancer specificity. PLoS Genet 14(4): e1007289. PubMed ID: 29617378

    Wang, G., Gutzwiller, L., Li-Kroeger, D. and Gebelein, B. (2017). A Hox complex activates and potentiates the Epidermal Growth Factor signaling pathway to specify Drosophila oenocytes. PLoS Genet 13(7): e1006910. PubMed ID: 28715417

    Gresser, A. L., Gutzwiller, L. M., Gauck, M. K., Hartenstein, V., Cook, T. A. and Gebelein, B. (2015). Rhomboid enhancer activity defines a subset of Drosophila neural precursors required for proper feeding, growth and viability. PLoS One 10: e0134915. PubMed ID: 26252385

    Jukam, D., Xie, B., Rister, J., Terrell, D., Charlton-Perkins, M., Pistillo, D., Gebelein, B., Desplan, C. and Cook, T. (2013). Opposite Feedbacks in the Hippo Pathway for Growth Control and Neural Fate. Science. PubMed ID: 23989952

    Bryantsev, A. L., Duong, S., Brunetti, T. M., Chechenova, M. B., Lovato, T. L., Nelson, C., Shaw, E., Uhl, J. D., Gebelein, B. and Cripps, R. M. (2012). Extradenticle and homothorax control adult muscle fiber identity in Drosophila. Dev Cell 23: 664-673. PubMed ID: 22975331

  • Walter Gehring Division of Cell Biology, Biozentrum, University of Basel
    Papadopoulos, D. K., Skouloudaki, K., Adachi, Y., Samakovlis, C. and Gehring, W. J. (2012). Dimer formation via the homeodomain is required for function and specificity of Sex combs reduced in Drosophila. Dev Biol 367: 78-89. PubMed ID: 22564794

    Blanco, J., Pauli, T., Seimiya, M., Udolph, G. and Gehring, W. J. (2010). Genetic interactions of eyes absent, twin of eyeless and orthodenticle regulate sine oculis expression during ocellar development in Drosophila. Dev Biol 344: 1088-1099. PubMed ID: 20580700

    Grieder, N. C., Morata, G., Affolter, M. and Gehring, W. J. (2009). Spalt major controls the development of the notum and of wing hinge primordia of the Drosophila melanogaster wing imaginal disc. Dev Biol 329: 315-326. PubMed ID: 19298807

  • Erika Geisbrecht School of Biological Sciences University of Missouri-Kansas City
    Guo, Y., Zeng, Q., Brooks, D. and Geisbrecht, E. R. (2023). A conserved STRIPAK complex is required for autophagy in muscle tissue. Mol Biol Cell 34(9): ar91. PubMed ID: 37379167

    Kawakami, J., Brooks, D., Zalmai, R., Hartson, S. D., Bouyain, S. and Geisbrecht, E. R. (2022). Complex protein interactions mediate Drosophila Lar function in muscle tissue. PLoS One 17(5): e0269037. PubMed ID: 35622884

    Brooks, D., Bawa, S., Bontrager, A., Stetsiv, M., Guo, Y. and Geisbrecht, E. R. (2022). Independent pathways control muscle tissue size and sarcomere remodeling. Dev Biol 490: 1-12. PubMed ID: 35760368 Bawa, S., Piccirillo, R. and Geisbrecht, E. R. (2021). TRIM32: A Multifunctional Protein Involved in Muscle Homeostasis, Glucose Metabolism, and Tumorigenesis. Biomolecules 11(3). PubMed ID: 33802079

    Brooks, D., Naeem, F., Stetsiv, M., Goetting, S. C., Bawa, S., Green, N., Clark, C., Bashirullah, A. and Geisbrecht, E. R. (2020). Drosophila NUAK functions with Starvin/BAG3 in autophagic protein turnover. PLoS Genet 16(4): e1008700. PubMed ID: 32320396

    Green, N., Walker, J., Bontrager, A., Zych, M. and Geisbrecht, E. R. (2018). A tissue communication network coordinating innate immune response during muscle stress. J Cell Sci. PubMed ID: 30478194

    Vishal, K., Bawa, S., Brooks, D., Bauman, K. and Geisbrecht, E. R. (2018). Thin is required for cell death in the Drosophila abdominal muscles by targeting DIAP1. Cell Death Dis 9(7): 740. PubMed ID: 29970915

    Vishal, K., Brooks, D. S., Bawa, S., Gameros, S., Stetsiv, M. and Geisbrecht, E. R. (2017). Adult Muscle Formation Requires Drosophila Moleskin for Proliferation of Wing Disc-Associated Muscle Precursors. Genetics. PubMed ID: 28249984

    Green, N., Odell, N., Zych, M., Clark, C., Wang, Z. H., Biersmith, B., Bajzek, C., Cook, K. R., Dushay, M. S. and Geisbrecht, E. (2016). A Common suite of coagulation proteins function in Drosophila muscle attachment. Genetics [Epub ahead of print]. PubMed ID: 27585844

    Wang, Z. H., Clark, C. and Geisbrecht, E. R. (2016). Drosophila clueless is involved in Parkin-dependent mitophagy by promoting VCP-mediated Marf degradation. Hum Mol Genet [Epub ahead of print]. PubMed ID: 26931463

    Wang, Z. H., Rabouille, C. and Geisbrecht, E. R. (2015). Loss of a Clueless-dGRASP complex results in ER stress and blocks Integrin exit from the perinuclear endoplasmic reticulum in Drosophila larval muscle. Biol Open. PubMed ID: 25862246

  • Bill Gelbart Molecular and Cellular Biology, Harvard University
    Matthews, B. B., Dos Santos, G., Crosby, M. A., Emmert, D. B., St Pierre, S. E., Gramates, L. S., Zhou, P., Schroeder, A. J., Falls, K., Russo, S. M. and Gelbart, W. M. (2015). Gene model annotations for Drosophila melanogaster: Impact of high-throughput data. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26109357

    Matthews, B. B., Dos Santos, G., Crosby, M. A., Emmert, D. B., St Pierre, S. E., Gramates, L. S., Zhou, P., Schroeder, A. J., Falls, K., Russo, S. M. and Gelbart, W. M. (2015). Gene model annotations for Drosophila melanogaster: Impact of high-throughput data. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26109357

    Fang, R., et al. (2012). Automatic categorization of diverse experimental information in the bioscience literature. BMC Bioinformatics 13: 16. PubMed ID: 22280404

  • Vladimir Gelfand Department of Cell and Molecular Biology, Northwestern University School of Medicine, Chicago
    Lu, W., Lakonishok, M. and Gelfand, V. I. (2023). Drosophila oocyte specification is maintained by the dynamic duo of microtubule polymerase Mini spindles/XMAP215 and dynein. bioRxiv. PubMed ID: 36945460

    Lu, W., Lakonishok, M., Gelfand, V. I. (2023). The dynamic duo of microtubule polymerase Mini spindles/XMAP215 and cytoplasmic dynein is essential for maintaining Drosophila oocyte fate. Proc Natl Acad Sci U S A, 120(39):e2303376120 PubMed ID: 37722034

    Lu, W., Lakonishok, M., Serpinskaya, A. S. and Gelfand, V. I. (2022). A novel mechanism of bulk cytoplasmic transport by cortical dynein in Drosophila ovary. Elife 11. PubMed ID: 35170428

    Del Castillo, U., Norkett, R., Lu, W., Serpinskaya, A. and Gelfand, V. I. (2022). Ataxin-2 is essential for cytoskeletal dynamics and neurodevelopment in Drosophila. iScience 25(1): 103536. PubMed ID: 34977501

    Del Castillo, U., Muller, H. J. and Gelfand, V. I. (2020). Kinetochore protein Spindly controls microtubule polarity in Drosophila axons. Proc Natl Acad Sci U S A 117(22): 12155-12163. PubMed ID: 32430325

    Norkett, R., Del Castillo, U., Lu, W. and Gelfand, V. I. (2020). Ser/Thr kinase Trc controls neurite outgrowth in Drosophila by modulating microtubule-microtubule sliding. Elife 9. PubMed ID: 32022690

    Lu, W., Lakonishok, M., Liu, R., Billington, N., Rich, A., Glotzer, M., Sellers, J. R. and Gelfand, V. I. (2020). Competition between kinesin-1 and myosin-V defines Drosophila posterior determination. Elife 9. PubMed ID: 32057294

    Lu, W., Lakonishok, M., Serpinskaya, A. S., Kirchenbuechler, D., Ling, S. C. and Gelfand, V. I. (2018). Ooplasmic flow cooperates with transport and anchorage in Drosophila oocyte posterior determination. J Cell Biol. PubMed ID: 30037924

    Lu, W., Winding, M., Lakonishok, M., Wildonger, J. and Gelfand, V.I. (2016). Microtubule-microtubule sliding by kinesin-1 is essential for normal cytoplasmic streaming in Drosophila oocytes. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27512034

    Winding, M., Kelliher, M.T., Lu, W., Wildonger, J. and Gelfand, V.I. (2016). Role of kinesin-1-based microtubule sliding in Drosophila nervous system development. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 27512046

    Jolly, A. L., Luan, C. H., Dusel, B. E., Dunne, S. F., Winding, M., Dixit, V. J., Robins, C., Saluk, J. L., Logan, D. J., Carpenter, A. E., Sharma, M., Dean, D., Cohen, A. R. and Gelfand, V. I. (2016). A Genome-wide RNAi Screen for Microtubule Bundle Formation and Lysosome Motility Regulation in Drosophila S2 Cells. Cell Rep 14: 611-620. PubMed ID: 26774481

    Del Castillo, U., Winding, M., Lu, W. and Gelfand, V.I. (2015). Interplay between kinesin-1 and cortical dynein during axonal outgrowth and microtubule organization in Drosophila neurons. Elife 4 [Epub ahead of print]. PubMed ID: 26615019

  • Pavel Georgiev Institute of Gene Biology, Russian Academy of Sciences, Moscow
    Kyrchanova, O., Sokolov, V., Tikhonov, M., Manukyan, G., Schedl, P. and Georgiev, P. (2023). Transcriptional Readthrough Interrupts Boundary Function in Drosophila. Int J Mol Sci 24(14). PubMed ID: 37511131

    Kyrchanova, O. V., Postika, N. Y., Sokolov, V. V. and Georgiev, P. G. (2022). Fragments of the Fab-3 and Fab-4 Boundaries of the Drosophila melanogaster Bithorax Complex That Include CTCF Sites Are not Effective Insulators. Dokl Biochem Biophys 502(1): 21-24. PubMed ID: 35275301

    Kyrchanova, O., Klimenko, N., Postika, N., Bonchuk, A., Zolotarev, N., Maksimenko, O. and Georgiev, P. (2021). Drosophila architectural protein CTCF is not essential for fly survival and is able to function independently of CP190. Biochim Biophys Acta Gene Regul Mech 1864(10): 194733. PubMed ID: 34311130

    Melnikova, L. S., Molodina, V. V., Kostyuchenko, M. V., Georgiev, P. G. and Golovnin, A. K. (2021). The BEAF-32 Protein Directly Interacts with Z4/putzig and Chriz/Chromator Proteins in Drosophila melanogaster. Dokl Biochem Biophys 498(1): 184-189. PubMed ID: 34189647

    Sabirov, M., Kyrchanova, O., Pokholkova, G. V., Bonchuk, A., Klimenko, N., Belova, E., Zhimulev, I. F., Maksimenko, O. and Georgiev, P. (2021). Mechanism and functional role of the interaction between CP190 and the architectural protein Pita in Drosophila melanogaster. Epigenetics Chromatin 14(1): 16. PubMed ID: 33752739

    Postika, N., Schedl, P., Georgiev, P. and Kyrchanova, O. (2021). Mapping of functional elements of the Fab-6 boundary involved in the regulation of the Abd-B hox gene in Drosophila melanogaster. Sci Rep 11(1): 4156. PubMed ID: 33603202

    Bonchuk, A., Boyko, K., Fedotova, A., Nikolaeva, A., Lushchekina, S., Khrustaleva, A., Popov, V. and Georgiev, P. (2021). Structural basis of diversity and homodimerization specificity of zinc-finger-associated domains in Drosophila. Nucleic Acids Res 49(4): 2375-2389. PubMed ID: 33638995

    Melnikova, L. S., Kostyuchenko, M. V., Georgiev, P. G. and Golovnin, A. K. (2020). The Chriz Protein Promotes the Recruitment of the Z4 Protein to the STAT-Dependent Promoters. Dokl Biochem Biophys 490(1): 29-33. PubMed ID: 32342309

    Bonchuk, A., Kamalyan, S., Mariasina, S., Boyko, K., Popov, V., Maksimenko, O. and Georgiev, P. (2020). N-terminal domain of the architectural protein CTCF has similar structural organization and ability to self-association in bilaterian organisms. Sci Rep 10(1): 2677. PubMed ID: 32060375

    Tikhonova, E., Fedotova, A., Bonchuk, A., Mogila, V., Larschan, E. N., Georgiev, P. and Maksimenko, O. (2019). The simultaneous interaction of MSL2 with CLAMP and DNA provides redundancy in the initiation of dosage compensation in Drosophila males. Development. PubMed ID: 31320325

  • Marios Georgiou School of Life Sciences, Queen's Medical Centre, Nottingham, U. K.
    Rusu, A. D., Cornhill, Z. E., Coutiño, B. C., Uribe, M. C., Lourdusamy, A., Markus, Z., May, S. T., Rahman, R. and Georgiou, M. (2021). CG7379 and ING1 suppress cancer cell invasion by maintaining cell-cell junction integrity. Open Biol 11(9): 210077. PubMed ID: 34493070

    Canales Coutino, B., Cornhill, Z. E., Couto, A., Mack, N. A., Rusu, A. D., Nagarajan, U., Fan, Y. N., Hadjicharalambous, M. R., Castellanos Uribe, M., Burrows, A., Lourdusamy, A., Rahman, R., May, S. T. and Georgiou, M. (2020). A Genetic Analysis of Tumor Progression in Drosophila Identifies the Cohesin Complex as a Suppressor of Individual and Collective Cell Invasion. iScience 23(6): 101237. PubMed ID: 32629605

    Couto, A., Mack, N. A., Favia, L. and Georgiou, M. (2017). An apicobasal gradient of Rac activity determines protrusion form and position. Nat Commun 8: 15385. PubMed ID: 28524861

    Mack, N. A. and Georgiou, M. (2014). The interdependence of the Rho GTPases and apicobasal cell polarity. Small GTPases 5(2): 10. PubMed ID: 25469537

  • Bertram Gerber Universität Leipzig Institut für Biologie, Genetik
    Konig, C. and Gerber, B. (2022). Age-related decrease in appetitive associative memory in fruit flies. J Exp Biol 225(21). PubMed ID: 36373856

    Schleyer, M., Weiglein, A., Thoener, J., Strauch, M., Hartenstein, V., Kantar Weigelt, M., Schuller, S., Saumweber, T., Eichler, K., Rohwedder, A., Merhof, D., Zlatic, M., Thum, A. S. and Gerber, B. (2020). Identification of Dopaminergic Neurons That Can Both Establish Associative Memory and Acutely Terminate Its Behavioral Expression. J Neurosci 40(31): 5990-6006. PubMed ID: 32586949

    Toshima, N., Kantar Weigelt, M., Weiglein, A., Boetzl, F. A. and Gerber, B. (2019). An amino-acid mixture can be both rewarding and punishing to larval Drosophila. J Exp Biol. PubMed ID: 31672727

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

    Konig, C., Khalili, A., Niewalda, T., Gao, S. and Gerber, B. (2019). An optogenetic analogue of second-order reinforcement in Drosophila. Biol Lett 15(7): 20190084. PubMed ID: 31266421

    Weiglein, A., Gerstner, F., Mancini, N., Schleyer, M. and Gerber, B. (2019). One-trial learning in larval Drosophila. Learn Mem 26(4): 109-120. PubMed ID: 30898973

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

    Chen, Y. C., Mishra, D., Glass, S. and Gerber, B. (2017). Behavioral evidence for enhanced processing of the minor component of binary odor mixtures in larval Drosophila. Front Psychol 8: 1923. PubMed ID: 29163299

    Croset, V., Schleyer, M., Arguello, J. R., Gerber, B. and Benton, R. (2016). A molecular and neuronal basis for amino acid sensing in the Drosophila larva. Sci Rep 6: 34871. PubMed ID: 27982028 Rohwedder, A., Wenz, N. L., Stehle, B., Huser, A., Yamagata, N., Zlatic, M., Truman, J. W., Tanimoto, H., Saumweber, T., Gerber, B. and Thum, A. S. (2016). Four individually identified paired dopamine neurons signal reward in larval Drosophila. Curr Biol. PubMed ID: 26877086

    Konig, C., Schleyer, M., Leibiger, J., El-Keredy, A. and Gerber, B. (2015). Bitter-Sweet Processing in Larval Drosophila. Chem Senses 40: 445. PubMed ID: 26078222

    Niewalda, T., Michels, B., Jungnickel, R., Diegelmann, S., Kleber, J., Kahne, T. and Gerber, B. (2015). Synapsin determines memory strength after punishment- and relief-learning. J Neurosci 35: 7487-7502. PubMed ID: 25972175

    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

  • J. Peter Gergen Department of Biochemistry and Cell Biology, Stony Brook University Stony Brook, NY
    Prazak, L., Iwasaki, Y., Kim, A. R., Kozlov, K., King, K. and Gergen, J. P. (2021). A dual role for DNA-binding by Runt in activation and repression of sloppy paired transcription. Mol Biol Cell: mbcE20080509. PubMed ID: 34432496

    Koromila, T., Gao, F., Iwasaki, Y., He, P., Pachter, L., Gergen, J. P. and Stathopoulos, A. (2020). Odd-paired is a pioneer-like factor that coordinates with Zelda to control gene expression in embryos. Elife 9. PubMed ID: 32701060

    Bell, K., Skier, K., Chen, K. H. and Gergen, J. P. (2019). Two pair-rule responsive enhancers regulate wingless transcription in the Drosophila blastoderm embryo. Dev Dyn. PubMed ID: 31837063

    Hang, S. and Gergen, J. P. (2017). Different modes of enhancer-specific regulation by Runt and Even-skipped during Drosophila segmentation. Mol Biol Cell. PubMed ID: 28077616

    Wang, X., Hang, S., Prazak, L. and Gergen, J. P. (2010). NELF potentiates gene transcription in the Drosophila embryo. PLoS One 5(7): e11498. PubMed ID: 20634899

    Funakoshi, Y., Negishi, Y., Gergen, J. P., Seino, J., Ishii, K., Lennarz, W. J., Matsuo, I., Ito, Y., Taniguchi, N. and Suzuki, T. (2010). Evidence for an essential deglycosylation-independent activity of PNGase in Drosophila melanogaster. PLoS One 5(5): e10545. PubMed ID: 20479940

  • Offer Gerlitz School of Medicine, IMRIC, Developmental Biology and Cancer Research, Hebrew University, Jerusalem
    Shore, T., Levi, T., Kalifa, R., Dreifuss, A., Rekler, D., Weinberg-Shukron, A., Nevo, Y., Bialistoky, T., Moyal, V., Gold, M. Y., Leebhoff, S., Zangen, D., Deshpande, G. and Gerlitz, O. (2022). Nucleoporin107 mediates female sexual differentiation via Dsx. Elife 11. PubMed ID: 35311642

    Brandwine, T., Ifrah, R., Bialistoky, T., Zaguri, R., Rhodes-Mordov, E., Mizrahi-Meissonnier, L., Sharon, D., Katanaev, V. L., Gerlitz, O. and Minke, B. (2021). Knockdown of Dehydrodolichyl Diphosphate Synthase in the Drosophila Retina Leads to a Unique Pattern of Retinal Degeneration. Front Mol Neurosci 14: 693967. PubMed ID: 34290587

    Levi, T., Sloutskin, A., Kalifa, R., Juven-Gershon, T. and Gerlitz, O. (2020). Efficient In Vivo Introduction of Point Mutations Using ssODN and a Co-CRISPR Approach. Biol Proced Online 22: 14. PubMed ID: 32684853

    Bialistoky, T., Manry, D., Smith, P., Ng, C., Kim, Y., Zamir, S., Moyal, V., Kalifa, R., Schedl, P., Gerlitz, O. and Deshpande, G. (2019). Functional analysis of Niemann-Pick disease type C family protein, NPC1a, in Drosophila melanogaster. Development 146(10). PubMed ID: 31092503

    Deshpande, G., Barr, J., Gerlitz, O., Lebedeva, L., Shidlovskii, Y. and Schedl, P. (2017). Cells on the move: Modulation of guidance cues during germ cell migration. Fly (Austin): [Epub ahead of print] PubMed ID: 28300473

    Deshpande, G., Manry, D., Jourjine, N., Mogila, V., Mozes, H., Bialistoky, T., Gerlitz, O. and Schedl, P. (2016). Functioning of an ABC transporter, Mdr49, in Hh signaling and germ cell migration. Development [Epub ahead of print]. PubMed ID: 27122170

    Weinberg-Shukron, A., Renbaum, P., Kalifa, R., Zeligson, S., Ben-Neriah, Z., Dreifuss, A., Abu-Rayyan, A., Maatuk, N., Fardian, N., Rekler, D., Kanaan, M., Samson, A. O., Levy-Lahad, E., Gerlitz, O. and Zangen, D. (2015). A mutation in the nucleoporin-107 gene causes XX gonadal dysgenesis. J Clin Invest 125: 4295-4304. PubMed ID: 26485283

    Ziv, O., Finkelstein, R., Suissa, Y., Dinur, T., Deshpande, G. and Gerlitz, O. (2012). Inverse regulation of target genes at the brink of the BMP morphogen activity gradient. J Cell Sci 125: 5811-5818. PubMed ID: 22956540

    Suissa, Y., Ordan, E., Deshpande, G. and Gerlitz, O. (2011). Males and females: creating differences while maintaining the similarities. Fly (Austin) 5: 25-28. PubMed ID: 21057219

  • Marc Gershow Department of Physics, New York University
    Jovanic, T., Winding, M., Cardona, A., Truman, J. W., Gershow, M. and Zlatic, M. (2019). Neural substrates of Drosophila larval anemotaxis. Curr Biol. PubMed ID: 30744969

    Gepner, R., Mihovilovic Skanata, M., Bernat, N. M., Kaplow, M. and Gershow, M. (2015). Computations underlying photo-taxis, odor-taxis, and multi-sensory integration. Elife 4. PubMed ID: 25945916

    Klein, M., Afonso, B., Vonner, A. J., Hernandez-Nunez, L., Berck, M., Tabone, C. J., Kane, E. A., Pieribone, V. A., Nitabach, M. N., Cardona, A., Zlatic, M., Sprecher, S. G., Gershow, M., Garrity, P. A. and Samuel, A. D. (2015). Sensory determinants of behavioral dynamics in Drosophila thermotaxis. Proc Natl Acad Sci U S A 112: E220-229. PubMed ID: 25550513

    Kane, E. A., Gershow, M., Afonso, B., Larderet, I., Klein, M., Carter, A. R., de Bivort, B. L., Sprecher, S. G. and Samuel, A. D. (2013). Sensorimotor structure of Drosophila larva phototaxis. Proc Natl Acad Sci U S A 110: E3868-3877. PubMed ID: 24043822


  • Jason Gerstner Elson S. Floyd College of Medicine, Washington State University, Spokane
    Vanderheyden, W. M., Van Dongen, H. P. A., Frank, M. G. and Gerstner, J. R. (2019). Sleep pressure regulates mushroom body neural-glial interactions in Drosophila. Matters Sel 2019. PubMed ID: 31938713

    Vanderheyden, W. M., Goodman, A. G., Taylor, R. H., Frank, M. G., Van Dongen, H. P. A. and Gerstner, J. R. (2018). Astrocyte expression of the Drosophila TNF-alpha homologue, Eiger, regulates sleep in flies. PLoS Genet 14(10): e1007724. PubMed ID: 30379810

    Gerstner, J. R., Perron, I. J., Riedy, S. M., Yoshikawa, T., Kadotani, H., Owada, Y., Van Dongen, H. P. A., Galante, R. J., Dickinson, K., Yin, J. C. P., Pack, A. I. and Frank, M. G. (2017). Normal sleep requires the astrocyte brain-type fatty acid binding protein FABP7. Sci Adv 3(4): e1602663. PubMed ID: 28435883

    Gerstner, J. R., Lenz, O., Vanderheyden, W. M., Chan, M. T., Pfeiffenberger, C. and Pack, A. I. (2017). Amyloid-beta induces sleep fragmentation that is rescued by fatty acid binding proteins in Drosophila. J Neurosci Res 95(8): 1548-1564. PubMed ID: 27320125

  • Pam Geyer Molecular and Cellular Biology, University of Iowa
    Duan, T., Thyagarajan, S., Amoiroglou, A., Rogers, G. C. and Geyer, P. K. (2023). Analysis of a rare progeria variant of Barrier-to-autointegration factor in Drosophila connects centromere function to tissue homeostasis. Cell Mol Life Sci 80(3): 73. PubMed ID: 36842139

    Kitzman, S. C., Duan, T., Pufall, M. A. and Geyer, P. K. (2021). Checkpoint activation drives global gene expression changes in Drosophila nuclear lamina mutants. G3 (Bethesda). PubMed ID: 34893833

    Duan, T., Cupp, R. and Geyer, P. K. (2021). Drosophila female germline stem cells undergo mitosis without nuclear breakdown. Curr Biol. PubMed ID: 33548191

    Duan, T., Kitzman, S. C. and Geyer, P. K. (2020). Survival of Drosophila germline stem cells requires the chromatin binding protein Barrier-to-autointegration factor. Development. PubMed ID: 32345742

    Glenn, S. E. and Geyer, P. K. (2018). Investigation of the developmental requirements of Drosophila HP1 and insulator protein partner, HIPP1. G3 (Bethesda). PubMed ID: 30514714

    Barton, L. J., Duan, T., Ke, W., Luttinger, A., Lovander, K. E., Soshnev, A. A. and Geyer, P. K. (2018). Nuclear lamina dysfunction triggers a germline stem cell checkpoint. Nat Commun 9(1): 3960. PubMed ID: 30262885

    Duan, T. and Geyer, P. K. (2018). Spermiogenesis and male fertility require the function of Suppressor of Hairy-Wing in somatic cyst cells of Drosophila. Genetics. PubMed ID: 29739818

    Barton, L. J., Lovander, K. E., Pinto, B. S. and Geyer, P. K. (2016). Drosophila male and female germline stem cell niches require the nuclear lamina protein Otefin. Dev Biol. PubMed ID: 27174470

    Barton, L. and Geyer, P. (2014). Stacking the deck for the next generation. Mol Reprod Dev. PubMed ID: 24771577

    Barton, L. J., Wilmington, S. R., Martin, M. J., Skopec, H. M., Lovander, K. E., Pinto, B. S. and Geyer, P. K. (2014). Unique and Shared Functions of Nuclear Lamina LEM Domain Proteins in Drosophila. Genetics. PubMed ID: 24700158

    Barton, L. J., Pinto, B. S., Wallrath, L. L. and Geyer, P. K. (2013). The Drosophila nuclear lamina protein otefin is required for germline stem cell survival. Dev Cell 25: 645-654. PubMed ID: 23806619

    Hohl, A. M., Thompson, M., Soshnev, A. A., Wu, J., Morris, J., Hsieh, T. S., Wu, C. T. and Geyer, P. K. (2012). Restoration of topoisomerase 2 function by complementation of defective monomers in Drosophila. Genetics 192: 843-856. PubMed ID: 22923380

  • Amin Ghabrial Cell and Developmental Biology, University of Pennsylvania, Philadelphia
    Francis, D., Burguete, A. S. and Ghabrial, A. (2022). Regulation of Archease by the mTOR-vATPase axis. Development. PubMed ID: 36111596

    Schweizer Burguete, A. B. and Ghabrial, A. S. (2020). Dissection of the Role of CCM Genes in Tubulogenesis Using the Drosophila Tracheal System as a Model. Methods Mol Biol 2152: 179-189. PubMed ID: 32524553

    Burguete, A. S., Francis, D., Rosa, J. and Ghabrial, A. (2019). The regulation of cell size and branch complexity in the terminal cells of the Drosophila tracheal system. Dev Biol 451(1): 79-85. PubMed ID: 30735663

    Rosa, J. B., Metzstein, M. M. and Ghabrial, A. S. (2018). An Ichor-dependent apical extracellular matrix regulates seamless tube shape and integrity. PLoS Genet 14(1): e1007146. PubMed ID: 29309404

    Francis, D. and Ghabrial, A. S. (2015). Compensatory branching morphogenesis of stalk cells in the Drosophila trachea. Development 142: 2048-2057. PubMed ID: 25977367

    Schottenfeld-Roames, J., Rosa, J. B. and Ghabrial, A. S. (2014). Seamless tube shape is constrained by endocytosis-dependent regulation of active moesin. Curr Biol 24: 1756-1764. PubMed ID: 25065756

    Song, Y., Eng, M. and Ghabrial, A. S. (2013). Focal Defects in Single-Celled Tubes Mutant for Cerebral Cavernous Malformation 3, GCKIII, or NSF2. Dev Cell 25: 507-519. PubMed ID: 23763949

    Ghabrial, A. S., Levi, B. P. and Krasnow, M. A. (2011). A systematic screen for tube morphogenesis and branching genes in the Drosophila tracheal system. PLoS Genet 7: e1002087. PubMed ID: 21750678

    Schottenfeld, J., Song, Y. and Ghabrial, A. S. (2010). Tube continued: morphogenesis of the Drosophila tracheal system. Curr Opin Cell Biol 22: 633-639. PubMed ID: 20739171

  • Michel Gho Laboratory of Developmental Biology, Université Pierre et Marie Curie, Paris
    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. 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 [Epub ahead of print]. PubMed ID: 27471258

    Salle, J., Campbell, S. D., Gho, M. and Audibert, A. (2012). CycA is involved in the control of endoreplication dynamics in the Drosophila bristle lineage. Development 139: 547-557. PubMed ID: 22223681

    Buffin, E. and Gho, M. (2010). Laser microdissection of sensory organ precursor cells of Drosophila microchaetes. PLoS One 5: e9285. PubMed ID: 20174573

  • Sujay Ghosh Department of Zoology, University of Calcutta
    Das, D., Begum, M., Paul, P., Dutta, I., Mandal, S., Ghosh, P. and Ghosh, S. (2022). Effects of plant growth retardant daminozide (Alar) on neuromuscular co-ordination behavior in Drosophila melanogaster. J Toxicol Environ Health A 85(22): 921-936. PubMed ID: 35996764

    Begum, M., Paul, P., Das, D., Chakraborty, K., Bhattacharjee, A. and Ghosh, S. (2021). Genes regulating development and behavior exhibited altered expression in Drosophila melanogaster exposed to bisphenol A: use of real-time quantitative PCR (qRT-PCR) and droplet digital PCR (ddPCR) in genotoxicity study. Environ Sci Pollut Res Int 28(6): 7090-7104. PubMed ID: 33025430

    Roy, S. S. and Ghosh, S. (2020). Genes regulating wing patterning in Drosophila melanogaster show reduced expression under exposure of Daminozide, the fruit ripening retardant. Environ Toxicol Pharmacol 75: 103322. PubMed ID: 31877500

  • Angela Giangrande Institute of Genetics and Molecular and Cellular Biology, Strasbourg
    Voutyraki, C., Choromidis, A., Meligkounaki, A., Vlachopoulos, N. A., Theodorou, V., Grammenoudi, S., Athanasiadis, E., Monticelli, S., Giangrande, A., Delidakis, C. and Zacharioudaki, E. (2023). Growth deregulation and interaction with host hemocytes contribute to tumor progression in a Drosophila brain tumor model. Proc Natl Acad Sci U S A 120(33): e2221601120. PubMed ID: 37549261

    Pavlidaki, A., Panic, R., Monticelli, S., Riet, C., Yuasa, Y., Cattenoz, P. B., Nait-Oumesmar, B. and Giangrande, A. (2022). An anti-inflammatory transcriptional cascade conserved from flies to humans. Cell Rep 41(3): 111506. PubMed ID: 36261018

    Cattenoz, P. B., Monticelli, S., Pavlidaki, A. and Giangrande, A. (2021). Toward a Consensus in the Repertoire of Hemocytes Identified in Drosophila. Front Cell Dev Biol 9: 643712. PubMed ID: 33748138

    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

    Trebuchet, G., Cattenoz, P. B., Zsamboki, J., Mazaud, D., Siekhaus, D. E., Fanto, M. and Giangrande, A. (2018). The Repo homeodomain transcription factor suppresses hematopoiesis in Drosophila and preserves the glial fate. J Neurosci. PubMed ID: 30504274

    Bazzi, W., Cattenoz, P. B., Delaporte, C., Dasari, V., Sakr, R., Yuasa, Y. and Giangrande, A. (2018). Embryonic hematopoiesis modulates the inflammatory response and larval hematopoiesis in Drosophila. Elife 7. PubMed ID: 29992900

    Abekhoukh, S., Sahin, H. B., Grossi, M., Zongaro, S., Maurin, T., Madrigal, I., Kazue-Sugioka, D., Raas-Rothschild, A., Doulazmi, M., Carrera, P., Stachon, A., Scherer, S., Nascimento, M. R., Trembleau, A., Arroyo, I., Peter, S., Smith, I. M., Mila, M., Smith, A. C., Giangrande, A., Caille, I. and Bardoni, B. (2017). New insights into the regulatory function of CYFIP1 in the context of WAVE- and FMRP-containing complexes. Dis Model Mech. PubMed ID: 28183735

    Cattenoz, P. B., Delaporte, C., Bazzi, W. and Giangrande, A. (2016). An evolutionary conserved interaction between the Gcm transcription factor and the SF1 nuclear receptor in the female reproductive system. Sci Rep 6: 37792. PubMed ID: 27886257

    Gupta, T., Kumar, A., Pierre, C., VijayRaghavan, K. and Giangrande, A. (2016). The Glide/Gcm fate determinant controls initiation of collective cell migration by regulating Frazzled. Elife 5 [Epub ahead of print]. PubMed ID: 27740455

    Cattenoz, P. B., Popkova, A., Southall, T., Aiello, G., Brand, A. and Giangrande, A. (2015). Functional conservation of the Glide/Gcm regulatory network controlling glia, hemocyte and tendon cell differentiation in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 26567182

  • Mariagrazia Giansanti Department of Biology and Biotechnology, Institute of Molecular Biology and Pathology, Rome
    Frappaolo, A., Piergentili, R. and Giansanti, M. G. (2022). Microtubule and Actin Cytoskeletal Dynamics in Male Meiotic Cells of Drosophila melanogaster. Cells 11(4). PubMed ID: 35203341

    Sechi, S., Karimpour-Ghahnavieh, A., Frappaolo, A., Di Francesco, L., Piergentili, R., Schinina, E., D'Avino, P. P. and Giansanti, M. G. (2021). Identification of GOLPH3 Partners in Drosophila Unveils Potential Novel Roles in Tumorigenesis and Neural Disorders. Cells 10(9). PubMed ID: 34571985

    Sechi, S., Frappaolo, A., Karimpour-Ghahnavieh, A., Fraschini, R. and Giansanti, M. G. (2020). A novel coordinated function of Myosin II with GOLPH3 controls centralspindlin localization during cytokinesis in Drosophila. J Cell Sci 133(21). PubMed ID: 33037125

    Sechi, S., Frappaolo, A., Karimpour-Ghahnavieh, A., Gottardo, M., Burla, R., Di Francesco, L., Szafer-Glusman, E., Schinina, E., Fuller, M. T., Saggio, I., Riparbelli, M. G., Callaini, G. and Giansanti, M. G. (2019). Drosophila Doublefault protein coordinates multiple events during male meiosis by controlling mRNA translation. Development 146(22). PubMed ID: 31645358

    Frappaolo, A., Sechi, S., Kumagai, T., Karimpour-Ghahnavieh, A., Tiemeyer, M. and Giansanti, M. G. (2018). Modeling congenital disorders of N-linked glycoprotein glycosylation in Drosophila melanogaster. Front Genet 9: 436. PubMed ID: 30333856

    Sechi, S., Frappaolo, A., Fraschini, R., Capalbo, L., Gottardo, M., Belloni, G., Glover, D. M., Wainman, A. and Giansanti, M. G. (2017). Rab1 interacts with GOLPH3 and controls Golgi structure and contractile ring constriction during cytokinesis in Drosophila melanogaster. Open Biol 7(1). PubMed ID: 28100664

    Sechi, S., Frappaolo, A., Belloni, G. and Giansanti, M. G. (2015). The roles of the oncoprotein GOLPH3 in contractile ring assembly and membrane trafficking during cytokinesis. Biochem Soc Trans 43: 117-121. PubMed ID: 25619256

    Sechi, S., Colotti, G., Belloni, G., Mattei, V., Frappaolo, A., Raffa, G. D., Fuller, M. T. and Giansanti, M. G. (2014). GOLPH3 Is Essential for Contractile Ring Formation and Rab11 Localization to the Cleavage Site during Cytokinesis in Drosophila melanogaster. PLoS Genet 10: e1004305. PubMed ID: 24786584

    Sechi, S., Colotti, G., Belloni, G., Mattei, V., Frappaolo, A., Raffa, G. D., Fuller, M. T. and Giansanti, M. G. (2014). GOLPH3 Is Essential for Contractile Ring Formation and Rab11 Localization to the Cleavage Site during Cytokinesis in Drosophila melanogaster. PLoS Genet 10: e1004305. PubMed ID: 24786584

    Giansanti, M. G., Sechi, S., Frappaolo, A., Belloni, G. and Piergentili, R. (2012). Cytokinesis in Drosophila male meiosis. Spermatogenesis 2: 185-196. PubMed ID: 23094234

  • Allen Gibbs School of Life Sciences, University of Nevada, Las Vegas
    Etges, W. J., de Oliveira, C. C., Rajpurohit, S. and Gibbs, A. G. (2017). Effects of temperature on transcriptome and cuticular hydrocarbon expression in ecologically differentiated populations of desert Drosophila. Ecol Evol 7(2): 619-637. PubMed ID: 28116058

    Hardy, C. M., Birse, R. T., Wolf, M. J., Yu, L., Bodmer, R. and Gibbs, A. G. (2015). Obesity-associated cardiac dysfunction in starvation-selected Drosophila melanogaster. Am J Physiol Regul Integr Comp Physiol: ajpregu 00160 02015. PubMed ID: 26136533

    Slocumb, M.E., Regalado, J.M., Yoshizawa, M., Neely, G.G., Masek, P., Gibbs, A.G. and Keene, A.C. (2015). Enhanced sleep is an evolutionarily adaptive response to starvation stress in Drosophila. PLoS One 10: e0131275. PubMed ID: 26147198

    Rajpurohit, S., Nedved, O. and Gibbs, A. G. (2013). Meta-analysis of geographical clines in desiccation tolerance of Indian drosophilids. Comp Biochem Physiol A Mol Integr Physiol 164: 391-398. PubMed ID: 23182926

    Merkey, A. B., Wong, C. K., Hoshizaki, D. K. and Gibbs, A. G. (2011). Energetics of metamorphosis in Drosophila melanogaster. J Insect Physiol 57: 1437-1445. PubMed ID: 21810426

    Bond, N. D., Hoshizaki, D. K. and Gibbs, A. G. (2010). The role of 20-hydroxyecdysone signaling in Drosophila pupal metabolism. Comp Biochem Physiol A Mol Integr Physiol 157: 398-404. PubMed ID: 20817116

  • Matthew Gibson Stowers Institute for Medical Research, Kansas City. Missouri
    Akiyama, T., Seidel, C. W. and Gibson, M. C. (2022). The feedback regulator Nord controls Dpp/BMP signaling via extracellular interaction with Dally in the Drosophila wing. Dev Biol 488: 91-103. PubMed ID: 35609633Bauerly, E., Akiyama, T., Staber, C., Yi, K. and Gibson, M. C. (2022). Impact of cilia-related genes on mitochondrial dynamics during Drosophila spermatogenesis. Dev Biol 482: 17-27. PubMed ID: 34822845

    Bauerly, E., Yi, K. and Gibson, M. C. (2020). Wampa is a dynein subunit required for axonemal assembly and male fertility in Drosophila. Dev Biol. PubMed ID: 32387369

    Ramanathan, S. P., Krajnc, M. and Gibson, M. C. (2019). Cell-size pleomorphism drives aberrant clone dispersal in proliferating epithelia. Dev Cell. PubMed ID: 31495693

    Nakajima, Y. I., Lee, Z. T., McKinney, S. A., Swanson, S. K., Florens, L. and Gibson, M. C. (2019). Junctional tumor suppressors interact with 14-3-3 proteins to control planar spindle alignment. J Cell Biol. PubMed ID: 31088859

    Akiyama, T., User, S. D. and Gibson, M. C. (2018). Somatic clones heterozygous for recessive disease alleles of BMPR1A exhibit unexpected phenotypes in Drosophila. Elife 7. PubMed ID: 29745898

    Akiyama, T. and Gibson, M. C. (2015). Decapentaplegic and growth control in the developing Drosophila wing. Nature 527: 375-378. PubMed ID: 26550824

    Liang, L., Haug, J. S., Seidel, C. W. and Gibson, M. C. (2014). Functional Genomic Analysis of the Periodic Transcriptome in the Developing Drosophila Wing. Dev Cell. PubMed ID: 24684830

    Ikmi, A., Gaertner, B., Seidel, C., Srivastava, M., Zeitlinger, J. and Gibson, M. C. (2014). Molecular Evolution of the Yap/Yorkie Proto-Oncogene and Elucidation of Its Core Transcriptional Program. Mol Biol Evol. PubMed ID: 24509725

    Nakajima, Y., Meyer, E. J., Kroesen, A., McKinney, S. A. and Gibson, M. C. (2013). Epithelial junctions maintain tissue architecture by directing planar spindle orientation. Nature 500: 359-362. PubMed ID: 23873041

  • Jadwiga Giebultowicz Center for Genome Research and Biocomputing, Oregon State University, Corvallis
    Yang, J., Song, Y., Law, A. D., Rogan, C. J., Shimoda, K., Djukovic, D., Anderson, J. C., Kretzschmar, D., Hendrix, D. A. and Giebultowicz, J. M. (2022). Chronic blue light leads to accelerated aging in Drosophila by impairing energy metabolism and neurotransmitter levels. Front Aging 3: 983373. PubMed ID: 36118990

    Long, D. M., Frame, A. K., Reardon, P. N., Cumming, R. C., Hendrix, D. A., Kretzschmar, D. and Giebultowicz, J. M. (2020). Lactate dehydrogenase expression modulates longevity and neurodegeneration in Drosophila melanogaster. Aging (Albany NY) 12. PubMed ID: 32484787

    Nash, T. R., Chow, E. S., Law, A. D., Fu, S. D., Fuszara, E., Bilska, A., Bebas, P., Kretzschmar, D. and Giebultowicz, J. M. (2019). Daily blue-light exposure shortens lifespan and causes brain neurodegeneration in Drosophila. NPJ Aging Mech Dis 5: 8. PubMed ID: 31636947

    Long, D. M. and Giebultowicz, J. M. (2017). Age-related changes in the expression of the circadian clock protein PERIOD in Drosophila glial cells. Front Physiol 8: 1131. PubMed ID: 29375400

    Chow, E. S., Long, D. M. and Giebultowicz, J. M. (2016). Circadian rhythm in mRNA expression of the glutathione synthesis gene Gclc is controlled by peripheral glial clocks in Drosophila melanogaster. Physiol Entomol 41(4): 369-377. PubMed ID: 28503020

    Egenriether, S. M., Chow, E. S., Krauth, N. and Giebultowicz, J. M. (2015). Accelerated food source location in aging Drosophila. Aging Cell. PubMed ID: 26102220

    Blake, M. R., Holbrook, S. D., Kotwica-Rolinska, J., Chow, E. S., Kretzschmar, D. and Giebultowicz, J. M. (2015). Manipulations of amyloid precursor protein cleavage disrupt the circadian clock in aging Drosophila. Neurobiol Dis 77: 117-126. PubMed ID: 25766673

    Long, D. M., Blake, M. R., Dutta, S., Holbrook, S. D., Kotwica-Rolinska, J., Kretzschmar, D. and Giebultowicz, J. M. (2014). Relationships between the Circadian System and Alzheimer's Disease-Like Symptoms in Drosophila. PLoS One 9: e106068. PubMed ID: 25171136

    Rakshit, K., Giebultowicz, J. M. (2013) Cryptochrome restores dampened circadian rhythms and promotes health span in aging Drosophila. Aging Cell. PubMed ID: 23692507

    Beaver, L. M., Klichko, V. I., Chow, E. S., Kotwica-Rolinska, J., Williamson, M., Orr, W. C., Radyuk, S. N. and Giebultowicz, J. M. (2012). Circadian regulation of glutathione levels and biosynthesis in Drosophila melanogaster. PLoS One 7: e50454. PubMed ID: 23226288

  • Jadwiga Giebultowicz Center for Genome Research and Biocomputing, Oregon State University, Corvallis
    Rakshit, K., Wambua, R., Giebultowicz, T. M. and Giebultowicz, J. M. (2013). Effects of exercise on circadian rhythms and mobility in aging Drosophila melanogaster. Exp Gerontol. PubMed ID: 23916842

    Rakshit, K. and Giebultowicz, J. M. (2013). Cryptochrome restores dampened circadian rhythms and promotes healthspan in aging Drosophila. Aging Cell. PubMed ID: 23692507

    Beaver, L. M., Klichko, V. I., Chow, E. S., Kotwica-Rolinska, J., Williamson, M., Orr, W. C., Radyuk, S. N. and Giebultowicz, J. M. (2012). Circadian regulation of glutathione levels and biosynthesis in Drosophila melanogaster. PLoS One 7: e50454. PubMed ID: 23226288

  • Régis Giet Institute of Genetics & Development of Rennes
    Thomas, A., Gallaud, E., Pascal, A., Serre, L., Arnal, I., Richard-Parpaillon, L., Savoian, M. S. and Giet, R. (2021). Peripheral astral microtubules ensure asymmetric furrow positioning in neural stem cells. Cell Rep 37(4): 109895. PubMed ID: 34706235

    Metivier, M., Gallaud, E., Thomas, A., Pascal, A., Gagne, J. P., Poirier, G. G., Chretien, D., Gibeaux, R., Richard-Parpaillon, L., Benaud, C. and Giet, R. (2020). Drosophila Tubulin-Specific Chaperone E Recruits Tubulin around Chromatin to Promote Mitotic Spindle Assembly. Curr Biol. PubMed ID: 33259793

    Metivier, M., Monroy, B. Y., Gallaud, E., Caous, R., Pascal, A., Richard-Parpaillon, L., Guichet, A., Ori-McKenney, K. M. and Giet, R. (2019). Dual control of Kinesin-1 recruitment to microtubules by Ensconsin in Drosophila neuroblasts and oocytes. Development 146(8). PubMed ID: 30936181

    Magnaghi-Jaulin, L., Eot-Houllier, G., Gallaud, E. and Giet, R. (2019). Aurora A Protein Kinase: To the Centrosome and Beyond. Biomolecules 9(1). PubMed ID: 30650622

    Beaven, R., Bastos, R. N., Spanos, C., Rome, P., Cullen, C. F., Rappsilber, J., Giet, R., Goshima, G. and Ohkura, H. (2017). 14-3-3 regulation of Ncd reveals a new mechanism for targeting proteins to the spindle in oocytes. J Cell Biol 216(10): 3029-3039. PubMed ID: 28860275

    Caous, R., Richard-Parpaillon, L. and Giet, R. (2016). Drosophila Aurora A regulates mitotic timing in cancer stem cells: Possible therapeutic implications. Mol Cell Oncol 3(3): e1140261. PubMed ID: 27314090

  • Lilach Gilboa Weizmann Institute
    Banisch, T. U., Maimon, I., Dadosh, T. and Gilboa, L. (2017). Escort cells generate a dynamic compartment for germline stem cell differentiation via combined Stat and Erk signalling. Development 144(11): 1937-1947. PubMed ID: 28559239

    Hitrik, A., Popliker, M., Gancz, D., Mukamel, Z., Lifshitz, A., Schwartzman, O., Tanay, A. and Gilboa, L. (2016). Combgap promotes ovarian niche development and chromatin association of EcR-binding regions in BR-C. PLoS Genet 12: e1006330. PubMed ID: 27846223

    Gilboa, L. (2015). Organizing stem cell units in the Drosophila ovary. Curr Opin Genet Dev 32C: 31-36. PubMed ID: 25703842

    Lengil, T., Gancz, D. and Gilboa, L. (2015). Activin signaling balances proliferation and differentiation of ovarian niche precursors and enables adjustment of niche numbers. Development. PubMed ID: 25633355

    Maimon, I., Popliker, M. and Gilboa, L. (2014). Without children is required for Stat-mediated zfh1 transcription and for germline stem cell differentiation. Development 141: 2602-2610. PubMed ID: 24903753

    Gancz, D. and Gilboa, L. (2013). Insulin and Target of rapamycin signaling orchestrate the development of ovarian niche-stem cell units in Drosophila. Development. PubMed ID: 24026119

    Maimon, I. and Gilboa, L. (2011). Dissection and staining of Drosophila larval ovaries. J Vis Exp. PubMed ID: 21610675

    Gilboa, L. and Boutros, M. (2010). Celebrating 100 years of Drosophila research. EMBO Rep 11: 724-726. PubMed ID: 20847739

  • David Gilmour Biochemistry and Molecular Biology, Penn State University
    Dollinger, R., Deng, E. B., Schultz, J., Wu, S., Deorio, H. R. and Gilmour, D. S. (2023). DSIF directly facilitates promoter proximal pausing of RNA Polymerase II. J Biol Chem: 105106. PubMed ID: 37517697

    Lu, F., Portz, B. and Gilmour, D. S. (2019). The C-terminal domain of RNA polymerase II is a multivalent targeting sequence that supports Drosophila development with only consensus heptads. Mol Cell. PubMed ID: 30765194

    Baumann, D. G. and Gilmour, D. S. (2017). A sequence-specific core promoter-binding transcription factor recruits TRF2 to coordinately transcribe ribosomal protein genes. Nucleic Acids Res 45(18): 10481-10491. PubMed ID: 28977400

    Baumann, D. G., Dai, M. S., Lu, H. and Gilmour, D. S. (2017). GFZF, a glutathione S-transferase protein implicated in cell cycle regulation and hybrid inviability, is a transcriptional co-activator. Mol Cell Biol. PubMed ID: 29158293

    Portz, B., Lu, F., Gibbs, E. B., Mayfield, J. E., Rachel Mehaffey, M., Zhang, Y. J., Brodbelt, J. S., Showalter, S. A. and Gilmour, D. S. (2017). Structural heterogeneity in the intrinsically disordered RNA polymerase II C-terminal domain. Nat Commun 8: 15231. PubMed ID: 28497792

    Gibbs, E. B., Lu, F., Portz, B., Fisher, M. J., Medellin, B. P., Laremore, T. N., Zhang, Y. J., Gilmour, D. S. and Showalter, S. A. (2017). Phosphorylation induces sequence-specific conformational switches in the RNA polymerase II C-terminal domain. Nat Commun 8: 15233. PubMed ID: 28497798

    Qiu, Y. and Gilmour, D. S. (2017). Identification of Regions in the Spt5 Subunit of DSIF That Are Involved in Promoter Proximal Pausing. J Biol Chem. PubMed ID: 28213523

    Li, J. and Gilmour, D. S. (2015). Reconstitution of factor-dependent, promoter proximal pausing in Drosophila nuclear extracts. Methods Mol Biol 1276: 133-152. PubMed ID: 25665561

    Achary, B. G., Campbell, K. M., Co, I. S. and Gilmour, D. S. (2014). RNAi screen in Drosophila larvae identifies histone deacetylase 3 as a positive regulator of the hsp70 heat shock gene expression during heat shock. Biochim Biophys Acta. PubMed ID: 24607507

    Li, J., Liu, Y., Rhee, H. S., Ghosh, S. K., Bai, L., Pugh, B. F. and Gilmour, D. S. (2013). Kinetic Competition between Elongation Rate and Binding of NELF Controls Promoter-Proximal Pausing. Mol Cell 50: 711-722. PubMed ID: 23746353

    Li, J., Gilmour, D. S. (2013) Distinct mechanisms of transcriptional pausing orchestrated by GAGA factor and M1BP, a novel transcription factor. EMBO J. PubMed ID: 23708796

  • Ed Giniger Axon Guidance and Neural Connectivity Unit, NINDS, NIH
    Koca, Y., Vuong, L. T., Singh, J., Giniger, E. and Mlodzik, M. (2022). Notch-dependent Abl signaling regulates cell motility during ommatidial rotation in Drosophila. Cell Rep 41(10): 111788. PubMed ID: 36476875

    Fang, H. Y., Forghani, R., Clarke, A., McQueen, P. G., Chandrasekaran, A., O'Neill, K. M., Losert, W., Papoian, G. A. and Giniger, E. (2023). Enabled primarily controls filopodial morphology, not actin organization, in the TSM1 growth cone in Drosophila. Mol Biol Cell 34(8): ar83. PubMed ID: 37223966

    Shukla, A. K., Johnson, K. and Giniger, E. (2021). Common features of aging fail to occur in Drosophila raised without a bacterial microbiome. iScience 24(7): 102703. PubMed ID: 34235409

    Marquilly, C., Busto, G. U., Leger, B. S., Boulanger, A., Giniger, E., Walker, J. A., Fradkin, L. G. and Dura, J. M. (2021). Htt is a repressor of Abl activity required for APP-induced axonal growth. PLoS Genet 17(1): e1009287. PubMed ID: 33465062

    Clarke, A., McQueen, P. G., Fang, H. Y., Kannan, R., Wang, V., McCreedy, E., Wincovitch, S. and Giniger, E. (2020). Abl signaling directs growth of a pioneer axon in Drosophila by shaping the intrinsic fluctuations of actin. Mol Biol Cell: mbcE19100564. PubMed ID: 31967946

    Clarke, A., McQueen, P. G., Fang, H. Y., Kannan, R., Wang, V., McCreedy, E., Buckley, T., Johannessen, E. M., Wincovitch, S. and Giniger, E. (2020). Dynamic morphogenesis of a pioneer axon in Drosophila and its regulation by Abl tyrosine kinase. Mol Biol Cell: mbcE19100563. PubMed ID: 31967935

    Shukla, A. K. and Giniger, E. (2019). Reduced autophagy efficiency induces innate immune activation leading to neurodegeneration. Autophagy. PubMed ID: 30898009

    Spurrier, J., Shukla, A. K., Buckley, T., Smith-Trunova, S., Kuzina, I., Gu, Q. and Giniger, E. (2019). Expression of a fragment of Ankyrin 2 disrupts the structure of the axon initial segment and causes axonal degeneration in Drosophila. Mol Neurobiol. PubMed ID: 30666562

    Shukla, A. K., Spurrier, J., Kuzina, I. and Giniger, E. (2019). Hyperactive innate immunity causes degeneration of dopamine neurons upon altering activity of Cdk5. Cell Rep 26(1): 131-144.e134. PubMed ID: 30605670

    Spurrier, J., Shukla, A. K., McLinden, K., Johnson, K. and Giniger, E. (2018). Altered expression of the Cdk5 activator-like protein, Cdk5alpha, causes neurodegeneration in part by accelerating the rate of aging. Dis Model Mech. PubMed ID: 29469033

    Kannan, R., Cox, E., Wang, L., Kuzina, I., Gu, Q. and Giniger, E. (2018). Tyrosine phosphorylation and proteolytic cleavage of Notch are required for non-canonical Notch/Abl signaling in Drosophila axon guidance. Development 145(2). PubMed ID: 29343637

    Kannan, R., Song, J. K., Karpova, T., Clarke, A., Shivalkar, M., Wang, B., Kotlyanskaya, L., Kuzina, I., Gu, Q. and Giniger, E. (2017). The Abl pathway bifurcates to balance Enabled and Rac signaling in axon patterning in Drosophila. Development. PubMed ID: 28087633

  • Santhosh Girirajan Department of Biochemistry and Molecular Biology, Penn State, University Park, PA
    Pizzo, L., Lasser, M., Yusuff, T., Jensen, M., Ingraham, P., Huber, E., Singh, M. D., Monahan, C., Iyer, J., Desai, I., Karthikeyan, S., Gould, D. J., Yennawar, S., Weiner, A. T., Pounraja, V. K., Krishnan, A., Rolls, M. M., Lowery, L. A. and Girirajan, S. (2021). Functional assessment of the "two-hit" model for neurodevelopmental defects in Drosophila and X. laevis. PLoS Genet 17(4): e1009112. PubMed ID: 33819264

    Yusuff, T., Jensen, M., Yennawar, S., Pizzo, L., Karthikeyan, S., Gould, D. J., Sarker, A., Gedvilaite, E., Matsui, Y., Iyer, J., Lai, Z. C. and Girirajan, S. (2020). Drosophila models of pathogenic copy-number variant genes show global and non-neuronal defects during development. PLoS Genet 16(6): e1008792. PubMed ID: 32579612

    Iyer, J., Singh, M. D., Jensen, M., Patel, P., Pizzo, L., Huber, E., Koerselman, H., Weiner, A. T., Lepanto, P., Vadodaria, K., Kubina, A., Wang, Q., Talbert, A., Yennawar, S., Badano, J., Manak, J. R., Rolls, M. M., Krishnan, A. and Girirajan, S. (2018). Pervasive genetic interactions modulate neurodevelopmental defects of the autism-associated 16p11.2 deletion in Drosophila melanogaster. Nat Commun 9(1): 2548. PubMed ID: 26994292

    ones, M. A., Amr, S., Ferebee, A., Huynh, P., Rosenfeld, J. A., Miles, M. F., Davies, A. G., Korey, C. A., Warrick, J. M., Shiang, R., Elsea, S. H., Girirajan, S. and Grotewiel, M. (2014). Genetic studies in Drosophila and humans support a model for the concerted function of CISD2, PPT1 and CLN3 in disease. Biol Open 3(5): 342-352. PubMed ID: 24705017

  • Gunter Gisselmann Lehrstuhl fur Zellphysiologie, Ruhr Universitat Bochum
    Rollecke, K., Werner, M., Ziemba, P. M., Neuhaus, E. M., Hatt, H. and Gisselmann, G. (2013). Amiloride Derivatives Are Effective Blockers of Insect Odorant Receptors. Chem Senses. PubMed ID: 23329732

    Deng, Y., Zhang, W., Farhat, K., Oberland, S., Gisselmann, G. and Neuhaus, E. M. (2011). The stimulatory Galpha(s) protein is involved in olfactory signal transduction in Drosophila. PLoS One 6: e18605. PubMed ID: 21490930

    Bredendiek, N., Hutte, J., Steingraber, A., Hatt, H., Gisselmann, G. and Neuhaus, E. M. (2011). Go alpha is involved in sugar perception in Drosophila. Chem Senses 36: 69-81. PubMed ID: 20940344

  • Alvaro Glavic Department of Biology, University of Chile, Santiago de Chile
    Molina, E., Cataldo, V. F., Eggers, C., Munoz-Madrid, V. and Glavic, A. (2022). p53 Related Protein Kinase is Required for Arp2/3-Dependent Actin Dynamics of Hemocytes in Drosophila melanogaster. Front Cell Dev Biol 10: 859105. PubMed ID: 35721516

    Molina-Pelayo, C., Olguin, P., Mlodzik, M. and Glavic, A. (2022). The conserved Pelado/ZSWIM8 protein regulates actin dynamics by promoting linear actin filament polymerization. Life Sci Alliance 5(12). PubMed ID: 35940847

    Edwards-Jorquera, S. S., Bosveld, F., Bellaiche, Y. A., Lennon-Dumenil, A. M. and Glavic, A. (2020). Trpml controls actomyosin contractility and couples migration to phagocytosis in fly macrophages. J Cell Biol 219(3). PubMed ID: 31940424

    Gonzalez-Itier, S., Contreras, E. G., Larrain, J., Glavic, A. and Faunes, F. (2018). A role for Lin-28 in growth and metamorphosis in Drosophila melanogaster. Mech Dev. PubMed ID: 29908237

    Contreras, E. G., Sierralta, J. and Glavic, A. (2018). p53 is required for brain growth but is dispensable for resistance to nutrient restriction during Drosophila larval development. PLoS One 13(4): e0194344. PubMed ID: 29621246

    Ibar, C. and Glavic, A. (2017). Drosophila p115 is required for Cdk1 activation and G2/M cell cycle transition. Mech Dev [Epub ahead of print]. PubMed ID: 28396045

    Rojas-Benitez, D., Eggers, C. and Glavic, A. (2017). Modulation of the Proteostasis Machinery to Overcome Stress Caused by Diminished Levels of t6A-Modified tRNAs in Drosophila. Biomolecules 7(1). PubMed ID: 28272317

    Rojas-Benitez, D., Ibar, C. and Glavic, A. (2013). The Drosophila EKC/KEOPS complex: Roles in protein synthesis homeostasis and animal growth. Fly (Austin) 7. PubMed ID: 23823807

    Ibar, C., Cataldo, V. F., Vasquez-Doorman, C., Olguin, P., Glavic, A. (2013) Drosophila p53-related protein kinase is required for PI3K/TOR pathway-dependent growth. Development 140: 1282-1291. PubMed ID: 23444356

    Olguin, P., Glavic, A. and Mlodzik, M. (2011). Intertissue mechanical stress affects Frizzled-mediated planar cell polarity in the Drosophila notum epidermis. Curr Biol 21: 236-242. PubMed ID: 21276726

  • David Glover Department of Genetics, University of Cambridge
    Santos, I. B., Wainman, A., Garrido-Maraver, J., Pires, V., Riparbelli, M. G., Kovacs, L., Callaini, G., Glover, D. M. and Tavares A, A. (2023). Mob4 is essential for spermatogenesis in Drosophila melanogaster. Genetics 224(4). PubMed ID: 37259670

    Sperling, A. L., Fabian, D. K., Garrison, E. and Glover, D. M. (2023). A genetic basis for facultative parthenogenesis in Drosophila. Curr Biol. PubMed ID: 37516115

    Kovacs, L., Fatalska, A. and Glover, D. M. (2022). Targeting Drosophila Sas6 to mitochondria reveals its high affinity for Gorab. Biol Open 11(11). PubMed ID: 36331102

    Braun, A. L., Meghini, F., Villa-Fombuena, G., Guermont, M., Fernandez-Martinez, E., Qian, Z., Dolores Martin-Bermudo, M., Gonzalez-Reyes, A., Glover, D. M. and Kimata, Y. (2021). The careful control of Polo kinase by APC/C-Ube2C ensures the intercellular transport of germline centrosomes during Drosophila oogenesis. Open Biol 11(6): 200371. PubMed ID: 34186008

    Lattao, R., Rangone, H., Llamazares, S. and Glover, D. M. (2021). Mauve/LYST limits fusion of lysosome-related organelles and promotes centrosomal recruitment of microtubule nucleating proteins. Dev Cell 56(7): 1000-1013.e1006. PubMed ID: 33725482

    Fatalska, A., Stepinac, E., Richter, M., Kovacs, L., Pietras, Z., Puchinger, M., Dong, G., Dadlez, M. and Glover, D. M. (2021). The dimeric Golgi protein Gorab binds to Sas6 as a monomer to mediate centriole duplication. Elife 10. PubMed ID: 33704067

    Karman, Z., Rethi-Nagy, Z., Abraham, E., Fabri-Ordogh, L., Csonka, A., Vilmos, P., Debski, J., Dadlez, M., Glover, D. M. and Lipinszki, Z. (2020). Novel perspectives of target-binding by the evolutionarily conserved PP4 phosphatase. Open Biol 10(12): 200343. PubMed ID: 33352067

    Panda, P., Kovacs, L., Dzhindzhev, N., Fatalska, A., Persico, V., Geymonat, M., Riparbelli, M. G., Callaini, G. and Glover, D. M. (2020). Tissue specific requirement of Drosophila Rcd4 for centriole duplication and ciliogenesis. J Cell Biol 219(8). PubMed ID: 32543652

    Kovacs, L., Chao-Chu, J., Schneider, S., Gottardo, M., Tzolovsky, G., Dzhindzhev, N. S., Riparbelli, M. G., Callaini, G. and Glover, D. M. (2018). Gorab is a Golgi protein required for structure and duplication of Drosophila centrioles. Nat Genet. PubMed ID: 29892014

    Dzhindzhev, N. S., Tzolovsky, G., Lipinszki, Z., Abdelaziz, M., Debski, J., Dadlez, M. and Glover, D. M. (2017). Two-step phosphorylation of Ana2 by Plk4 is required for the sequential loading of Ana2 and Sas6 to initiate procentriole formation. Open Biol 7(12). PubMed ID: 29263250

  • Tanja Godenschwege Max Planck Florida Institute for Neuroscience, Department of Biological Sciences, College of Science, Florida Atlantic University, Jupiter
    Ermanoska, B., Asselbergh, B., Morant, L., Petrovic-Erfurth, M. L., Hosseinibarkooie, S., Leitao-Gonçalves, R., Almeida-Souza, L., Bervoets, S., Sun, L., Lee, L., Atkinson, D., Khanghahi, A., Tournev, I., Callaerts, P., Verstreken, P., Yang, X. L., Wirth, B., Rodal, A. A., Timmerman, V., Goode, B. L., Godenschwege, T. A. and Jordanova, A. (2023). Tyrosyl-tRNA synthetase has a noncanonical function in actin bundling. Nat Commun 14(1): 999. PubMed ID: 36890170

    Nawaratne, V., Kudumala, S., Kakad, P. P. and Godenschwege, T. A. (2021). The conserved MASRPF motif in the Attractin homolog, Distracted, is required for association with Drosophila E3-ligase Mgrn1. MicroPubl Biol 2021. PubMed ID: 34235405

    Penserga, T., Kudumala, S. R., Poulos, R. and Godenschwege, T. A. (2019). A role for Drosophila Amyloid precursor protein in retrograde trafficking of L1-Type cell adhesion molecule Neuroglian. Front Cell Neurosci 13: 322. PubMed ID: 31354437

    Kakad, P. P., Penserga, T., Davis, B. P., Henry, B., Boerner, J., Riso, A., Pielage, J. and Godenschwege, T. A. (2018). An ankyrin-binding motif regulates nuclear levels of L1-type neuroglian and expression of the oncogene Myc in Drosophila neurons. J Biol Chem. PubMed ID: 30257867

    Augustin, H., McGourty, K., Allen, M. J., Madem, S. K., Adcott, J., Kerr, F., Wong, C. T., Vincent, A., Godenschwege, T., Boucrot, E. and Partridge, L. (2017). Reduced insulin signaling maintains electrical transmission in a neural circuit in aging flies. PLoS Biol 15(9): e2001655. PubMed ID: 28902870

    Lee, L. H. and Godenschwege, T. A. (2014). Structure-function analyses of tyrosine phosphatase PTP69D in giant fiber synapse formation of Drosophila. Mol Cell Neurosci 64C: 24-31. PubMed ID: 25433167

    Kudumala, S., Freund, J., Hortsch, M. and Godenschwege, T. A. (2013). Differential Effects of Human L1CAM Mutations on Complementing Guidance and Synaptic Defects in Drosophila melanogaster. PLoS One 8: e76974. PubMed ID: 24155914

    Mejia, M., Heghinian, M. D., Mari, F. and Godenschwege, T. A. (2013). New Tools for Targeted Disruption of Cholinergic Synaptic Transmission in Drosophila melanogaster. PLoS One 8: e64685. PubMed ID: 23737994

    Mejia, M., Heghinian, M. D., Busch, A., Mari, F. and Godenschwege, T. A. (2012). Paired nanoinjection and electrophysiology assay to screen for bioactivity of compounds using the Drosophila melanogaster giant fiber system. J Vis Exp. PubMed ID: 22525737

    Boerner, J. and Godenschwege, T. A. (2011). Whole mount preparation of the adult Drosophila ventral nerve cord for giant fiber dye injection. J Vis Exp. PubMed ID: 21673644

  • Dorothea Godt Dept. Cell and Systems Biology, University of Toronto
    Panchal, T., Chen, X., Alchits, E., Oh, Y., Poon, J., Kouptsova, J., Laski, F. A. and Godt, D. (2017). Specification and spatial arrangement of cells in the germline stem cell niche of the Drosophila ovary depend on the Maf transcription factor Traffic jam. PLoS Genet 13(5): e1006790. PubMed ID: 28542174

    Glowinski, C., Liu, R. H., Chen, X., Darabie, A. and Godt, D. (2014). Myosin VIIA regulates microvillus morphogenesis and interacts with cadherin Cad99C in Drosophila oogenesis. J Cell Sci. PubMed ID: 25236597

    Gunawan, F., Arandjelovic, M., Godt, D. (2013) The Maf factor Traffic jam both enables and inhibits collective cell migration in Drosophila oogenesis. Development. PubMed ID: 23720044

    Sarpal, R., Pellikka, M., Patel, R. R., Hui, F. Y., Godt, D. and Tepass, U. (2012). Mutational analysis supports a core role for Drosophila alpha-catenin in adherens junction function. J Cell Sci 125: 233-245. PubMed ID: 22266901

    Karpowicz, P., Pellikka, M., Chea, E., Godt, D., Tepass, U. and van der Kooy, D. (2009). The germline stem cells of Drosophila melanogaster partition DNA non-randomly. Eur J Cell Biol 88: 397-408. PubMed ID: 19395121

  • Michael Goldberg Department of Molecular Biology and Genetics, Cornell University
    Yamamoto, T. M., Blake-Hodek, K., Williams, B. C., Lewellyn, A. L., Goldberg, M. L. and Maller, J. L. (2011). Regulation of Greatwall kinase during Xenopus oocyte maturation. Mol Biol Cell 22: 2157-2164. PubMed ID: 21551066

    Wainman, A., Giansanti, M. G., Goldberg, M. L. and Gatti, M. (2012). The Drosophila RZZ complex - roles in membrane trafficking and cytokinesis. J Cell Sci 125: 4014-4025. PubMed ID: 22685323

    Kim, M.Y., Bucciarelli, E., Morton, D. G., Williams, B. C., Blake-Hodek, K., Pellacani, C., Von Stetina, J. R., Hu, X., Somma, M. P., Drummond-Barbosa, D. and Goldberg, M. L. (2012). Bypassing the Greatwall-Endosulfine pathway: plasticity of a pivotal cell-cycle regulatory module in Drosophila melanogaster and Caenorhabditis elegans. Genetics 191(4): 1181-97

    Yamamoto, T. M., Blake-Hodek, K., Williams, B. C., Lewellyn, A. L., Goldberg, M. L. and Maller, J. L. (2011). Regulation of Greatwall kinase during Xenopus oocyte maturation. Mol Biol Cell 22: 2157-2164. PubMed ID: 21551066

  • Lawrence Goldstein HHMI and Department of Cellular and Molecular Medicine, U. C. San Diego
    Almenar-Queralt, A., Kim, S. N., Benner, C., Herrera, C. M., Kang, D. E., Garcia-Bassets, I. and Goldstein, L. S. (2013). Presenilins Regulate Neurotrypsin Gene Expression and Neurotrypsin-Dependent Agrin Cleavage via CREB Modulation. J Biol Chem. PubMed ID: 24145027

    Duncan, J. E., Lytle, N. K., Zuniga, A. and Goldstein, L. S. (2013). The Microtubule Regulatory Protein Stathmin Is Required to Maintain the Integrity of Axonal Microtubules in Drosophila. PLoS One 8: e68324. PubMed ID: 23840848

    Henthorn, K. S., Roux, M. S., Herrera, C. and Goldstein, L. S. (2011). A role for kinesin heavy chain in controlling vesicle transport into dendrites in Drosophila. Mol Biol Cell 22: 4038-4046. PubMed ID: 21880894

    Falzone, T. L., Gunawardena, S., McCleary, D., Reis, G. F. and Goldstein, L. S. (2010). Kinesin-1 transport reductions enhance human tau hyperphosphorylation, aggregation and neurodegeneration in animal models of tauopathies. Hum Mol Genet 19: 4399-4408. PubMed ID: 20817925

  • Aaron Goldstrohm Biological Sciences, University of Minnesota, Minneapolis
    Arvola, R. M., Chang, C. T., Buytendorp, J. P., Levdansky, Y., Valkov, E., Freddolino, P. L. and Goldstrohm, A. C. (2019). Unique repression domains of Pumilio utilize deadenylation and decapping factors to accelerate destruction of target mRNAs. Nucleic Acids Res. PubMed ID: 31863588

    Qiu, C., Goldstrohm, A. C. and Tanaka Hall, T. M. (2019). Preparation of cooperative RNA recognition complexes for crystallographic structural studies. Methods Enzymol 623: 1-22. PubMed ID: 31239042

    Arvola, R. M., Weidmann, C. A., Tanaka Hall, T. M. and Goldstrohm, A. C. (2017). Combinatorial control of messenger RNAs by Pumilio, Nanos and Brain Tumor Proteins. RNA Biol 14(11): 1445-1456. PubMed ID: 28318367

  • Kent Golic Molecular Biology, University of Utah
    Hill, H., Bonser, D. and Golic, K. G. (2023). Dicentric chromosome breakage in Drosophila melanogaster is influenced by pericentric heterochromatin and occurs in non-conserved hotspots. Genetics. PubMed ID: 37010100

    Bhandari, J., Karg, T. and Golic, K. (2019). Homolog dependent repair following dicentric chromosome breakage in Drosophila melanogaster. Genetics. PubMed ID: 31053594

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

    Hill, H. and Golic, K. G. (2015). Preferential breakpoints in the recovery of broken dicentric chromosomes in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 26294667

    Chakraborty, R., Li, Y., Zhou, L. and Golic, K.G. (2015). Corp regulates P53 in Drosophila melanogaster via a negative feedback loop. PLoS Genet 11: e1005400. PubMed ID: 26230084

    Golic, K. G. (2013). RNA-guided nucleases: a new era for engineering the genomes of model and nonmodel organisms. Genetics 195: 303-308. PubMed ID: 24089463

    Titen, S. W., Lin, H. C., Bhandari, J. and Golic, K. G. (2014). Chk2 and p53 regulate the transmission of healed chromosomes in the Drosophila male germline. PLoS Genet 10: e1004130. PubMed ID: 24586185

    Xie, H. B., Cammarato, A., Rajasekaran, N. S., Zhang, H., Suggs, J. A., Lin, H. C., Bernstein, S. I., Benjamin, I. J. and Golic, K. G. (2013). The NADPH Metabolic Network Regulates Human alphaB-crystallin Cardiomyopathy and Reductive Stress in Drosophila melanogaster. PLoS Genet 9: e1003544. PubMed ID: 23818860

    Kurzhals, R. L., Titen, S. W., Xie, H. B. and Golic, K. G. (2011). Chk2 and p53 are haploinsufficient with dependent and independent functions to eliminate cells after telomere loss. PLoS Genet 7: e1002103. PubMed ID: 21655087

    Golic, M. M. and Golic, K. G. (2011). A simple and rapid method for constructing ring-X chromosomes in Drosophila melanogaster. Chromosoma 120: 159-164. PubMed ID: 21085980

  • Zhefeng Gong Faculty of Basic Medicine, Zhejiang University School of Medicine
    Gong, C., Ouyang, Z., Zhao, W., Wang, J., Li, K., Zhou, P., Zhao, T., Zheng, N. and Gong, Z. (2019). A neuronal pathway that commands deceleration in Drosophila larval light-avoidance. Neurosci Bull. PubMed ID: 30810958

    Zhao, W., Zhou, P., Gong, C., Ouyang, Z., Wang, J., Zheng, N. and Gong, Z. (2019). A disinhibitory mechanism biases Drosophila innate light preference. Nat Commun 10(1): 124. PubMed ID: 30631066

    Fei, Y., Zhu, D., Sun, Y., Gong, C., Huang, S. and Gong, Z. (2018). Repeated failure in reward pursuit alters innate Drosophila larval behaviors. Neurosci Bull. PubMed ID: 29951979

    Zhao, W., Gong, C., Ouyang, Z., Wang, P., Wang, J., Zhou, P., Zheng, N. and Gong, Z. (2017). Turns with multiple and single head cast mediate Drosophila larval light avoidance. PLoS One 12(7): e0181193. PubMed ID: 28700684

    Li, Q. and Gong, Z. (2015). Cold-sensing regulates Drosophila growth through insulin-producing cells. Nat Commun 6: 10083. PubMed ID: 26648410

    Guo, C., Du, Y., Yuan, D., Li, M., Gong, H., Gong, Z. and Liu, L. (2014). A conditioned visual orientation requires the ellipsoid body in Drosophila. Learn Mem 22: 56-63. PubMed ID: 25512578

    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 57: 391-402. PubMed ID: 24744088

    Liu, J., Gong, Z. and Liu, L. (2014). gamma-glutamyl transpeptidase 1 specifically suppresses green-light avoidance via GABAA receptors in Drosophila. J Neurochem 130: 408-418. PubMed ID: 24702462

  • Nicolas Gompel Faculty of Biology, LMU Munich
    Ling, L., Muhling, B., Jaenichen, R. and Gompel, N. (2023). Increased chromatin accessibility promotes the evolution of a transcriptional silencer in Drosophila. Sci Adv 9(7): eade6529. PubMed ID: 36800429

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

    Xin, Y., Le Poul, Y., Ling, L., Museridze, M., Muhling, B., Jaenichen, R., Osipova, E. and Gompel, N. (2020). Enhancer evolutionary co-option through shared chromatin accessibility input. Proc Natl Acad Sci U S A 117(34): 20636-20644. PubMed ID: 32778581

    Green, J. E., Cavey, M., Medina Caturegli, E., Aigouy, B., Gompel, N. and Prud'homme, B. (2019). Evolution of ovipositor length in Drosophila suzukii is driven by enhanced cell size expansion and anisotropic tissue reorganization. Curr Biol 29(12): 2075-2082.e2076. PubMed ID: 31178315

    Bracker, L. B., Schmid, C. A., Bolini, V. A., Holz, C. A., Prud'homme, B., Sirota, A. and Gompel, N. (2019). Quantitative and discrete evolutionary changes in the egg-laying behavior of single Drosophila females. Front Behav Neurosci 13: 118. PubMed ID: 31191270

    Bozek, M., Cortini, R., Storti, A. E., Unnerstall, U., Gaul, U. and Gompel, N. (2019). ATAC-seq reveals regional differences in enhancer accessibility during the establishment of spatial coordinates in the Drosophila blastoderm. Genome Res. PubMed ID: 30962180

    Hinaux, H., Bachem, K., Battistara, M., Rossi, M., Xin, Y., Jaenichen, R., Le Poul, Y., Arnoult, L., Kobler, J. M., Grunwald Kadow, I. C., Rodermund, L., Prud'homme, B. and Gompel, N. (2018). Revisiting the developmental and cellular role of the pigmentation gene yellow in Drosophila using a tagged allele. Dev Biol 438(2): 111-123. PubMed ID: 29634916

    Hussain, A., Pooryasin, A., Zhang, M., Loschek, L. F., La Fortezza, M., Friedrich, A. B., Blais, C. M., Ucpunar, H. K., Yepez, V. A., Lehmann, M., Gompel, N., Gagneur, J., Sigrist, S. J. and Grunwald Kadow, I. C. (2018). Inhibition of oxidative stress in cholinergic projection neurons fully rescues aging-associated olfactory circuit degeneration in Drosophila. Elife 7. PubMed ID: 29345616

    Karageorgi, M., Bracker, L. B., Lebreton, S., Minervino, C., Cavey, M., Siju, K. P., Grunwald Kadow, I. C., Gompel, N. and Prud'homme, B. (2017). Evolution of Multiple Sensory Systems Drives Novel Egg-Laying Behavior in the Fruit Pest Drosophila suzukii. Curr Biol 27(6): 847-853. PubMed ID: 28285999

  • Graydon Gonsalvez Cellular Biology and Anatomy, Medical College of Georgia, Augusta
    Neiswender, H., Baker, F. C., Veeranan-Karmegam, R., Allen, P. and Gonsalvez, G. B. (2023). dTtc1, a conserved tetratricopeptide repeat protein, is required for maturation of Drosophila egg chambers via its role in stabilizing electron transport chain components. Front Cell Dev Biol 11: 1148773. PubMed ID: 37333987

    Baker, F. C., Neiswender, H., Veeranan-Karmegam, R. and Gonsalvez, G. B. (2021). In vivo proximity biotin ligation identifies the interactome of Egalitarian, a Dynein cargo adaptor. Development 148(22). PubMed ID: 35020877

    Neiswender, H., Goldman, C. H., Veeranan-Karmegam, R. and Gonsalvez, G. B. (2021). Dynein light chain-dependent dimerization of Egalitarian is essential for maintaining oocyte fate in Drosophila. Dev Biol 478: 76-88. PubMed ID: 34181915

    Goldman, C. H., Neiswender, H., Baker, F., Veeranan-Karmegam, R., Misra, S. and Gonsalvez, G. B. (2021). Optimal RNA binding by Egalitarian, a Dynein cargo adaptor, is critical for maintaining oocyte fate in Drosophila. RNA Biol: 1-14. PubMed ID: 33904382

    Goldman, C. H., Neiswender, H., Veeranan-Karmegam, R. and Gonsalvez, G. B. (2019). The Egalitarian binding partners Dynein light chain and Bicaudal-D act sequentially to link mRNA to the Dynein motor. Development 146(15). PubMed ID: 31391195

    Veeranan-Karmegam, R., Boggupalli, D. P., Liu, G. and Gonsalvez, G. B. (2016). A new isoform of Drosophila non-muscle Tropomyosin 1 interacts with Kinesin-1 and functions in oskar mRNA localization. J Cell Sci 129: 4252-4264. PubMed ID: 27802167

    Sanghavi, P., Liu, G., Veeranan-Karmegam, R., Navarro, C. and Gonsalvez, G. B. (2016). Multiple roles for Egalitarian in polarization of the Drosophila egg chamber. Genetics [Epub ahead of print]. PubMed ID: 27017624

    Hicks, L., Liu, G., Ukken, F. P., Lu, S., Bollinger, K. E., O'Connor-Giles, K. and Gonsalvez, G. B. (2015). Depletion or over-expression of Sh3px1 results in dramatic changes in cell morphology. Biol Open. PubMed ID: 26459243

    Liu, G., Sanghavi, P., Bollinger, K. E., Perry, L., Marshall, B., Roon, P., Tanaka, T., Nakamura, A. and Gonsalvez, G. B. (2015). Efficient endocytic uptake and maturation in Drosophila oocytes requires Dynamitin/p50. Genetics. PubMed ID: 26265702

    Sanghavi, P., Laxani, S., Li, X., Bullock, S. L. and Gonsalvez, G. B. (2013). Dynein Associates with oskar mRNPs and Is Required For Their Efficient Net Plus-End Localization in Drosophila Oocytes. PLoS One 8: e80605. PubMed ID: 24244700

  • Cayetano González Institute for Research in Biomedicine, Barcelona
    Goupil, A., Heinen, J. P., Salame, R., Rossi, F., Reina, J., Pennetier, C., Simon, A., Skorski, P., Louzao, A., Bardin, A. J., Basto, R. and Gonzalez, C. (2022). Illuminati: a form of gene expression plasticity in Drosophila neural stem cells. Development 149(22). PubMed ID: 36399062

    Molnar, C., Louzao, A. and Gonzalez, C. (2020). Context-Dependent Tumorigenic Effect of Testis-Specific Mitochondrial Protein Tiny Tim 2 in Drosophila Somatic Epithelia. Cells 9(8). PubMed ID: 32781577

    Molnar, C., Heinen, J. P., Reina, J., Llamazares, S., Palumbo, E., Breschi, A., Gay, M., Villarreal, L., Vilaseca, M., Pollarolo, G. and Gonzalez, C. (2019). The histone code reader PHD finger protein 7 controls sex-linked disparities in gene expression and malignancy in Drosophila. Sci Adv 5(8): eaaw7965. PubMed ID: 31453329

    Reina, J., Gottardo, M., Riparbelli, M. G., Llamazares, S., Callaini, G. and Gonzalez, C. (2018). Centrobin is essential for C-tubule assembly and flagellum development in Drosophila melanogaster spermatogenesis. J Cell Biol. PubMed ID: 29712734

    Rossi, F., Stephan-Otto Attolini, C., Mosquera, J. L. and Gonzalez, C. (2018). Drosophila larval brain neoplasms present tumour-type dependent genome instability. G3 (Bethesda). PubMed ID: 29467187

    Rossi, F. and Gonzalez, C. (2015). Studying tumor growth in Drosophila using the tissue allograft method. Nat Protoc 10: 1525-1534. PubMed ID: 26357008

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

    Pampalona, J., Januschke, J., Sampaio, P. and Gonzalez, C. (2015). Time-lapse recording of centrosomes and other organelles in Drosophila neuroblasts. Methods Cell Biol 129: 301-315. PubMed ID: 26175445

    Gonzalez, C. (2013) Drosophila melanogaster: a model and a tool to investigate malignancy and identify new therapeutics. Nat Rev Cancer. PubMed ID: 23388617

    Januschke, J., Reina, J., Llamazares, S., Bertran, T., Rossi, F., Roig, J., Gonzalez, C. (2013) Centrobin controls mother-daughter centriole asymmetry in Drosophila neuroblasts. Nat Cell Biol. PubMed ID: 23354166

  • Josefa González Institute of Evolutionary Biology (CSIC-Universitat Pompeu Fabra), Barcelona, Spain
    Merenciano, M. and Gonzalez, J. (2023). The Interplay Between Developmental Stage and Environment Underlies the Adaptive Effect of a Natural Transposable Element Insertion. Mol Biol Evol 40(3). PubMed ID: 36811953

    Rech, G. E., Radio, S., Guirao-Rico, S., Aguilera, L., Horvath, V., Green, L., Lindstadt, H., Jamilloux, V., Quesneville, H. and Gonzalez, J. (2022). Population-scale long-read sequencing uncovers transposable elements associated with gene expression variation and adaptive signatures in Drosophila. Nat Commun 13(1): 1948. PubMed ID: 35413957

    Ullastres, A., Merenciano, M. and Gonzalez, J. (2021). Regulatory regions in natural transposable element insertions drive interindividual differences in response to immune challenges in Drosophila. Genome Biol 22(1): 265. PubMed ID: 34521452

    Bogaerts-Marquez, M., Guirao-Rico, S., Gautier, M. and Gonzalez, J. (2020). Temperature, rainfall and wind variables underlie environmental adaptation in natural populations of Drosophila melanogaster. Mol Ecol. PubMed ID: 33350518

    Kapun, M., Barron, M. G., Staubach, F..... Gonzalez, J. (2020). Genomic analysis of European Drosophila melanogaster populations reveals longitudinal structure, continent-wide selection, and previously unknown DNA viruses. Mol Biol Evol. PubMed ID: 32413142

    Salces-Ortiz, J., Vargas-Chavez, C., Guio, L., Rech, G. E. and Gonzalez, J. (2020). Transposable elements contribute to the genomic response to insecticides in Drosophila melanogaster. Philos Trans R Soc Lond B Biol Sci 375(1795): 20190341. PubMed ID: 32075557

    Villanueva-Canas, J. L., Horvath, V., Aguilera, L. and Gonzalez, J. (2019). Diverse families of transposable elements affect the transcriptional regulation of stress-response genes in Drosophila melanogaster. Nucleic Acids Res. PubMed ID: 31175824

    Rech, G. E., Bogaerts-Marquez, M., Barron, M. G., Merenciano, M., Villanueva-Canas, J. L., Horvath, V., Fiston-Lavier, A. S., Luyten, I., Venkataram, S., Quesneville, H., Petrov, D. A. and Gonzalez, J. (2019). Stress response, behavior, and development are shaped by transposable element-induced mutations in Drosophila. PLoS Genet 15(2): e1007900. PubMed ID: 30753202

    Lerat, E., Goubert, C., Guirao-Rico, S., Merenciano, M., Dufour, A. B., Vieira, C. and Gonzalez, J. (2018). Population specific dynamics and selection patterns of transposable element insertions in European natural populations. Mol Ecol. PubMed ID: 30506554

    Mateo, L., Rech, G. E. and Gonzalez, J. (2018). Genome-wide patterns of local adaptation in Western European Drosophila melanogaster natural populations. Sci Rep 8(1): 16143. PubMed ID: 30385770

    Guio, L., Vieira, C. and Gonzalez, J. (2018). Stress affects the epigenetic marks added by natural transposable element insertions in Drosophila melanogaster. Sci Rep 8(1): 12197. PubMed ID: 30111890

    Le Manh, H., Guio, L., Merenciano, M., Rovira, Q., Barrón, M.G. and González, J. (2017). Natural and laboratory mutations in kuzbanian are associated with zinc stress phenotypes in Drosophila melanogaster. Sci Rep 7: 42663. PubMed ID: 28218276

    Merenciano, M., Ullastres, A., de Cara, M. A., Barron, M. G. and Gonzalez, J. (2016). Multiple independent retroelement insertions in the promoter of a stress response gene have variable molecular and functional effects in Drosophila. PLoS Genet 12: e1006249. PubMed ID: 27517860

  • Marcos Gonzalez-Gaitan Biochemistry Department, University of Geneva
    Daeden, A., Mietke, A., Derivery, E., Seum, C., Julicher, F. and Gonzalez-Gaitan, M. (2023). Polarized branched Actin modulates cortical mechanics to produce unequal-size daughters during asymmetric division. Nat Cell Biol 25(2): 235-245. PubMed ID: 36747081

    Derivery, E., Seum, C., Daeden, A., Loubery, S., Holtzer, L., Julicher, F. and Gonzalez-Gaitan, M. (2015). Polarized endosome dynamics by spindle asymmetry during asymmetric cell division. Nature 528: 280-285. PubMed ID: 26659188

    Romanova-Michaelides, M., Aguilar-Hidalgo, D., Julicher, F. and Gonzalez-Gaitan, M. (2015). The wing and the eye: a parsimonious theory for scaling and growth control? Wiley Interdiscip Rev Dev Biol. PubMed ID: 26108346

    Montagne, C. and Gonzalez-Gaitan, M. (2014). Sara endosomes and the asymmetric division of intestinal stem cells. Development 141: 2014-2023. PubMed ID: 24803650

    Wartlick, O., Julicher, F. and Gonzalez-Gaitan, M. (2014). Growth control by a moving morphogen gradient during Drosophila eye development. Development 141: 1884-1893. PubMed ID: 24757005

    Loubery, S. and Gonzalez-Gaitan, M. (2014). Monitoring notch/delta endosomal trafficking and signaling in Drosophila. Methods Enzymol 534: 301-321. PubMed ID: 24359961

    Kicheva, A., Holtzer, L., Wartlick, O., Schmidt, T., Gonzalez-Gaitan, M. (2013) Quantitative imaging of morphogen gradients in Drosophila imaginal discs. Cold Spring Harb Protoc 2013. PubMed ID: 23637364

    Wartlick, O., Mumcu, P., Kicheva, A., Bittig, T., Seum, C., Julicher, F. and Gonzalez-Gaitan, M. (2011). Dynamics of Dpp signaling and proliferation control. Science 331: 1154-1159. PubMed ID: 21385708

    Assaker, G., Ramel, D., Wculek, S. K., Gonzalez-Gaitan, M. and Emery, G. (2010). Spatial restriction of receptor tyrosine kinase activity through a polarized endocytic cycle controls border cell migration. Proc Natl Acad Sci U S A 107: 22558-22563. PubMed ID: 21149700

  • Acaimo González Reyes Centro Andaluz de Biología del Desarrollo, Sevilla, Spain
    Villa-Fombuena, G., Lobo-Pecellin, M., Marin-Menguiano, M., Rojas-Rios, P. and Gonzalez-Reyes, A. (2021). Live imaging of the Drosophila ovarian niche shows spectrosome and centrosome dynamics during asymmetric germline stem cell division. Development. PubMed ID: 34370012

    Diaz-Torres, A., Rosales-Nieves, A. E., Pearson, J. R., Santa-Cruz Mateos, C., Marin-Menguiano, M., Marshall, O. J., Brand, A. H. and Gonzalez-Reyes, A. (2021). Stem cell niche organization in the Drosophila ovary requires the ECM component Perlecan. Curr Biol. PubMed ID: 33621481

    Lobo-Pecellin, M., Marin-Menguiano, M. and Gonzalez-Reyes, A. (2019). mastermind regulates niche ageing independently of the Notch pathway in the Drosophila ovary. Open Biol 9(11): 190127. PubMed ID: 31744422

    Diaz de la Loza, M. C., Diaz-Torres, A., Zurita, F., Rosales-Nieves, A. E., Moeendarbary, E., Franze, K., Martin-Bermudo, M. D. and Gonzalez-Reyes, A. (2017). Laminin levels regulate tissue migration and anterior-posterior polarity during egg morphogenesis in Drosophila. Cell Rep 20(1): 211-223. PubMed ID: 28683315

    Valencia-Exposito, A., Grosheva, I., Miguez, D. G., Gonzalez-Reyes, A. and Martin-Bermudo, M. D. (2016). Myosin light-chain phosphatase regulates basal actomyosin oscillations during morphogenesis. Nat Commun 7: 10746. PubMed ID: 26888436

    Pearson, J. R., Zurita, F., Tomas-Gallardo, L., Diaz-Torres, A., Diaz de la Loza Mdel, C., Franze, K., Martin-Bermudo, M. D. and Gonzalez-Reyes, A. (2016). ECM-Regulator timp Is Required for Stem Cell Niche Organization and Cyst Production in the Drosophila Ovary. PLoS Genet 12: e1005763. PubMed ID: 26808525

    Rojas-Ríos, P., Guerrero, I. and González-Reyes, A. (2012). Cytoneme-mediated delivery of hedgehog regulates the expression of bone morphogenetic proteins to maintain germline stem cells in Drosophila. PLoS Biol 10(4): e1001298. PubMed ID: 22509132

    Cobreros, L., Fernández-Miñán, A., López-Schier, H., González-Reyes A. and Martín-Bermudo, M. D. (2010). The Ste-20 kinase misshapen regulates the invasive behaviour of epithelial cells in the Drosophila ovary. EMBO Reports 11(12): 943-949. PubMed ID: 21102643

  • Julie Goodliffe University of North Carolina at Charlotte
    Daneshvar, K., Nath, S., Khan, A., Shover, W., Richardson, C., Goodliffe, J. M. (2013) MicroRNA miR-308 regulates dMyc through a negative feedback loop in Drosophila. Biol Open 2: 1-9. PubMed ID: 23336071

    Daneshvar, K., Khan, A. and Goodliffe, J. M. (2011). Myc localizes to histone locus bodies during replication in Drosophila. PLoS One 6: e23928. PubMed ID: 21886841

    Khan, A., Shover, W. and Goodliffe, J. M. (2009). Su(z)2 antagonizes auto-repression of Myc in Drosophila, increasing Myc levels and subsequent trans-activation. gPLoS One 4: e5076. PubMed ID: 19333393

  • Stephen Goodwin University of Oxford
    Neville, M. C., Eastwood, A., Allen, A. M., de Haan, A., Nojima, T. and Goodwin, S. F. (2021). Generation and characterization of fruitless P1 promoter mutant in Drosophila melanogaster. J Neurogenet: 1-10. PubMed ID: 34338589

    Nojima, T., Rings, A., Allen, A. M., Otto, N., Verschut, T. A., Billeter, J. C., Neville, M. C. and Goodwin, S. F. (2021). A sex-specific switch between visual and olfactory inputs underlies adaptive sex differences in behavior. Curr Biol. PubMed ID: 33508219

    Zhang, Y., Ng, R., Neville, M. C., Goodwin, S. F. and Su, C. Y. (2020). Distinct Roles and Synergistic Function of Fru(M) Isoforms in Drosophila Olfactory Receptor Neurons. Cell Rep 33(11): 108516. PubMed ID: 33326795

    Allen, A. M., Neville, M. C., Birtles, S., Croset, V., Treiber, C. D., Waddell, S. and Goodwin, S. F. (2020). A single-cell transcriptomic atlas of the adult Drosophila ventral nerve cord. Elife 9. PubMed ID: 32314735

    Bruggemeier, B., Porter, M. A., Vigoreaux, J. O. and Goodwin, S. F. (2018). Female Drosophila melanogaster respond to song-amplitude modulations. Biol Open. PubMed ID: 29666051

    Bath, E., Bowden, S., Peters, C., Reddy, A., Tobias, J. A., Easton-Calabria, E., Seddon, N., Goodwin, S. F. and Wigby, S. (2017). Sperm and sex peptide stimulate aggression in female Drosophila. Nat Ecol Evol 1(6): 0154. PubMed ID: 28580431

    Pavlou, H. J., Lin, A. C., Neville, M. C., Nojima, T., Diao, F., Chen, B. E., White, B. H. and Goodwin, S. F. (2016). Neural circuitry coordinating male copulation. Elife 5. PubMed ID: 27855059

    Rezaval, C., Pattnaik, S., Pavlou, H. J., Nojima, T., Bruggemeier, B., D'Souza, L. A., Dweck, H. K. and Goodwin, S. F. (2016). Activation of latent courtship circuitry in the brain of Drosophila females induces male-like behaviors. Curr Biol 26: 2508-2515. PubMed ID: 27568592

    Nojima, T., Neville, M. C. and Goodwin, S. F. (2014). Fruitless isoforms and target genes specify the sexually dimorphic nervous system underlying Drosophila reproductive behavior. Fly (Austin) 8. PubMed ID: 24813187

    Rezaval, C., Nojima, T., Neville, M. C., Lin, A. C. and Goodwin, S. F. (2014). Sexually Dimorphic Octopaminergic Neurons Modulate Female Postmating Behaviors in Drosophila. Curr Biol. PubMed ID: 24631243

    Neville, M. C., Nojima, T., Ashley, E., Parker, D. J., Walker, J., Southall, T., Van de Sande, B., Marques, A. C., Fischer, B., Brand, A. H., Russell, S., Ritchie, M. G., Aerts, S. and Goodwin, S. F. (2014). Male-Specific Fruitless Isoforms Target Neurodevelopmental Genes to Specify a Sexually Dimorphic Nervous System. Curr Biol. PubMed ID: 24440396

  • Martin Göpfert Dept. of Cellular Neurobiology, Göttingen
    Spalthoff, C., Salgado, V. L., Balu, N., David, M. D., Hehlert, P., Huang, H., Jones, J. E., Kandasamy, R., Knudsen, G. A., Lelito, K. R., Machamer, J. B., Nesterov, A., Tomalski, M., Wahl, G. D., Wedel, B. J. and Gopfert, M. C. (2023). The Novel Pyridazine Pyrazolecarboxamide Insecticide Dimpropyridaz Inhibits Chordotonal Organ Function Upstream of Trpv Channels. Pest Manag Sci. PubMed ID: 36622360

    Katana, R., Guan, C., Zanini, D., Larsen, M. E., Giraldo, D., Geurten, B. R. H., Schmidt, C. F., Britt, S. G. and Gopfert, M. C. (2019). Chromophore-independent roles of opsin apoproteins in Drosophila mechanoreceptors. Curr Biol 29(17): 2961-2969.e2964. PubMed ID: 31447373

    Zanini, D., Giraldo, D., Warren, B., Katana, R., Andres, M., Reddy, S., Pauls, S., Schwedhelm-Domeyer, N., Geurten, B. R. H. and Gopfert, M. C. (2018). Proprioceptive Opsin Functions in Drosophila Larval Locomotion. Neuron. PubMed ID: 29551493

    Prahlad, A., Spalthoff, C., Kong, D., Grosshans, J., Gopfert, M. C. and Schmidt, C. F. (2017). Mechanical properties of a Drosophila larval chordotonal organ. Biophys J 113(12): 2796-2804. PubMed ID: 29262372

    Corthals, K., Heukamp, A. S., Kossen, R., Grosshennig, I., Hahn, N., Gras, H., Gopfert, M. C., Heinrich, R. and Geurten, B. R. H. (2017). Neuroligins Nlg2 and Nlg4 Affect Social Behavior in Drosophila melanogaster. Front Psychiatry 8: 113. PubMed ID: 28740469

    Guo, Y., Wang, Y., Zhang, W., Meltzer, S., Zanini, D., Yu, Y., Li, J., Cheng, T., Guo, Z., Wang, Q., Jacobs, J. S., Sharma, Y., Eberl, D. F., Gopfert, M. C., Jan, L. Y., Jan, Y. N. and Wang, Z. (2016). Transmembrane channel-like (tmc) gene regulates Drosophila larval locomotion. Proc Natl Acad Sci U S A. PubMed ID: 27298354

    Karak, S., Jacobs, J. S., Kittelmann, M., Spalthoff, C., Katana, R., Sivan-Loukianova, E., Schon, M. A., Kernan, M. J., Eberl, D. F. and Gopfert, M. C. (2015). Diverse roles of axonemal dyneins in Drosophila auditory neuron function and mechanical amplification in hearing. Sci Rep 5: 17085. PubMed ID: 26608786

    Zhang, W., Cheng, L.E., Kittelmann, M., Li, J., Petkovic, M., Cheng, T., Jin, P., Guo, Z., Göpfert, M.C., Jan, L.Y. and Jan, Y.N. (2015). Ankyrin repeats convey force to gate the NOMPC mechanotransduction channel. Cell 162: 1391-1403. PubMed ID: 26359990

    Nesterov, A., Spalthoff, C., Kandasamy, R., Katana, R., Rankl, N. B., Andres, M., Jahde, P., Dorsch, J. A., Stam, L. F., Braun, F. J., Warren, B., Salgado, V. L. and Gopfert, M. C. (2015). TRP Channels in Insect Stretch Receptors as Insecticide Targets. Neuron 86: 665-671. PubMed ID: 25950634

    Albert, J. T. and Gopfert, M. C. (2015). Hearing in Drosophila. Curr Opin Neurobiol 34C: 79-85. PubMed ID: 25710304

    Geurten, B. R., Jahde, P., Corthals, K. and Gopfert, M. C. (2014). Saccadic body turns in walking Drosophila. Front Behav Neurosci 8: 365. PubMed ID: 25386124

  • Michael Gordon Department of Zoology, University of British Columbia, Vancouver
    McDowell, S. A. T., Stanley, M. and Gordon, M. D. (2022). A molecular mechanism for high salt taste in Drosophila. Curr Biol. PubMed ID: 35772408

    Musso, P. Y., Junca, P. and Gordon, M. D. (2021). A neural circuit linking two sugar sensors regulates satiety-dependent fructose drive in Drosophila. Sci Adv 7(49): eabj0186. PubMed ID: 34851668

    Stanley, M., Ghosh, B., Weiss, Z. F., Christiaanse, J. and Gordon, M. D. (2021). Mechanisms of lactic acid gustatory attraction in Drosophila. Curr Biol. PubMed ID: 34197729

    Lau, C. K. S., Jelen, M. and Gordon, M. D. (2021). A closed-loop optogenetic screen for neurons controlling feeding in Drosophila. G3 (Bethesda) 11(5). PubMed ID: 33714999

    Musso, P. Y., Junca, P., Jelen, M., Feldman-Kiss, D., Zhang, H., Chan, R. C. and Gordon, M. D. (2019). Closed-loop optogenetic activation of peripheral or central neurons modulates feeding in freely moving Drosophila. Elife 8. PubMed ID: 31322499

    Jaeger, A. H., Stanley, M., Weiss, Z. F., Musso, P. Y., Chan, R. C., Zhang, H., Feldman-Kiss, D. and Gordon, M. D. (2018). A complex peripheral code for salt taste in Drosophila. Elife 7. PubMed ID: 30307393

    LeDue, E.E., Mann, K., Koch, E., Chu, B., Dakin, R. and Gordon, M.D. (2016). Starvation-induced depotentiation of bitter taste in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 27720624

    LeDue, E. E., Chen, Y. C., Jung, A. Y., Dahanukar, A. and Gordon, M. D. (2015). Pharyngeal sense organs drive robust sugar consumption in Drosophila. Nat Commun 6: 6667. PubMed ID: 25807033

    Chu, B., Chui, V., Mann, K. and Gordon, M. D. (2014). Presynaptic Gain Control Drives Sweet and Bitter Taste Integration in Drosophila. Curr Biol. PubMed ID: 25131672

    Pool, A. H., Kvello, P., Mann, K., Cheung, S. K., Gordon, M. D., Wang, L. and Scott, K. (2014). Four GABAergic interneurons impose feeding restraint in Drosophila. Neuron 83: 164-177. PubMed ID: 24991960

    Mann, K., Gordon, M. D. and Scott, K. (2013). A pair of interneurons influences the choice between feeding and locomotion in Drosophila. Neuron 79: 754-765. PubMed ID: 23972600

  • Nicole Gorfinkiel Haim Centro De Biologia Molecular Severo Ochoa, Madrid
    Duque, J. and Gorfinkiel, N. (2016). Integration of actomyosin contractility with cell-cell adhesion during dorsal closure. Development. PubMed ID: 27836966

    Jurado, J., de Navascues, J. and Gorfinkiel, N. (2016). α-Catenin stabilises Cadherin-Catenin complexes and modulates actomyosin dynamics to allow pulsatile apical contraction. J Cell Sci [Epub ahead of print]. PubMed ID: 27831494

    Duque, J. and Gorfinkiel, N. (2016). Integration of actomyosin contractility with cell-cell adhesion during dorsal closure. Development [Epub ahead of print]. PubMed ID: 27836966

    Machado, P. F., Duque, J., Etienne, J., Martinez-Arias, A., Blanchard, G. B. and Gorfinkiel, N. (2015). Emergent material properties of developing epithelial tissues. BMC Biol 13: 98. PubMed ID: 26596771

    Fischer, S. C., Blanchard, G. B., Duque, J., Adams, R. J., Arias, A. M., Guest, S. D. and Gorfinkiel, N. (2014). Contractile and mechanical properties of epithelia with perturbed actomyosin dynamics. PLoS One 9: e95695. PubMed ID: 24759936

    Callejo, A., Bilioni, A., Mollica, E., Gorfinkiel, N., Andres, G., Ibanez, C., Torroja, C., Doglio, L., Sierra, J. and Guerrero, I. (2011). Dispatched mediates Hedgehog basolateral release to form the long-range morphogenetic gradient in the Drosophila wing disk epithelium. Proc Natl Acad Sci U S A 108: 12591-12598. PubMed ID: 21690386

    Mateus, A. M., Gorfinkiel, N., Schamberg, S. and Martinez Arias, A. (2011). Endocytic and recycling endosomes modulate cell shape changes and tissue behaviour during morphogenesis in Drosophila. PLoS One 6: e18729. PubMed ID: 21533196

  • Maureen Gorman Department of Biochemistry and Molecular Biophysic, Manhattan, KS
    Weber, J. J., Brummett, L. M., Coca, M. E., Tabunoki, H., Kanost, M. R., Ragan, E. J., Park, Y. and Gorman, M. J. (2022). Phenotypic analyses, protein localization, and bacteriostatic activity of Drosophila melanogaster transferrin-1. Insect Biochem Mol Biol 147: 103811. PubMed ID: 35781032

    Weber, J. J., Kanost, M. R. and Gorman, M. J. (2020). Iron binding and release properties of transferrin-1 from Drosophila melanogaster and Manduca sexta: Implications for insect iron homeostasis. Insect Biochem Mol Biol 125: 103438. PubMed ID: 32735914

    Zhang, R., Miner, J. J., Gorman, M. J., Rausch, K., Ramage, H., White, J. P., Zuiani, A., Zhang, P., Fernandez, E., Zhang, Q., Dowd, K. A., Pierson, T. C., Cherry, S. and Diamond, M. S. (2016). A CRISPR screen defines a signal peptide processing pathway required by flaviviruses. Nature 535(7610): 164-168. PubMed ID: 27383988

  • Sharon Gorski British Columbia Genome Sciences Center, Vancouver
    Choutka, C., DeVorkin, L., Go, N. E., Hou, Y. C., Moradian, A., Morin, G. B. and Gorski, S. M. (2017). Hsp83 loss suppresses proteasomal activity resulting in an upregulation of caspase-dependent compensatory autophagy. Autophagy: 1-17. PubMed ID: 28806103

    Chittaranjan, S., Xu, J., Kuzyk, M., Dullat, H. K., Wilton, J., DeVorkin, L., Lebovitz, C., Morin, G. B., Marra, M. A. and Gorski, S. M. (2015). The Drosophila TIPE family member Sigmar interacts with the Ste20-like kinase Misshapen and modulates JNK signaling, cytoskeletal remodeling and autophagy. Biol Open. PubMed ID: 25836674

    DeVorkin, L. and Gorski, S. M. (2014). Genetic manipulation of autophagy in the Drosophila ovary. Cold Spring Harb Protoc 2014: pdb prot080358. PubMed ID: 25183818

    DeVorkin, L. and Gorski, S. M. (2014). Monitoring Autophagy in Drosophila Using Fluorescent Reporters in the UAS-GAL4 System. Cold Spring Harb Protoc 2014: pdb prot080341. PubMed ID: 25183817

    DeVorkin, L. and Gorski, S. M. (2014). Monitoring Autophagic Flux Using Ref(2)P, the Drosophila p62 Ortholog. Cold Spring Harb Protoc 2014: pdb prot080333. PubMed ID: 25183816

    DeVorkin, L. and Gorski, S. M. (2014). LysoTracker Staining to Aid in Monitoring Autophagy in Drosophila. Cold Spring Harb Protoc 2014: pdb prot080325. PubMed ID: 25183815

    DeVorkin, L., Go, N. E., Hou, Y. C., Moradian, A., Morin, G. B. and Gorski, S. M. (2014). The Drosophila effector caspase Dcp-1 regulates mitochondrial dynamics and autophagic flux via SesB. J Cell Biol 205: 477-492. PubMed ID: 24862573

  • Gohta Goshima Division of Biological Sciences, Nagoya University
    Edzuka, T. and Goshima, G. (2018). Drosophila kinesin-8 stabilizes the kinetochore-microtubule interaction. J Cell Biol. PubMed ID: 30538142

    Moriwaki, T. and Goshima, G. (2016). Five factors can reconstitute all three phases of microtubule polymerization dynamics. J Cell Biol 215: 357-368. PubMed ID: 27799364

    Ito, A. and Goshima, G. (2015). Microcephaly protein Asp focuses the minus ends of spindle microtubules at the pole and within the spindle. J Cell Biol 211: 999-1009. PubMed ID: 26644514

    Moutinho-Pereira, S., Stuurman, N., Afonso, O., Hornsveld, M., Aguiar, P., Goshima, G., Vale, R. D. and Maiato, H. (2013). Genes involved in centrosome-independent mitotic spindle assembly in Drosophila S2 cells. Proc Natl Acad Sci U S A 110: 19808-19813. PubMed ID: 24255106

    Li, W., Moriwaki, T., Tani, T., Watanabe, T., Kaibuchi, K. and Goshima, G. (2012). Reconstitution of dynamic microtubules with Drosophila XMAP215, EB1, and Sentin. J Cell Biol 199: 849-862. PubMed ID: 23185033

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

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

    Folmsbee, S. S., Morales-Nebreda, L., Van Hengel, J., Tyberghein, K., Van Roy, F., Budinger, G. R., Bryce, P. J. and Gottardi, C. J. (2015). The cardiac protein alphaT-catenin contributes to chemical-induced asthma. Am J Physiol Lung Cell Mol Physiol 308: L253-258. PubMed ID: 25480337

    Valenti, F., Ibetti, J., Komiya, Y., Baxter, M., Lucchese, A. M., Derstine, L., Covaciu, C., Rizzo, V., Vento, R., Russo, G., Macaluso, M., Cotelli, F., Castiglia, D., Gottardi, C. J., Habas, R., Giordano, A. and Bellipanni, G. (2015). The Increase in Maternal Expression of axin1 and axin2 Contribute to the Zebrafish Mutant Ichabod Ventralized Phenotype. J Cell Biochem 116: 418-430. PubMed ID: 25335865

    McEwen, A. E., Maher, M. T., Mo, R. and Gottardi, C. J. (2014). E-cadherin phosphorylation occurs during its biosynthesis to promote its cell surface stability and adhesion. Mol Biol Cell 25: 2365-2374. PubMed ID: 24966173


  • Alex Gould MRC, Mill Hill, London
    Lubojemska, A., Stefana, M. I., Sorge, S., Bailey, A. P., Lampe, L., Yoshimura, A., Burrell, A., Collinson, L. and Gould, A. P. (2021). Adipose triglyceride lipase protects renal cell endocytosis in a Drosophila dietary model of chronic kidney disease. PLoS Biol 19(5): e3001230. PubMed ID: 33945525

    Froldi, F., Pachnis, P., Szuperak, M., Costas, O., Fernando, T., Gould, A. P. and Cheng, L. Y. (2019). Histidine is selectively required for the growth of Myc-dependent dedifferentiation tumours in the Drosophila CNS. Embo j. PubMed ID: 30804004

    Sawala, A. and Gould, A. P. (2018). Sex-lethal in neurons controls female body growth in Drosophila. Fly (Austin): 1-9. PubMed ID: 30126340

    Obata, F., Fons, C. O. and Gould, A. P. (2018). Early-life exposure to low-dose oxidants can increase longevity via microbiome remodelling in Drosophila. Nat Commun 9(1): 975. PubMed ID: 29515102

    Stefana, M. I., Driscoll, P. C., Obata, F., Pengelly, A. R., Newell, C. L., MacRae, J. I. and Gould, A. P. (2017). Developmental diet regulates Drosophila lifespan via lipid autotoxins. Nat Commun 8(1): 1384. PubMed ID: 29123106

    Sawala, A. and Gould, A. P. (2017). The sex of specific neurons controls female body growth in Drosophila. PLoS Biol 15(10): e2002252. PubMed ID: 28976974

    Cinnamon, E., Makki, R., Sawala, A., Wickenberg, L.P., Blomquist, G.J., Tittiger, C., Paroush, Z. and Gould, A.P. (2016). Spidey/Kar regulates oenocyte growth via PI3-kinase signaling. PLoS Genet 12: e1006154. PubMed ID: 27500738

    Bailey, A. P., Koster, G., Guillermier, C., Hirst, E. M., MacRae, J. I., Lechene, C. P., Postle, A. D. and Gould, A. P. (2015). Antioxidant Role for Lipid Droplets in a Stem Cell Niche of Drosophila. Cell 163: 340-353. PubMed ID: 26451484

    Ragan, T. J., Bailey, A. P., Gould, A. P. and Driscoll, P. C. (2013). Volume determination with two standards allows absolute quantification and improved chemometric analysis of metabolites by NMR from sub-microlitre samples. Anal Chem. PubMed ID: 24251761

    Cheng, L. Y., Bailey, A. P., Leevers, S. J., Ragan, T. J., Driscoll, P. C. and Gould, A. P. (2011). Anaplastic lymphoma kinase spares organ growth during nutrient restriction in Drosophila. Cell 146: 435-447. PubMed ID: 21816278

  • Shubha Govind Biology Department, City College of New York
    Heavner, M. E., Ramroop, J., Gueguen, G., Ramrattan, G., Dolios, G., Scarpati, M., Kwiat, J., Bhattacharya, S., Wang, R., Singh, S. and Govind, S. (2017). Novel organelles with elements of bacterial and eukaryotic secretion systems weaponize parasites of Drosophila. Curr Biol 27(18): 2869-2877.e2866. PubMed ID: 28889977

    Heavner, M. E., Hudgins, A. D., Rajwani, R., Morales, J. and Govind, S. (2014). Harnessing the natural -parasitoid model for integrating insect immunity with functional venomics. Curr Opin Insect Sci 6: 61-67. PubMed ID: 25642411

    Small, C., Ramroop, J., Otazo, M., Huang, L. H., Saleque, S. and Govind, S. (2013). An Unexpected Link Between Notch Signaling and ROS in Restricting the Differentiation of Hematopoietic Progenitors in Drosophila. Genetics. PubMed ID: 24318532

    Heavner, M. E., Gueguen, G., Rajwani, R., Pagan, P. E., Small, C. and Govind, S. (2013). Partial venom gland transcriptome of a Drosophila parasitoid wasp, Leptopilina heterotoma, reveals novel and shared bioactive profiles with stinging Hymenoptera. Gene 526: 195-204. PubMed ID: 23688557

    Kalamarz, M. E., Paddibhatla, I., Nadar, C. and Govind, S. (2012). Sumoylation is tumor-suppressive and confers proliferative quiescence to hematopoietic progenitors in Drosophila melanogaster larvae. Biol Open 1: 161-172. PubMed ID: 23213407

    Gueguen, G., Onemola, B. and Govind, S. (2012). Association of a new Wolbachia strain with, and its effects on, Leptopilina victoriae, a virulent wasp parasitic to Drosophila spp. Appl Environ Microbiol 78: 5962-5966. PubMed ID: 22685158

  • Yacine Graba Development Biology Institute of Marseille Luminy
    Barthez, M., Poplineau, M., Elrefaey, M., Caruso, N., Graba, Y. and Saurin, A. J. (2020). Human ZKSCAN3 and Drosophila M1BP are functionally homologous transcription factors in autophagy regulation. Sci Rep 10(1): 9653. PubMed ID: 32541927

    Rinaldi, L., Saurin, A. J. and Graba, Y. (2018). Fattening the perspective of Hox protein specificity through SLiMming. Int J Dev Biol 62(11-12): 755-766. PubMed ID: 30604845

    Zouaz, A., Auradkar, A., Delfini, M. C., Macchi, M., Barthez, M., Ela Akoa, S., Bastianelli, L., Xie, G., Deng, W. M., Levine, S. S., Graba, Y. and Saurin, A. J. (2017). The Hox proteins Ubx and AbdA collaborate with the transcription pausing factor M1BP to regulate gene transcription. EMBO J [Epub ahead of print]. PubMed ID: 28871058

    Banreti, A., Hudry, B., Sass, M., Saurin, A. J. and Graba, Y. (2013). Hox Proteins Mediate Developmental and Environmental Control of Autophagy. Dev Cell. PubMed ID: 24389064

    Sambrani, N., Pradel, J. and Graba, Y. (2013). Distinct genetic requirements for BX-C mediated specification of abdominal denticles. Dev Dyn. PubMed ID: 24155218

    Sambrani, N., Hudry, B., Maurel-Zaffran, C., Zouaz, A., Mishra, R., Merabet, S., Graba, Y. (2013) Distinct Molecular Strategies for Hox-Mediated Limb Suppression in Drosophila: From Cooperativity to Dispensability/Antagonism in TALE Partnership. PLoS Genet 9: e1003307. PubMed ID: 23505377

    Hudry, B., Remacle, S., Delfini, M. C., Rezsohazy, R., Graba, Y. and Merabet, S. (2012). Hox proteins display a common and ancestral ability to diversify their interaction mode with the PBC class cofactors. PLoS Biol 10: e1001351. PubMed ID: 22745600

    Hainaut, M., Sagnier, T., Berenger, H., Pradel, J., Graba, Y. and Miotto, B. (2012). The MYST-containing protein Chameau is required for proper sensory organ specification during Drosophila thorax morphogenesis. PLoS One 7: e32882. PubMed ID: 22412942

    Merabet, S., Litim-Mecheri, I., Karlsson, D., Dixit, R., Saadaoui, M., Monier, B., Brun, C., Thor, S., Vijayraghavan, K., Perrin, L., Pradel, J. and Graba, Y. (2011). Insights into Hox protein function from a large scale combinatorial analysis of protein domains. PLoS Genet 7: e1002302. PubMed ID: 22046139

  • Caroline Grabbe Department of Molecular Biology, Umea University
    Khoshnood, B., Dacklin, I. and Grabbe, C. (2017). A proteomics approach to identify targets of the ubiquitin-like molecule Urm1 in Drosophila melanogaster. PLoS One 12(9): e0185611. PubMed ID: 28953965

    Khoshnood, B., Dacklin, I. and Grabbe, C. (2015). Urm1: an essential regulator of JNK signaling and oxidative stress in Drosophila melanogaster. Cell Mol Life Sci. PubMed ID: 26715182

    Shirinian, M., Kambris, Z., Hamadeh, L., Grabbe, C., Journo, C., Mahieux, R. and Bazarbachi, A. (2015). A Transgenic Drosophila melanogaster Model To Study Human T-Lymphotropic Virus Oncoprotein Tax-1-Driven Transformation In Vivo. J Virol 89: 8092-8095. PubMed ID: 25995252

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

  • Jonathan Graff Department of Developmental Biology, Southwestern Medical Center, Dallas
    Ugrankar, R., Theodoropoulos, P., Akdemir, F., Henne, W. M. and Graff, J. M. (2018). Circulating glucose levels inversely correlate with Drosophila larval feeding through insulin signaling and SLC5A11. Commun Biol 1: 110. PubMed ID: 30271990
    Ugrankar, R., Berglund, E., Akdemir, F., Tran, C., Kim, M.S., Noh, J., Schneider, R., Ebert, B. and Graff, J.M. (2015). Drosophila glucome screening identifies Ck1alpha as a regulator of mammalian glucose metabolism. Nat Commun 6: 7102. PubMed ID: 25994086

    Stenesen, D., Suh, J. M., Seo, J., Yu, K., Lee, K. S., Kim, J. S., Min, K. J., Graff, J. M. (2013) Adenosine Nucleotide Biosynthesis and AMPK Regulate Adult Life Span and Mediate the Longevity Benefit of Caloric Restriction in Flies. Cell Metab 17: 101-112. PubMed ID: 23312286

    Seo, J., Fortuno, E. S., 3rd, Suh, J. M., Stenesen, D., Tang, W., Parks, E. J., Adams, C. M., Townes, T. and Graff, J. M. (2009). Atf4 regulates obesity, glucose homeostasis, and energy expenditure. Diabetes 58: 2565-2573. PubMed ID: 19690063

  • Brenton Graveley UConn Health, Farmington, CT
    Brooks, A. N., Duff, M. O., May, G., Yang, L., Bolisetty, M., Landolin, J., Wan, K., Sandler, J., Celniker, S. E., Graveley, B. R. and Brenner, S. E. (2015). Regulation of alternative splicing in Drosophila by 56 RNA binding proteins. Genome Res [Epub ahead of print]. PubMed ID: 26294686

    Stoiber, M. H., Olson, S., May, G. E., Duff, M. O., Manent, J., Obar, R., Guruharsha, K., Artavanis-Tsakonas, S., Brown, J. B., Graveley, B. R. and Celniker, S. E. (2015). Extensive cross-regulation of post-transcriptional regulatory networks in Drosophila. Genome Res [Epub ahead of print]. PubMed ID: 26294687

    Coolon, J. D., Stevenson, K. R., McManus, C. J., Yang, B., Graveley, B. R. and Wittkopp, P. J. (2015). Molecular mechanisms and evolutionary processes contributing to accelerated divergence of gene expression on the Drosophila X chromosome. Mol Biol Evol. PubMed ID: 26041937

    Duff, M.O., Olson, S., Wei, X., Garrett, S.C., Osman, A., Bolisetty, M., Plocik, A., Celniker, S.E. and Graveley, B.R. (2015). Genome-wide identification of zero nucleotide recursive splicing in Drosophila. Nature [Epub ahead of print]. PubMed ID: 25970244

    Westholm, J. O., Miura, P., Olson, S., Shenker, S., Joseph, B., Sanfilippo, P., Celniker, S. E., Graveley, B. R. and Lai, E. C. (2014). Genome-wide analysis of drosophila circular RNAs reveals their structural and sequence properties and age-dependent neural accumulation. Cell Rep 9: 1966-1980. PubMed ID: 25544350

    Coolon, J. D., McManus, C. J., Stevenson, K. R., Graveley, B. R. and Wittkopp, P. J. (2014). Tempo and mode of regulatory evolution in Drosophila. Genome Res 24: 797-808. PubMed ID: 24567308

    McManus, C. J., Coolon, J. D., Eipper-Mains, J., Wittkopp, P. J. and Graveley, B. R. (2014). Evolution of splicing regulatory networks in Drosophila. Genome Res 24: 786-796. PubMed ID: 24515119

  • Rachel Green Molecular Biology and Genetics, Johns Hopkins
    Hur, J. K., Zinchenko, M. K., Djuranovic, S., Green, R. (2013) Regulation of Argonaute slicer activity by guide RNA 3' end interactions with the N-terminal lobe. J Biol Chem 288: 7829-7840. PubMed ID: 23329841

    Djuranovic, S., Nahvi, A. and Green, R. (2012). miRNA-mediated gene silencing by translational repression followed by mRNA deadenylation and decay. Science 336: 237-240. PubMed ID: 22499947

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

    Graveley, B. R., et al. (2011). The developmental transcriptome of Drosophila melanogaster. Nature 471: 473-479. PubMed ID: 21179090

  • Ralph Greenspan Kavli Institute for Brain and Mind, La Jolla, CA
    Wu, S. T., Chen, J. Y., Martin, V., Ng, R., Zhang, Y., Grover, D., Greenspan, R. J., Aljadeff, J. and Su, C. Y. (2022). Valence opponency in peripheral olfactory processing. Proc Natl Acad Sci U S A 119(5). PubMed ID: 35091473

    Grover, D., Chen, J. Y., Xie, J., Li, J., Changeux, J. P. and Greenspan, R. J. (2022). Differential mechanisms underlie trace and delay conditioning in Drosophila. Nature 603(7900): 302-308. PubMed ID: 35173333

    Sun, R., Delly, J., Sereno, E., Wong, S., Chen, X., Wang, Y., Huang, Y. and Greenspan, R. J. (2020). Anti-instinctive Learning Behavior Revealed by Locomotion-Triggered Mild Heat Stress in Drosophila. Front Behav Neurosci 14: 41. PubMed ID: 32372923

    Grover, D., Katsuki, T., Li, J., Dawkins, T. J. and Greenspan, R. J. (2020). Imaging brain activity during complex social behaviors in Drosophila with Flyception2. Nat Commun 11(1): 623. PubMed ID: 32001689

    Aimon, S., Katsuki, T., Jia, T., Grosenick, L., Broxton, M., Deisseroth, K., Sejnowski, T. J. and Greenspan, R. J. (2019). Fast near-whole-brain imaging in adult Drosophila during responses to stimuli and behavior. PLoS Biol 17(2): e2006732. PubMed ID: 30768592

    Shih, C.T., Sporns, O., Yuan, S.L., Su, T.S., Lin, Y.J., Chuang, C.C., Wang, T.Y., Lo, C.C., Greenspan, R.J. and Chiang, A.S. (2015). Connectomics-based analysis of information flow in the Drosophila brain. Curr Biol 25: 1249-1258. PubMed ID: 25866397

    Shih, C. T., Sporns, O., Yuan, S. L., Su, T. S., Lin, Y. J., Chuang, C. C., Wang, T. Y., Lo, C. C., Greenspan, R. J. and Chiang, A. S. (2015). Connectomics-based analysis of information flow in the Drosophila brain. Curr Biol. PubMed ID: 25866397

    Lin, C. Y., Chuang, C. C., Hua, T. E., Chen, C. C., Dickson, B. J., Greenspan, R. J. and Chiang, A. S. (2013). A comprehensive wiring diagram of the protocerebral bridge for visual information processing in the Drosophila brain. Cell Rep 3: 1739-1753. PubMed ID: 23707064

    Houot, B., Fraichard, S., Greenspan, R. J. and Ferveur, J. F. (2012). Genes involved in sex pheromone discrimination in Drosophila melanogaster and their background-dependent effect. PLoS One 7: e30799. PubMed ID: 22292044

    Kent, C. F., Daskalchuk, T., Cook, L., Sokolowski, M. B. and Greenspan, R. J. (2009). The Drosophila foraging gene mediates elasticity and gene-environment interactions in behaviour, metabolites, and gene expression in response to food deprivation. PLoS Genet 5: e1000609. PubMed ID: 19696884

  • Thomas Gregor Department of Physics, Lewis-Sigler Institute for Integrative Genomics, Princeton University
    Bruckner, D. B., Chen, H., Barinov, L., Zoller, B. and Gregor, T. (2023). Stochastic motion and transcriptional dynamics of pairs of distal DNA loci on a compacted chromosome. Science 380(6652): 1357-1362. PubMed ID: 37384691

    Voortman, L., Anderson, C., Urban, E., Yuan, L., Tran, S., Neuhaus-Follini, A., Derrick, J., Gregor, T. and Johnston, R. J., Jr. (2022). Temporally dynamic antagonism between transcription and chromatin compaction controls stochastic photoreceptor specification in flies. Dev Cell 57(15): 1817-1832.e1815. PubMed ID: 35835116

    Seyboldt, R., Lavoie, J., Henry, A., Vanaret, J., Petkova, M. D., Gregor, T. and Francois, P. (2022). Latent space of a small genetic network: Geometry of dynamics and information. Proc Natl Acad Sci U S A 119(26): e2113651119. PubMed ID: 35737842

    Singh, A. P., Wu, P., Ryabichko, S., Raimundo, J., Swan, M., Wieschaus, E., Gregor, T. and Toettcher, J. E. (2022). Optogenetic control of the Bicoid morphogen reveals fast and slow modes of gap gene regulation. Cell Rep 38(12): 110543. PubMed ID: 35320726

    Petkova, M. D., Tkacik, G., Bialek, W., Wieschaus, E. F. and Gregor, T. (2019). Optimal decoding of cellular identities in a genetic network. Cell 176(4): 844-855.e815. PubMed ID: 30712870

    Zoller, B., Little, S. C. and Gregor, T. (2018). Diverse spatial expression patterns emerge from unified kinetics of transcriptional bursting. Cell 175(3): 835-847.e825. PubMed ID: 30340044

    Chen, H., Levo, M., Barinov, L., Fujioka, M., Jaynes, J. B. and Gregor, T. (2018). Dynamic interplay between enhancer-promoter topology and gene activity. Nat Genet. PubMed ID: 30038397

    Abouchar, L., Petkova, M. D., Steinhardt, C. R. and Gregor, T. (2014). Fly wing vein patterns have spatial reproducibility of a single cell. J R Soc Interface 11. PubMed ID: 24942847

    Petkova, M. D., Little, S. C., Liu, F. and Gregor, T. (2014). Maternal Origins of Developmental Reproducibility. Curr Biol. PubMed ID: 24856210

    Garcia, H. G., Tikhonov, M., Lin, A. and Gregor, T. (2013). Quantitative Imaging of Transcription in Living Drosophila Embryos Links Polymerase Activity to Patterning. Curr Biol. PubMed ID: 24139738

    Little, S. C., Tikhonov, M. and Gregor, T. (2013). Precise developmental gene expression arises from globally stochastic transcriptional activity. Cell 154: 789-800. PubMed ID: 23953111

  • Savraj Grewal Department of Biochemistry & Molecular Biology, University of Calgary
    Ding, K., Barretto, E. C., Johnston, M., Lee, B., Gallo, M. and Grewal, S. S. (2022). Transcriptome analysis of FOXO-dependent hypoxia gene expression identifies Hipk as a regulator of low oxygen tolerance in Drosophila. G3 (Bethesda). PubMed ID: 36200850

    Deshpande, R., Lee, B. and Grewal, S. S. (2022). Enteric bacterial infection in Drosophila induces whole-body alterations in metabolic gene expression independently of the immune deficiency signaling pathway. G3 (Bethesda) 12(11). PubMed ID: 35781508

    Sriskanthadevan-Pirahas, S., Turingan, M. J., Chahal, J. S., Thorson, E., Khan, S., Tinwala, A. Q. and Grewal, S. S. (2022). Adipose mitochondrial metabolism controls body growth by modulating systemic cytokine and insulin signaling. Cell Rep 39(6): 110802. PubMed ID: 35545043

    Deliu, L. P., Turingan, M., Jadir, D., Lee, B., Ghosh, A. and Grewal, S. S. (2022). Serotonergic neuron ribosomal proteins regulate the neuroendocrine control of Drosophila development. PLoS Genet 18(9): e1010371. PubMed ID: 36048889

    Deshpande, R., Lee, B. and Grewal, S. S. (2022). Enteric bacterial infection in Drosophila induces whole-body alterations in metabolic gene expression independently of the Immune Deficiency (Imd) signalling pathway. G3 (Bethesda). PubMed ID: 35781508

    Deshpande, R., Lee, B., Qiao, Y. and Grewal, S. S. (2022). TOR signaling is required for host lipid metabolic remodelling and survival following enteric infection in Drosophila. Dis Model Mech. PubMed ID: 35363274

    Barretto, E. C., Polan, D. M., Beevor-Potts, A. N., Lee, B. and Grewal, S. S. (2020). Tolerance to Hypoxia Is Promoted by FOXO Regulation of the Innate Immunity Transcription Factor NF-κB/Relish in Drosophila. Genetics. PubMed ID: 32513813

    Lee, B., Barretto, E. C. and Grewal, S. S. (2019). TORC1 modulation in adipose tissue is required for organismal adaptation to hypoxia in Drosophila. Nat Commun 10(1): 1878. PubMed ID: 31015407

    Sriskanthadevan-Pirahas, S., Lee, J. and Grewal, S. S. (2018). The EGF/Ras pathway controls growth in Drosophila via ribosomal RNA synthesis. Dev Biol 439(1):19-29. PubMed ID: 29660312

    Spurrier, J., Shukla, A. K., McLinden, K., Johnson, K. and Giniger, E. (2018). Altered expression of the Cdk5 activator-like protein, Cdk5alpha, causes neurodegeneration in part by accelerating the rate of aging. Dis Model Mech. PubMed ID: 29469033

  • Leslie Griffith Brandeis University
    Chen, N., Zhang, Y., Rivera-Rodriguez, E. J., Yu, A. D., Hobin, M., Rosbash, M. and Griffith, L. C. (2023). Widespread posttranscriptional regulation of cotransmission. Sci Adv 9(22): eadg9836. PubMed ID: 37267358

    Yan, W., Lin, H., Yu, J., Wiggin, T. D., Wu, L., Meng, Z., Liu, C. and Griffith, L. C. (2023). Subtype-Specific Roles of Ellipsoid Body Ring Neurons in Sleep Regulation in Drosophila. J Neurosci 43(5): 764-786. PubMed ID: 36535771

    Chen, N., Zhang, Y., Rivera-Rodriguez, E. J., Yu, A. D., Hobin, M., Rosbash, M. and Griffith, L. C. (2023). Widespread post-transcriptional regulation of co-transmission. bioRxiv. PubMed ID: 36909471

    Adel, M., Chen, N., Zhang, Y., Reed, M. L., Quasney, C. and Griffith, L. C. (2022). Pairing-Dependent Plasticity in a Dissected Fly Brain Is Input-Specific and Requires Synaptic CaMKII Enrichment and Nighttime Sleep. J Neurosci 42(21): 4297-4310. PubMed ID: 35474278

    Hobin, M., Dorfman, K., Adel, M., Rivera-Rodriguez, E. J., Kuklin, E. A., Ma, D. and Griffith, L. C. (2022). The Drosophila microRNA bantam regulates excitability in adult mushroom body output neurons to promote early night sleep. iScience 25(9): 104874. PubMed ID: 36034229

    Chen, N., Zhang, Y., Adel, M., Kuklin, E. A., Reed, M. L., Mardovin, J. D., Bakthavachalu, B., VijayRaghavan, K., Ramaswami, M. and Griffith, L. C. (2022). Local translation provides the asymmetric distribution of CaMKII required for associative memory formation. Curr Biol 32(12): 2730-2738. PubMed ID: 35545085 Wiggin, T. D., Hsiao, Y., Liu, J. B., Huber, R. and Griffith, L. C. (2021). Rest Is Required to Learn an Appetitively-Reinforced Operant Task in Drosophila. Front Behav Neurosci 15: 681593. PubMed ID: 34220464

    Flyer-Adams, J. G., Rivera-Rodriguez, E. J., Yu, J., Mardovin, J. D., Reed, M. L. and Griffith, L. C. (2020). Regulation of Olfactory Associative Memory by the Circadian Clock Output Signal Pigment-Dispersing Factor (PDF). J Neurosci 40(47): 9066-9077. PubMed ID: 33106351

    Wiggin, T. D., Goodwin, P. R., Donelson, N. C., Liu, C., Trinh, K., Sanyal, S. and Griffith, L. C. (2020). Covert sleep-related biological processes are revealed by probabilistic analysis in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 32303656

    McNeill, E. M., Warinner, C., Alkins, S., Taylor, A., Heggeness, H., DeLuca, T. F., Fulga, T. A., Wall, D. P., Griffith, L. C. and Van Vactor, D. (2020). The conserved microRNA miR-34 regulates synaptogenesis via coordination of distinct mechanisms in presynaptic and postsynaptic cells. Nat Commun 11(1): 1092. PubMed ID: 32107390

    Liu, C., Meng, Z., Wiggin, T. D., Yu, J., Reed, M. L., Guo, F., Zhang, Y., Rosbash, M. and Griffith, L. C. (2019). A serotonin-modulated circuit controls sleep architecture to regulate cognitive function independent of total sleep in Drosophila. Curr Biol 29(21): 3635-3646.e3635. PubMed ID: 31668619

    Donelson, N. C., Dixit, R., Pichardo-Casas, I., Chiu, E. Y., Ohman, R. T., Slawson, J. B., Klein, M., Fulga, T. A., Van Vactor, D. and Griffith, L. C. (2019). MicroRNAs regulate multiple aspects of locomotor behavior in Drosophila. G3 (Bethesda). PubMed ID: 31694853

    Goodwin, P. R., Meng, A., Moore, J., Hobin, M., Fulga, T. A., Van Vactor, D. and Griffith, L. C. (2018). MicroRNAs regulate sleep and sleep homeostasis in Drosophila. Cell Rep 23(13): 3776-3786. PubMed ID: 29949763

    Bronk, P., Kuklin, E. A., Gorur-Shandilya, S., Liu, C., Wiggin, T. D., Reed, M. L., Marder, E. and Griffith, L. C. (2018). Regulation of Eag by calcium/calmodulin controls presynaptic excitability in Drosophila. J Neurophysiol. PubMed ID: 29364071

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

    Ninova, M., Ronshaugen, M. and Griffiths-Jones, S. (2014). Conserved temporal patterns of microRNA expression in Drosophila support a developmental hourglass model. Genome Biol Evol. PubMed ID: 25169982

    Ninova, M., Ronshaugen, M. and Griffiths-Jones, S. (2014). Fast-evolving microRNAs are highly expressed in the early embryo of Drosophila virilis. RNA 20: 360-372. PubMed ID: 24448446

    Rogers, H. H. and Griffiths-Jones, S. (2012). Mitochondrial pseudogenes in the nuclear genomes of Drosophila. PLoS One 7: e32593. PubMed ID: 22412894

  • Cornelis Grimmelikhuijzen Center for Functional and Comparative Insect Genomics, Department of Biology, University of Copenhagen
    Ren, G. R., Hauser, F., Rewitz, K. F., Kondo, S., Engelbrecht, A. F., Didriksen, A. K., Schjott, S. R., Sembach, F. E., Li, S., Sogaard, K. C., Sondergaard, L. and Grimmelikhuijzen, C. J. (2015). CCHamide-2 Is an orexigenic brain-gut peptide in Drosophila. PLoS One 10: e0133017. PubMed ID: 26168160

    Ren, G. R., Folke, J., Hauser, F., Li, S. and Grimmelikhuijzen, C. J. (2015). The A- and B-type muscarinic acetylcholine receptors from Drosophila melanogaster couple to different second messenger pathways. Biochem Biophys Res Commun. PubMed ID: 25964087

    Li, S., Torre-Muruzabal, T., Sogaard, K. C., Ren, G. R., Hauser, F., Engelsen, S. M., Podenphanth, M. D., Desjardins, A. and Grimmelikhuijzen, C. J. (2013). Expression patterns of the Drosophila neuropeptide CCHamide-2 and its receptor may suggest hormonal signaling from the gut to the brain. PLoS One 8: e76131. PubMed ID: 24098432

  • Yaël Grosjean Centre des Sciences du Gout at de l'Alimentation, Dijon
    Ziegler, A. B., Maniere, G. and Grosjean, Y. (2018). JhI-21 plays a role in Drosophila insulin-like peptide release from larval IPCs via leucine transport. Sci Rep 8(1): 1908. PubMed ID: 29382949

    Depetris-Chauvin, A., Galagovsky, D., Chevalier, C., Maniere, G. and Grosjean, Y. (2017). Olfactory detection of a bacterial short-chain fatty acid acts as an orexigenic signal in Drosophila melanogaster larvae. Sci Rep 7(1): 14230. PubMed ID: 29079812

    Maniere, G., Ziegler, A. B., Geillon, F., Featherstone, D. E. and Grosjean, Y. (2016). Direct Sensing of Nutrients via a LAT1-like Transporter in Drosophila Insulin-Producing Cells. Cell Rep 17: 137-148. PubMed ID: 27681427

    Ziegler, A. B., Augustin, H., Clark, N. L., Berthelot-Grosjean, M., Simonnet, M. M., Steinert, J. R., Geillon, F., Maniere, G., Featherstone, D. E. and Grosjean, Y. (2016). The amino acid transporter JhI-21 coevolves with glutamate receptors, impacts NMJ physiology, and influences locomotor activity in Drosophila larvae. Sci Rep 6: 19692. PubMed ID: 26805723

    Ziegler, A. B., Menage, C., Gregoire, S., Garcia, T., Ferveur, J. F., Bretillon, L. and Grosjean, Y. (2015). Lack of dietary polyunsaturated fatty acids causes synapse dysfunction in the Drosophila visual system. PLoS One 10: e0135353. PubMed ID: 26308084

    Simonnet, M. M., Berthelot-Grosjean, M. and Grosjean, Y. (2014). Testing Drosophila Olfaction with a Y-maze Assay. J Vis Exp. PubMed ID: 24961243

    Ziegler, A. B., Berthelot-Grosjean, M. and Grosjean, Y. (2013). The smell of love in Drosophila. Front Physiol 4: 72. PubMed ID: 23576993

    Grosjean, Y., Rytz, R., Farine, J. P., Abuin, L., Cortot, J., Jefferis, G. S. and Benton, R. (2011). An olfactory receptor for food-derived odours promotes male courtship in Drosophila. Nature 478: 236-240. PubMed ID: 21964331

  • Jörg Großhans Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences
    Lv, Z., Zhang, N., Zhang, X., Grosshans, J. and Kong, D. (2022). The Lateral Epidermis Actively Counteracts Pulling by the Amnioserosa During Dorsal Closure. Front Cell Dev Biol 10: 865397. PubMed ID: 35652100

    Schmidt, A., Li, L., Yan, S. and Grosshans, J. (2022). Restriction of subapical proteins during cellularization depends on the onset of zygotic transcription and the formin Dia. Dev Biol 487: 110-121. PubMed ID: 35525304

    Lv, Z., de-Carvalho, J., Telley, I. A. and Grosshans, J. (2021). Cytoskeletal mechanics and dynamics in the Drosophila syncytial embryo. J Cell Sci 134(4). PubMed ID: 33597155

    Lv, Z., Rosenbaum, J., Mohr, S., Zhang, X., Kong, D., Preiß, H., Kruss, S., Alim, K., Aspelmeier, T. and Großhans, J. (2020). The Emergent Yo-yo Movement of Nuclei Driven by Cytoskeletal Remodeling in Pseudo-synchronous Mitotic Cycles. Curr Biol. PubMed ID: 32470369

    Liu, B., Gregor, I., Muller, H. A. and Grosshans, J. (2020). Fluorescence fluctuation analysis reveals PpV dependent Cdc25 protein dynamics in living embryos. PLoS Genet 16(4): e1008735. PubMed ID: 32251417

    Liu, B., Sung, H. W. and Grosshans, J. (2019). Multiple functions of the essential gene PpV in Drosophila early development. G3 (Bethesda). PubMed ID: 31484673

    Liu, B., Winkler, F., Herde, M., Witte, C. P. and Grosshans, J. (2019). A link between deoxyribonucleotide metabolites and embryonic cell-cycle control. Curr Biol. PubMed ID: 30880011

    Yan, S. and Grosshans, J. (2018). Localization and translation control of slam in Drosophila cellularization. Fly (Austin): 1-8. PubMed ID: 30211628

    Petrovsky, R., Krohne, G. and Grosshans, J. (2018). Overexpression of the lamina proteins Lamin and Kugelkern induces specific ultrastructural alterations in the morphology of the nuclear envelope of intestinal stem cells and enterocytes. Eur J Cell Biol 97(2): 102-113. PubMed ID: 29395481

    Schmidt, A., Lv, Z. and Grosshans, J. (2018). ELMO and Sponge specify subapical restriction of Canoe and formation of the subapical domain in early Drosophila embryos. Development 145(2). PubMed ID: 29361564

    Petrovsky, R. and Grosshans, J. (2018). Expression of lamina proteins Lamin and Kugelkern suppresses stem cell proliferation. Nucleus 9(1): 104-118. PubMed ID: 29210315

    Lv, Z., Rosenbaum, J., Aspelmeier, T. and Grosshans, J. (2018). A "molecular guillotine" reveals an interphase function of Kinesin-5. J Cell Sci. PubMed ID: 29361546

  • Michael Grotewiel Human and Molecular Genetics, Virginia Commonwealth University, Richmond
    Shell, B. C. and Grotewiel, M. (2022). Identification of additional dye tracers for measuring solid food intake and food preference via consumption-excretion in Drosophila. Sci Rep 12(1): 6201. PubMed ID: 35418664

    Shell, B. C., Luo, Y., Pletcher, S. and Grotewiel, M. (2021). Expansion and application of dye tracers for measuring solid food intake and food preference in Drosophila. Sci Rep 11(1): 20044. PubMed ID: 34625601

    Lee, K. M., Talikoti, A., Shelton, K. and Grotewiel, M. (2021). Tyramine synthesis, vesicular packaging, and the SNARE complex function coordinately in astrocytes to regulate Drosophila alcohol sedation. Addict Biol: e13019. PubMed ID: 33538092

    Lee, K. M., Mathies, L. D. and Grotewiel, M. (2019). Alcohol sedation in adult Drosophila is regulated by Cysteine proteinase-1 in cortex glia. Commun Biol 2: 252. PubMed ID: 31286069

    Schmitt, R. E., Shell, B. C., Lee, K. M., Shelton, K. L., Mathies, L. D., Edwards, A. C. and Grotewiel, M. (2019). Convergent evidence from humans and Drosophila melanogaster implicates the transcription factor MEF2B/Mef2 in alcohol sensitivity. Alcohol Clin Exp Res. PubMed ID: 31241765

    Schmitt, R. E., Messick, M. R., Shell, B. C., Dunbar, E. K., Fang, H. F., Shelton, K. L., Venton, B. J., Pletcher, S. D. and Grotewiel, M. (2019). Dietary yeast influences ethanol sedation in Drosophila via serotonergic neuron function. Addict Biol: e12779. PubMed ID: 31169340

    Shell, B. C., Schmitt, R. E., Lee, K. M., Johnson, J. C., Chung, B. Y., Pletcher, S. D. and Grotewiel, M. (2018). Measurement of solid food intake in Drosophila via consumption-excretion of a dye tracer. Sci Rep 8(1): 11536. PubMed ID: 30068981

    Sandhu, S., Kollah, A. P., Lewellyn, L., Chan, R. F. and Grotewiel, M. (2015). An inexpensive, scalable behavioral assay for measuring ethanol sedation sensitivity and rapid tolerance in Drosophila. J Vis Exp. PubMed ID: 25939022

    Chan, R. F., Lewellyn, L., DeLoyht, J. M., Sennett, K., Coffman, S., Hewitt, M., Bettinger, J. C., Warrick, J. M. and Grotewiel, M. (2014). Contrasting Influences of Drosophila white/mini-white on Ethanol Sensitivity in Two Different Behavioral Assays. Alcohol Clin Exp Res 38: 1582-1593. PubMed ID: 24890118

    Jones, M. A., Amr, S., Ferebee, A., Huynh, P., Rosenfeld, J. A., Miles, M. F., Davies, A. G., Korey, C. A., Warrick, J. M., Shiang, R., Elsea, S. H., Girirajan, S. and Grotewiel, M. (2014). Genetic studies in Drosophila and humans support a model for the concerted function of CISD2, PPT1 and CLN3 in disease. Biol Open 3: 342-352. PubMed ID: 24705017

  • Wes Grueber Department of Neuroscience, Columbia University
    Cooney, P. C., Li, W., Huang, Y., Hormigo, R., Tabachnik, T., Hillman, E. M. C., Grueber, W. B. and Zarin, A. A. (2023). Neuromuscular Basis of Drosophila Larval Escape Behavior. bioRxiv. PubMed ID: 36778508

    Galindo, S. E., Shin, G. J., Millard, S. S. and Grueber, W. B. (2023). Regulated alternative splicing of Dscam2 is required for somatosensory circuit wiring. bioRxiv. PubMed ID: 36909552

    Shrestha, B. R., Burgos, A. and Grueber, W. B. (2021). The Immunoglobulin Superfamily Member Basigin Is Required for Complex Dendrite Formation in Drosophila. Front Cell Neurosci 15: 739741. PubMed ID: 34803611

    Shin, G. J., Pero, M. E., Hammond, L. A., Burgos, A., Kumar, A., Galindo, S. E., Lucas, T., Bartolini, F. and Grueber, W. B. (2021). Integrins protect sensory neurons in models of paclitaxel-induced peripheral sensory neuropathy. Proc Natl Acad Sci U S A 118(15). PubMed ID: 33876743

    Burgos, A., Honjo, K., Ohyama, T., Qian, C. S., Shin, G. J., Gohl, D. M., Silies, M., Tracey, W. D., Zlatic, M., Cardona, A. and Grueber, W. B. (2018). Nociceptive interneurons control modular motor pathways to promote escape behavior in Drosophila. Elife 7. PubMed ID: 29528286

    O'Connor, R. M., Stone, E. F., Wayne, C. R., Marcinkevicius, E. V., Ulgherait, M., Delventhal, R., Pantalia, M. M., Hill, V. M., Zhou, C. G., McAllister, S., Chen, A., Ziegenfuss, J. S., Grueber, W. B., Canman, J. C. and Shirasu-Hiza, M. M. (2017). A Drosophila model of Fragile X syndrome exhibits defects in phagocytosis by innate immune cells. J Cell Biol 216(3): 595-605. PubMed ID: 28223318

    Corty, M.M., Tam, J. and Grueber, W.B. (2016). Dendritic diversification through transcription factor-mediated suppression of alternative morphologies. Development 143: 1351-1362. PubMed ID: 27095495

    Singhania, A. and Grueber, W. B. (2014). Development of the embryonic and larval peripheral nervous system of Drosophila. Wiley Interdiscip Rev Dev Biol 3: 193-210. PubMed ID: 24896657

    Kim, M. E., Shrestha, B. R., Blazeski, R., Mason, C. A. and Grueber, W. B. (2012). Integrins establish dendrite-substrate relationships that promote dendritic self-avoidance and patterning in drosophila sensory neurons. Neuron 73: 79-91. PubMed ID: 22243748

  • Yosef Gruenbaum Dept. of Genetics, The Hebrew University of Jerusalem,
    Lyakhovetsky, R. and Gruenbaum, Y. (2014). Studying lamins in invertebrate models. Adv Exp Med Biol 773: 245-262. PubMed ID: 24563351

    Vadasz, I., Dada, L. A., Briva, A., Helenius, I. T., Sharabi, K., Welch, L. C., Kelly, A. M., Grzesik, B. A., Budinger, G. R., Liu, J., Seeger, W., Beitel, G. J., Gruenbaum, Y. and Sznajder, J. I. (2012). Evolutionary conserved role of c-Jun-N-terminal kinase in CO2-induced epithelial dysfunction. PLoS One 7: e46696. PubMed ID: 23056407

    Azzam, Z. S., Sharabi, K., Guetta, J., Bank, E. M. and Gruenbaum, Y. (2010). The physiological and molecular effects of elevated CO2 levels. Cell Cycle 9: 1528-1532. PubMed ID: 20372066

  • Nataly Gruntenko Institute of Cytology and Genetics, Novosibirsk
    Bobrovskikh, M. A. and Gruntenko, N. E. (2023). Mechanisms of Neuroendocrine Stress Response in Drosophila and Its Effect on Carbohydrate and Lipid Metabolism. Insects 14(5). PubMed ID: 37233102

    Eremina, M. A., Menshanov, P. N., Shishkina, O. D. and Gruntenko, N. E. (2021). The transcription factor dfoxo controls the expression of insulin pathway genes and lipids content under heat stress in Drosophila melanogaster. Vavilovskii Zhurnal Genet Selektsii 25(5): 465-471. PubMed ID: 34595369

    Rauschenbach, I. Y., Karpova, E. K., Burdina, E. V., Adonyeva, N. V., Bykov, R. A., Ilinsky, Y. Y., Menshanov, P. N. and Gruntenko, N. E. (2016). Insulin-like peptide DILP6 regulates juvenile hormone and dopamine metabolism in Drosophila females. Gen Comp Endocrinol 243: 1-9. PubMed ID: 27823956

    Rauschenbach, I. Y., Karpova, E. K., Adonyeva, N. V., Andreenkova, O. V., Faddeeva, N. V., Burdina, E. K., Alekseev, A. A., Menshanov, P. N. and Gruntenko, N. E. (2014). Disruption of insulin signalling affects the neuroendocrine stress reaction in Drosophila females. J Exp Biol. PubMed ID: 25214494

    Rauschenbach, I. Y., Adonyeva, N. V., Faddeeva, N. V., Shumnaya, L. V. and Gruntenko, N. E. (2013). The role of the insulin signaling pathway in the control of the Drosophila sexual dimorphism with respect to thermal stress resistance. Dokl Biol Sci 452: 284-286. PubMed ID: 24150648

    Karpova, E. K., Adonyeva, N. V., Faddeeva, N. V., Romanova, I. V., Gruntenko, N. E. and Rauschenbach, I. Y. (2013). Insulin affects reproduction and juvenile hormone metabolism under normal and stressful conditions in Drosophila females. Dokl Biochem Biophys 452: 264-266. PubMed ID: 24150588

    Adonyeva, N. V., Bogomolova, E. V., Rauschenbach, I. Y. and Gruntenko, N. E. (2013). Insulin affects dopamine metabolism in Drosophila females under normal and stress conditions. Dokl Biochem Biophys 448: 40-42. PubMed ID: 23478985

    Rauschenbach, I. Y., Bogomolova, E. V., Karpova, E. K., Adonyeva, N. V., Faddeeva, N. V., Menshanov, P. N. and Gruntenko, N. E. (2011). Mechanisms of age-specific regulation of dopamine metabolism by juvenile hormone and 20-hydroxyecdysone in Drosophila females. J Comp Physiol B 181: 19-26. PubMed ID: 20878329

  • Ilona Grunwald Kadow Munich Center for Neurosciences
    Aimon, S., Cheng, K. Y., Gjorgjieva, J. and Grunwald Kadow, I. C. (2023). Global change in brain state during spontaneous and forced walk in Drosophila is composed of combined activity patterns of different neuron classes. Elife 12. PubMed ID: 37067152

    Ucpunar, H. K. and Grunwald Kadow, I. C. (2021). Flies Avoid Current Atmospheric CO(2) Concentrations. Front Physiol 12: 646401. PubMed ID: 33927640

    Kobler, J. M., Rodriguez Jimenez, F. J., Petcu, I. and Grunwald Kadow, I. C. (2020). Immune Receptor Signaling and the Mushroom Body Mediate Post-ingestion Pathogen Avoidance. Curr Biol. PubMed ID: 33007248

    Sayin, S., De Backer, J. F., Siju, K. P., Wosniack, M. E., Lewis, L. P., Frisch, L. M., Gansen, B., Schlegel, P., Edmondson-Stait, A., Sharifi, N., Fisher, C. B., Calle-Schuler, S. A., Lauritzen, J. S., Bock, D. D., Costa, M., Jefferis, G., Gjorgjieva, J. and Grunwald Kadow, I. C. (2019). A Neural Circuit Arbitrates between Persistence and Withdrawal in Hungry Drosophila. Neuron. PubMed ID: 31471123

    Sardana, J., Organisti, C. and Grunwald Kadow, I. C. (2018). Eph Receptor Effector Ephexin Mediates Olfactory Dendrite Targeting in Drosophila. Dev Neurobiol 78(9): 873-888. PubMed ID: 30019861

    Hinaux, H., Bachem, K., Battistara, M., Rossi, M., Xin, Y., Jaenichen, R., Le Poul, Y., Arnoult, L., Kobler, J. M., Grunwald Kadow, I. C., Rodermund, L., Prud'homme, B. and Gompel, N. (2018). Revisiting the developmental and cellular role of the pigmentation gene yellow in Drosophila using a tagged allele. Dev Biol 438(2): 111-123. PubMed ID: 29634916

  • Huaiyu Gu Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat Sen University, Guangzhou, China
    Chen, D., Zhu, X., Zhou, L., Wang, J., Tao, X., Wang, S., Sun, F., Kan, X., Han, Z. and Gu, Y. (2017). Gilgamesh is required for the maintenance of germline stem cells in Drosophila testis. Sci Rep 7(1): 5737. PubMed ID: 28720768

    Yan, Y., Xu, Y., Deng, S., Huang, N., Yang, Y., Qiu, J., Liu, J., Wang, X., Yang, G. and Gu, H. (2013). A pair of identified giant visual projection neurons demonstrates rhythmic activities before eclosion. Neurosci Lett. PubMed ID: 23827229

    Fang, L., Duan, J., Ran, D., Fan, Z., Yan, Y., Huang, N., Gu, H. and Zhu, Y. (2012). Amyloid-beta depresses excitatory cholinergic synaptic transmission in Drosophila. Neurosci Bull 28: 585-594. PubMed ID: 23054636

    Duan, J., Li, W., Yuan, D., Sah, B., Yan, Y. and Gu, H. (2012). Nitric oxide signaling modulates cholinergic synaptic input to projection neurons in Drosophila antennal lobes. Neuroscience 219: 1-9. PubMed ID: 22683719

    Huang, N., Yan, Y., Xu, Y., Jin, Y., Lei, J., Zou, X., Ran, D., Zhang, H., Luan, S. and Gu, H. (2013). Alumina nanoparticles alter rhythmic activities of local interneurons in the antennal lobe of Drosophila. Nanotoxicology 7: 212-220. PubMed ID: 22264077

  • Kun-Liang Guan Department of Pharmacology, University of California San Diego
    u, F. X., Zhang, Y., Park, H. W., Jewell, J. L., Chen, Q., Deng, Y., Pan, D., Taylor, S. S., Lai, Z. C. and Guan, K. L. (2013). Protein kinase A activates the Hippo pathway to modulate cell proliferation and differentiation. Genes Dev 27: 1223-1232. PubMed ID: 23752589

    Yu, F. X. and Guan, K. L. (2013). The Hippo pathway: regulators and regulations. Genes Dev 27: 355-371. PubMed ID: 23431053

    Li, L. and Guan, K. L. (2013). Microtubule-associated protein/microtubule affinity-regulating kinase 4 (MARK4) is a negative regulator of the mammalian target of rapamycin complex 1 (mTORC1). J Biol Chem 288: 703-708. PubMed ID: 23184942

    Hong, W. and Guan, K. L. (2012). The YAP and TAZ transcription co-activators: key downstream effectors of the mammalian Hippo pathway. Semin Cell Dev Biol 23: 785-793. PubMed ID: 22659496

    Valbuena, N., Guan, K. L. and Moreno, S. (2012). The Vam6 and Gtr1-Gtr2 pathway activates TORC1 in response to amino acids in fission yeast. J Cell Sci 125: 1920-1928. PubMed ID: 22344254

  • Isabelle Guénal Université de Versailles Saint-Quentin, Versailles, France
    Clavier, A., Rincheval-Arnold, A., Baillet, A., Mignotte, B. and Guenal, I. (2016). Two different specific JNK activators are required to trigger apoptosis or compensatory proliferation in response to Rbf1 in Drosophila. Cell Cycle 15: 283-294. PubMed ID: 26825229

    Clavier, A., Ruby, V., Rincheval-Arnold, A., Mignotte, B and Guénal, I. (2015). The Drosophila retinoblastoma protein, Rbf1, induces a debcl and drp1-dependent mitochondrial apoptosis. J Cell Sci [Epub ahead of print]. PubMed ID: 26208635

    Colin, J., Garibal, J., Clavier, A., Rincheval-Arnold, A., Gaumer, S., Mignotte, B. and Guenal, I. (2014). The drosophila Bcl-2 family protein Debcl is targeted to the proteasome by the beta-TrCP homologue slimb. Apoptosis 19: 1444-1456. PubMed ID: 25208640

    Colin, J., Garibal, J., Clavier, A., Rincheval-Arnold, A., Gaumer, S., Mignotte, B. and Guenal, I. (2014). The drosophila Bcl-2 family protein Debcl is targeted to the proteasome by the beta-TrCP homologue slimb. Apoptosis [Epub ahead of print]. PubMed ID: 25208640

    Clavier, A., Baillet, A., Rincheval-Arnold, A., Coleno-Costes, A., Lasbleiz, C., Mignotte, B. and Guenal, I. (2014). The pro-apoptotic activity of Drosophila Rbf1 involves dE2F2-dependent downregulation of diap1 and buffy mRNA. Cell Death Dis 5: e1405. PubMed ID: 25188515

    Milet, C., Rincheval-Arnold, A., Mignotte, B. and Guenal, I. (2010). The Drosophila retinoblastoma protein induces apoptosis in proliferating but not in post-mitotic cells. Cell Cycle 9: 97-103. PubMed ID: 20016284

  • Pilar Garcia Guerreiro Grup de Biología Evolutiva, Autonomous University of Barcelona
    Gamez-Visairas, V., Romero-Soriano, V., Marti-Carreras, J., Segarra-Carrillo, E. and Garcia Guerreiro, M. P. (2020). Drosophila Interspecific Hybridization Causes A Deregulation of the piRNA Pathway Genes. Genes (Basel) 11(2). PubMed ID: 32092860

    Romero-Soriano, V., Burlet, N., Vela, D., Fontdevila, A., Vieira, C. and Garcia Guerreiro, M.P. (2016). Drosophila females undergo genome expansion after interspecific hybridization. Genome Biol Evol [Epub ahead of print]. PubMed ID: 26872773

    Romero-Soriano, V. and Garcia Guerreiro, M. P. (2016). Expression of the retrotransposon Helena reveals a complex pattern of TE deregulation in Drosophila Hybrids. PLoS One 11: e0147903. PubMed ID: 26812285

    Vela, D., Fontdevila, A., Vieira, C. and Garcia Guerreiro, M. P. (2014). A genome-wide survey of genetic instability by transposition in Drosophila hybrids. PLoS One 9: e88992. PubMed ID: 24586475

    Garcia Guerreiro, M. P. (2012). What makes transposable elements move in the Drosophila genome? Heredity (Edinb) 108: 461-468. PubMed ID: 21971178

  • Isabel Guerrero Centro de Biología Molecular Severo Ochoa, Madrid
    Simon, E., Jimenez-Jimenez, C., Seijo-Barandiaran, I., Aguilar, G., Sanchez-Hernandez, D., Aguirre-Tamaral, A., Gonzalez-Mendez, L., Ripoll, P. and Guerrero, I. (2021). Glypicans define unique roles for the Hedgehog co-receptors boi and ihog in cytoneme-mediated gradient formation. Elife 10. PubMed ID: 34355694

    Aguirre-Tamaral, A. and Guerrero, I. (2021). Improving the understanding of cytoneme-mediated morphogen gradients by in silico modeling. PLoS Comput Biol 17(8): e1009245. PubMed ID: 34343167

    Gonzalez-Mendez, L., Gradilla, A. C., Sanchez-Hernandez, D., Gonzalez, E., Aguirre-Tamaral, A., Jimenez-Jimenez, C., Guerra, M., Aguilar, G., Andres, G., Falcon-Perez, J. M. and Guerrero, I. (2020). Polarized sorting of Patched enables cytoneme-mediated Hedgehog reception in the Drosophila wing disc. Embo j: e103629. PubMed ID: 32311148

    Simon, E., de la Puebla, S. F. and Guerrero, I. (2019). Drosophila Zic family member odd-paired is needed for adult post-ecdysis maturation. Open Biol 9(12): 190245. PubMed ID: 31847787

    Seijo-Barandiaran, I., Guerrero, I. and Bischoff, M. (2015). In Vivo Imaging of Hedgehog Transport in Drosophila Epithelia. Methods Mol Biol 1322: 9-18. PubMed ID: 26179035

    Simon, E. and Guerrero, I. (2015). The transcription factor optomotor-blind antagonizes Drosophila haltere growth by repressing decapentaplegic and hedgehog targets. PLoS One 10: e0121239. PubMed ID: 25793870

    Sanchez-Hernandez, D., Sierra, J., Ortigao-Farias, J. R. and Guerrero, I. (2013). The WIF domain of the human and Drosophila Wif-1 secreted factors confers specificity for Wnt or Hedgehog. Development 140: 4645. PubMed ID: 24194473

    Bischoff, M., Gradilla, A. C., Seijo, I., Andres, G., Rodriguez-Navas, C., Gonzalez-Mendez, L. and Guerrero, I. (2013). Cytonemes are required for the establishment of a normal Hedgehog morphogen gradient in Drosophila epithelia. Nat Cell Biol. PubMed ID: 24121526

    Gradilla, A. C. and Guerrero, I. (2013). Hedgehog on the move: a precise spatial control of Hedgehog dispersion shapes the gradient. Curr Opin Genet Dev. PubMed ID: 23747033

    Callejo, A., et al. (2011) Dispatched mediates Hedgehog basolateral release to form the long-range morphogenetic gradient in the Drosophila wing disk epithelium. Proc. Natl. Acad. Sci. 108: 12591-12598. PubMed ID: 21690386

  • Antoine Guichet Institute Jacque Monod
    Loh, M., Bernard, F. and Guichet, A. (2023). Kinesin-1 promotes centrosome clustering and nuclear migration in the Drosophila oocyte. Development 150(13). PubMed ID: 37334771

    Tissot, N., Lepesant, J. A., Bernard, F., Legent, K., Bosveld, F., Martin, C., Faklaris, O., Bellaiche, Y., Coppey, M. and Guichet, A. (2017). Distinct molecular cues ensure a robust microtubule-dependent nuclear positioning in the Drosophila oocyte. Nat Commun 8: 15168. PubMed ID: 28447612

    Le Droguen, P.M., Claret, S., Guichet, A. and Brodu, V. (2015). Microtubule-dependent apical restriction of recycling endosomes sustains adherens junctions during morphogenesis of the Drosophila tracheal system. Development 142: 363-374. PubMed ID: 25564624

    Claret, S., Jouette, J., Benoit, B., Legent, K. and Guichet, A. (2014). PI(4,5)P2 Produced by the PI4P5K SKTL Controls Apical Size by Tethering PAR-3 in Drosophila Epithelial Cells. Curr Biol. PubMed ID: 24768049

    Lecland, N., Debec, A., Delmas, A., Moutinho-Pereira, S., Malmanche, N., Bouissou, A., Dupre, C., Jourdan, A., Raynaud-Messina, B., Maiato, H., Guichet, A. (2013) Establishment and mitotic characterization of new Drosophila acentriolar cell lines from DSas-4 mutant. Biol Open 2: 314-323. PubMed ID: 23519377

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

    Parrott, B. B., Chiang, Y., Hudson, A., Sarkar, A., Guichet, A. and Schulz, C. (2011). Nucleoporin98-96 function is required for transit amplification divisions in the germ line of Drosophila melanogaster. PLoS One 6: e25087. PubMed ID: 21949861

    Brodu, V., Baffet, A. D., Le Droguen, P. M., Casanova, J. and Guichet, A. (2010). A developmentally regulated two-step process generates a noncentrosomal microtubule network in Drosophila tracheal cells. Dev Cell 18: 790-801. PubMed ID: 20493812

  • Shermali Gunawardena Laboratory of Axonal Transport and Neurodegenerative Disease, SUNY, Buffalo
    Krzystek, T. J., White, J. A., Rathnayake, R., Thurston, L., Hoffmar-Glennon, H., Li, Y. and Gunawardena, S. (2022). HTT (huntingtin) and RAB7 co-migrate retrogradely on a signaling LAMP1-containing late endosome during axonal injury. Autophagy: 1-22. PubMed ID: 36048753

    Banerjee, R., Chakraborty, P., Yu, M. C. and Gunawardena, S. (2021). A stop or go switch: glycogen synthase kinase 3β phosphorylation of the kinesin 1 motor domain at Ser314 halts motility without detaching from microtubules. Development 148(24). PubMed ID: 34940839

    White, J. A., 2nd, Krzystek, T. J., Hoffmar-Glennon, H., Thant, C., Zimmerman, K., Iacobucci, G., Vail, J., Thurston, L., Rahman, S. and Gunawardena, S. (2020). Excess Rab4 rescues synaptic and behavioral dysfunction caused by defective HTT-Rab4 axonal transport in Huntington's disease. Acta Neuropathol Commun 8(1): 97. PubMed ID: 32611447

    Iacobucci, G. J. and Gunawardena, S. (2017). Ethanol stimulates the in vivo axonal movement of neuropeptide dense core vesicles in Drosophila motor neurons. J Neurochem [Epub ahead of print]. PubMed ID: 28960313

    White, J. A., Anderson, E., Zimmerman, K., Zheng, K. H., Rouhani, R. and Gunawardena, S. (2015). Huntingtin differentially regulates the axonal transport of a sub-set of Rab-containing vesicles in vivo. Hum Mol Genet. PubMed ID: 26450517

    Kang, M. J., Hansen, T. J., Mickiewicz, M., Kaczynski, T. J., Fye, S. and Gunawardena, S. (2014). Disruption of Axonal Transport Perturbs Bone Morphogenetic Protein (BMP) - Signaling and Contributes to Synaptic Abnormalities in Two Neurodegenerative Diseases. PLoS One 9: e104617. PubMed ID: 25127478

    Iacobucci, G. J., Rahman, N. A., Valtuena, A. A., Nayak, T. K. and Gunawardena, S. (2014). Spatial and temporal characteristics of normal and perturbed vesicle transport. PLoS One 9: e97237. PubMed ID: 24878565

    Gunawardena, S., Yang, G. and Goldstein, L. S. (2013). Presenilin controls kinesin-1 and dynein function during APP-vesicle transport in vivo. Hum Mol Genet 22: 3828-3843. PubMed ID: 23710041

    Power, D., Srinivasan, S. and Gunawardena, S. (2012). In-vivo evidence for the disruption of Rab11 vesicle transport by loss of huntingtin. Neuroreport 23: 970-977. PubMed ID: 23032403

    Barandeh, F., Nguyen, P. L., Kumar, R., Iacobucci, G. J., Kuznicki, M. L., Kosterman, A., Bergey, E. J., Prasad, P. N. and Gunawardena, S. (2012). Organically modified silica nanoparticles are biocompatible and can be targeted to neurons in vivo. PLoS One 7: e29424. PubMed ID: 22238611

  • Ai-Ke Guo Institute of Neuroscience, Chinese Academy of Sciences, Shanghai
    Liu, T., Wang, Y., Tian, Y., Zhang, J., Zhao, J. and Guo, A. (2018). The receptor channel formed by ppk25, ppk29 and ppk23 can sense the Drosophila female pheromone 7,11-HD. Genes Brain Behav: e12529. PubMed ID: 30345606

    Liu, H., Zhou, B., Yan, W., Lei, Z., Zhao, X., Zhang, K. and Guo, A. (2014). Astrocyte-like glial cells physiologically regulate olfactory processing through the modification of ORN-PN synaptic strength in Drosophila. Eur J Neurosci. PubMed ID: 24964821

    Yi, W., Zhang, Y., Tian, Y., Guo, J., Li, Y. and Guo, A. (2013). A subset of cholinergic mushroom body neurons requires go signaling to regulate sleep in Drosophila. Sleep 36: 1809-1821. PubMed ID: 24293755

    Li, H., Li, Y., Lei, Z., Wang, K. and Guo, A. (2013). Transformation of odor selectivity from projection neurons to single mushroom body neurons mapped with dual-color calcium imaging. Proc Natl Acad Sci U S A. PubMed ID: 23818618

    Yang, X., Guo, A. (2013) Distinct acute zones for visual stimuli in different visual tasks in Drosophila. PLoS One 8: e61313. PubMed ID: 23585891

    Zhang, X., Ren, Q., Guo, A. (2013) Parallel pathways for cross-modal memory retrieval in Drosophila. J Neurosci 33: 8784-8793. PubMed ID: 23678121

    Zhang, X., Liu, H., Lei, Z., Wu, Z. and Guo, A. (2012). Lobula-specific visual projection neurons are involved in perception of motion-defined second-order motion in Drosophila. J Exp Biol. PubMed ID: 23077158

    Wu, Y., Ren, Q., Li, H. and Guo, A. (2012). The GABAergic anterior paired lateral neurons facilitate olfactory reversal learning in Drosophila. Learn Mem. 19(10): 478-86. PubMed Citation: 22988290

    Ren, Q., Li, H., Wu, Y., Ren, J. and Guo, A. (2012). A GABAergic inhibitory neural circuit regulates visual reversal learning in Drosophila. J Neurosci 32: 11524-11538. PubMed ID: 22915099

  • Ming Guo Molecular, Cellular and Integrative Physiology, UCLA
    Ma, P., Yun, J., Deng, H. and Guo, M. (2018). Atg1 mediated autophagy suppresses tissue degeneration in pink1/parkin mutants by promoting mitochondrial fission in Drosophila. Mol Biol Cell: mbcE18040243. PubMed ID: 30354903

    Zhang, T., Mishra, P., Hay, B. A., Chan, D. and Guo, M. (2017). Valosin-containing protein (VCP/p97) inhibitors relieve Mitofusin-dependent mitochondrial defects due to VCP disease mutants. Elife 6. PubMed ID: 28322724

    Dodson, M. W., Leung, L. K., Lone, M., Lizzio, M. A. and Guo, M. (2014). Novel alleles of the Drosophila LRRK2 homolog reveal a crucial role in endolysosomal functions and autophagy in vivo. Dis Model Mech. PubMed ID: 25288684

    Gross, G. G., Lone, G. M., Leung, L. K., Hartenstein, V. and Guo, M. (2013). X11/Mint Genes Control Polarized Localization of Axonal Membrane Proteins in Vivo. J Neurosci 33: 8575-8586. PubMed ID: 23658195

    Guo, M. (2012). Drosophila as a model to study mitochondrial dysfunction in Parkinson's disease. Cold Spring Harb Perspect Med 2. PubMed ID: 23024178

    Dodson, M. W., Zhang, T., Jiang, C., Chen, S. and Guo, M. (2012). Roles of the Drosophila LRRK2 homolog in Rab7-dependent lysosomal positioning. Hum Mol Genet 21: 1350-1363. PubMed ID: 22171073

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

  • Vladimir Gvozdev Laboratory of Biochemical Genetics of Animals, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow
    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

    Klenov, M. S., Lavrov, S. A., Korbut, A. P., Stolyarenko, A. D., Yakushev, E. Y., Reuter, M., Pillai, R. S. and Gvozdev, V. A. (2014). Impact of nuclear Piwi elimination on chromatin state in Drosophila melanogaster ovaries. Nucleic Acids Res. PubMed ID: 24782529

    Kibanov, M. V., Gvozdev, V. A. and Olenina, L. V. (2012). Germ granules in spermatogenesis of Drosophila: Evidences of contribution to the piRNA silencing. Commun Integr Biol 5: 130-133. PubMed ID: 22808315

    Olenkina, O. M., Egorova, K. S., Kibanov, M. V., Gervaziev, Y. V., Gvozdev, V. A. and Olenina, L. V. (2012). Promoter contribution to the testis-specific expression of Stellate gene family in Drosophila melanogaster. Gene 499: 143-153. PubMed ID: 22425977


  • Return to The Interactive Fly