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


  • Qi Dai Department of Molecular Biosciences, Stockholm University
    Ueberschar, M., Wang, H., Zhang, C., Kondo, S., Aoki, T., Schedl, P., Lai, E. C., Wen, J. and Dai, Q. (2019). BEN-solo factors partition active chromatin to ensure proper gene activation in Drosophila. Nat Commun 10(1): 5700. PubMed ID: 31836703

    Yao, L., Wang, S., Westholm, J. O., Dai, Q., Matsuda, R., Hosono, C., Bray, S., Lai, E. C. and Samakovlis, C. (2017). Genome-wide identification of Grainy head targets in Drosophila reveals regulatory interactions with the POU domain transcription factor Vvl. Development 144(17): 3145-3155. PubMed ID: 28760809

    Milton, C. C., Grusche, F. A., Degoutin, J. L., Yu, E., Dai, Q., Lai, E. C. and Harvey, K. F. (2014). The Hippo pathway regulates hematopoiesis in Drosophila melanogaster. Curr Biol 24(22): 2673-2680. PubMed ID: 25454587

  • Anupama Dahanukar Department of Entomology and Nematology, U. C. Davis
    Dey, M., Ganguly, A. and Dahanukar, A. (2023). An inhibitory mechanism for suppressing high salt intake in Drosophila. Chem Senses 48. PubMed ID: 37201555

    Clark, J. T., Ganguly, A., Ejercito, J., Luy, M., Dahanukar, A. and Ray, A. (2023). Chemosensory detection of aversive concentrations of ammonia and basic volatile amines in insects. iScience 26(1): 105777. PubMed ID: 36594011

    Ganguly, A., Dey, M., Scott, C., Duong, V. K. and Dahanukar, A. (2021). Dietary Macronutrient Imbalances Lead to Compensatory Changes in Peripheral Taste via Independent Signaling Pathways. J Neurosci. PubMed ID: 34753739

    Brown, E. B., Shah, K. D., Palermo, J., Dey, M., Dahanukar, A. and Keene, A. C. (2021). Ir56d-dependent fatty acid responses in Drosophila uncovers taste discrimination between different classes of fatty acids. Elife 10. PubMed ID: 33949306

    Chen, Y. D., Ahmad, S., Amin, K. and Dahanukar, A. (2019). A subset of brain neurons controls regurgitation in adult Drosophila melanogaster. J Exp Biol. PubMed ID: 31511344

    Chen, Y. D. and Dahanukar, A. (2017). Molecular and cellular organization of taste neurons in aedult Drosophila pharynx. Cell Rep 21(10): 2978-2991. PubMed ID: 29212040

    Ganguly, A., Pang, L., Duong, V.K., Lee, A., Schoniger, H., Varady, E. and Dahanukar, A. (2017). A molecular and cellular context-dependent role for Ir76b in detection of amino acid taste. Cell Rep 18: 737-750. PubMed ID: 28099851

    Freeman, E. G. and Dahanukar, A. (2015). Molecular neurobiology of Drosophila taste. Curr Opin Neurobiol 34: 140-148. PubMed ID: 26102453

    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

    Kain, P. and Dahanukar, A. (2015). Secondary taste neurons that convey sweet taste and starvation in the Drosophila brain. Neuron [Epub ahead of print]. PubMed ID: 25661186

    Liu, Y. C., Pearce, M. W., Honda, T., Johnson, T. K., Charlu, S., Sharma, K. R., Imad, M., Burke, R. E., Zinsmaier, K. E., Ray, A., Dahanukar, A., de Bruyne, M. and Warr, C. G. (2014). The Drosophila melanogaster Phospholipid Flippase dATP8B Is Required for Odorant Receptor Function. PLoS Genet 10: e1004209. PubMed ID: 24651716

    Freeman, E. G., Wisotsky, Z. and Dahanukar, A. (2014). Detection of sweet tastants by a conserved group of insect gustatory receptors. Proc Natl Acad Sci U S A 111: 1598-1603. PubMed ID: 24474785

  • Christian Dahmann Max Planck Institute of Molecular Cell Biology and Genetics, Dresden
    Pierini, G. and Dahmann, C. (2023). Hedgehog morphogen gradient is robust towards variations in tissue morphology in Drosophila. Sci Rep 13(1): 8454. PubMed ID: 37231029

    Sui, L. and Dahmann, C. (2020). Increased lateral tension is sufficient for epithelial folding in Drosophila. Development 147(23). PubMed ID: 33277300

    Wang, J., Michel, M., Bialas, L., Pierini, G. and Dahmann, C. (2023). Preferential recruitment and stabilization of Myosin II at compartment boundaries in Drosophila. J Cell Sci. PubMed ID: 36718636

    Sui, L. and Dahmann, C. (2020). Wingless counteracts epithelial folding by increasing mechanical tension at basal cell edges in Drosophila. Development 147(5). PubMed ID: 32161062

    Michel, M. and Dahmann, C. (2020). Tissue mechanical properties modulate cell extrusion in the Drosophila abdominal epidermis. Development 147(5). PubMed ID: 32161061

    Sui, L., Alt, S., Weigert, M., Dye, N., Eaton, S., Jug, F., Myers, E. W., Julicher, F., Salbreux, G. and Dahmann, C. (2018). Differential lateral and basal tension drive folding of Drosophila wing discs through two distinct mechanisms. Nat Commun 9(1): 4620. PubMed ID: 30397306

    Michel, M., Aliee, M., Rudolf, K., Bialas, L., Julicher, F. and Dahmann, C. (2016). The selector gene apterous and Notch are required to locally increase mechanical cell bond tension at the Drosophila dorsoventral compartment boundary. PLoS One 11: e0161668. PubMed ID: 27552097

    Michel, M. and Dahmann, C. (2016). Regulating mechanical tension at compartment boundaries in Drosophila. Fly (Austin): [Epub ahead of print]. PubMed ID: 27362651

    Aurich, F. and Dahmann, C. (2016). A mutation in fat2 uncouples tissue elongation from global tissue rotation. Cell Rep [Epub ahead of print]. PubMed ID: 26972006

    Rudolf, K., Umetsu, D., Aliee, M., Sui, L., Julicher, F. and Dahmann, C. (2015). A local difference in Hedgehog signal transduction increases mechanical cell bond tension and biases cell intercalations along the Drosophila anteroposterior compartment boundary. Development 142: 3845-3858. PubMed ID: 26577205

  • Ramanuj Dasgupta NYU School of Medicine
    Pancratov, R., Peng, F., Smibert, P., Yang, J. S., Olson, E. R., Guha-Gilford, C., Kapoor, A. J., Liang, F. X., Lai, E. C., Flaherty, M. S. and Dasgupta, R. (2013). The miR-310/13 cluster antagonizes beta-catenin function in the regulation of germ and somatic cell differentiation in the Drosophila testis. Development 140: 2904-2916. PubMed ID: 23821034

    Olson, E. R., Pancratov, R., Chatterjee, S. S., Changkakoty, B., Pervaiz, Z. and DasGupta, R. (2011). Yan, an ETS-domain transcription factor, negatively modulates the Wingless pathway in the Drosophila eye. EMBO Rep 12: 1047-1054. PubMed ID: 21869817

    Gonsalves, F. C., Klein, K., Carson, B. B., Katz, S., Ekas, L. A., Evans, S., Nagourney, R., Cardozo, T., Brown, A. M. and DasGupta, R. (2011). An RNAi-based chemical genetic screen identifies three small-molecule inhibitors of the Wnt/wingless signaling pathway. Proc Natl Acad Sci U S A 108: 5954-5963. PubMed ID: 21393571

  • Mary Dasso Section on Cell Cycle Regulation, NICHD, NIH, Bethesda, MD
    Chen, S., Lyanguzova, M., Kaufhold, R., Plevock Haase, K. M., Lee, H., Arnaoutov, A. and Dasso, M. (2021). Association of RanGAP to nuclear pore complex component, RanBP2/Nup358, is required for pupal development in Drosophila. Cell Rep 37(13): 110151. PubMed ID: 34965423

    Arnaoutov, A., Lee, H., Plevock Haase, K., Aksenova, V., Jarnik, M., Oliver, B., Serpe, M. and Dasso, M. (2020). IRBIT Directs Differentiation of Intestinal Stem Cell Progeny to Maintain Tissue Homeostasis. iScience 23(3): 100954. PubMed ID: 32179478

    Dasso, M. (2017). Catch and release: 14-3-3 controls Ncd in meiotic spindles. J Cell Biol 216(10): 3003-3005. PubMed ID: 28923976

    O'Rourke, J. G., Gareau, J. R., Ochaba, J., Song, W., Rasko, T., Reverter, D., Lee, J., Monteys, A. M., Pallos, J., Mee, L., Vashishtha, M., Apostol, B. L., Nicholson, T. P., Illes, K., Zhu, Y. Z., Dasso, M., Bates, G. P., Difiglia, M., Davidson, B., Wanker, E. E., Marsh, J. L., Lima, C. D., Steffan, J. S. and Thompson, L. M. (2013). SUMO-2 and PIAS1 modulate insoluble mutant huntingtin protein accumulation. Cell Rep 4(2): 362-375. PubMed ID: 23871671

  • Brigitte Dauwalder Biology and Biochemistry, University of Houston
    Love, C. R., Gautam, S., Lama, C., Le, N. H. and Dauwalder, B. (2023). The Drosophila dopamine 2-like receptor D2R (Dop2R) is required in the blood brain barrier for male courtship. Genes Brain Behav 22(1): e12836. PubMed ID: 36636829

    Lama, C., Love, C. R., Le, H. N., Waqar, M., Reeve, J. L., Lama, J. and Dauwalder, B. (2022). The nuclear receptor Hr46/Hr3 is required in the blood brain barrier of mature males for courtship. PLoS Genet 18(1): e1009519. PubMed ID: 35077443

    Saurabh, S., Vanaphan, N., Wen, W. and Dauwalder, B. (2018). High functional conservation of takeout family members in a courtship model system. PLoS One 13(9): e0204615. PubMed ID: 30261021
    Wijesekera, T.P., Saurabh, S and Dauwalder, B. (2016). Juvenile hormone is required in adult males for Drosophila courtship. PLoS One 11: e0151912. PubMed ID: 27003411

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

    Vanaphan, N., Dauwalder, B. and Zufall, R. A. (2012). Diversification of takeout, a male-biased gene family in Drosophila. Gene 491: 142-148. PubMed ID: 22020223

  • Shireen Davies Institute of Molecular Cell and Systems Biology. University of Glasgow
    Cabrero, P., Terhzaz, S., Dornan, A. J., Ghimire, S., Holmes, H. L., Turin, D. R., Romero, M. F., Davies, S. A. and Dow, J. A. T. (2020). Specialized stellate cells offer a privileged route for rapid water flux in Drosophila renal tubule. Proc Natl Acad Sci U S A 117(3): 1779-1787. PubMed ID: 31907321

    Martinez-Corrales, G., Cabrero, P., Dow, J. A. T., Terhzaz, S. and Davies, S. A. (2019). Novel roles for GATAe in growth, maintenance and proliferation of cell populations in the Drosophila renal tubule. Development 146(9). PubMed ID: 31036543

    Davies, S. A., Cabrero, P., Marley, R., Corrales, G. M., Ghimire, S., Dornan, A. J. and Dow, J. A. T. (2019). Epithelial function in the Drosophila Malpighian tubule: An in vivo renal model. Methods Mol Biol 1926: 203-221. PubMed ID: 30742274

    Overend, G., Luo, Y., Henderson, L., Douglas, A.E., Davies, S.A. and Dow, J.A. (2016). Molecular mechanism and functional significance of acid generation in the Drosophila midgut. Sci Rep 6: 27242. PubMed ID: 27250760

    Halberg, K.A., Rainey, S.M., Veland, I.R., Neuert, H., Dornan, A.J., Klämbt, C., Davies, S.A. and Dow, J.A. (2016). The cell adhesion molecule Fasciclin2 regulates brush border length and organization in Drosophila renal tubules. Nat Commun 7: 11266. PubMed ID: 27072072

    Cannell, E., Dornan, A. J., Halberg, K. A., Terhzaz, S., Dow, J. A. and Davies, S. A. (2016). The corticotropin-releasing factor-like diuretic hormone 44 (DH) and kinin neuropeptides modulate desiccation and starvation tolerance in Drosophila melanogaster. Peptides [Epub ahead of print]. PubMed ID: 26896569

    Brinzer, R. A., Henderson, L., Marchiondo, A. A., Woods, D. J., Davies, S. A. and Dow, J. A. (2015). Metabolomic profiling of permethrin-treated Drosophila melanogaster identifies a role for tryptophan catabolism in insecticide survival. Insect Biochem Mol Biol. PubMed ID: 26474926

    Terhzaz, S., Cabrero, P., Brinzer, R. A., Halberg, K. A., Dow, J. A. and Davies, S. A. (2015). A novel role of Drosophila cytochrome P450-4e3 in permethrin insecticide tolerance. Insect Biochem Mol Biol [Epub ahead of print]. PubMed ID: 26073628

    Halberg, K. A., Terhzaz, S., Cabrero, P., Davies, S. A. and Dow, J. A. (2015). Tracing the evolutionary origins of insect renal function. Nat Commun 6: 6800. PubMed ID: 25896425

    Terhzaz, S., Teets, N. M., Cabrero, P., Henderson, L., Ritchie, M. G., Nachman, R. J., Dow, J. A., Denlinger, D. L. and Davies, S. A. (2015). Insect capa neuropeptides impact desiccation and cold tolerance. Proc Natl Acad Sci U S A 112: 2882-2887. PubMed ID: 25730885

  • Pier Paolo D'Avino Department of Pathology, Cambridge Cancer Center
    Bassi, Z. I., Audusseau, M., Riparbelli, M. G., Callaini, G., D'Avino, P. P. (2013) Citron kinase controls a molecular network required for midbody formation in cytokinesis. Proc Natl Acad Sci U S A. PubMed ID: 23716662

    Colozza, G., Montembault, E., Quenerch'du, E., Riparbelli, M. G., D'Avino, P. P. and Callaini, G. (2011). Drosophila nucleoporin Nup154 controls cell viability, proliferation and nuclear accumulation of Mad transcription factor. Tissue Cell 43: 254-261. PubMed ID: 21696798

    Montembault, E., Zhang, W., Przewloka, M. R., Archambault, V., Sevin, E. W., Laue, E. D., Glover, D. M. and D'Avino, P. P. (2010). Nessun Dorma, a novel centralspindlin partner, is required for cytokinesis in Drosophila spermatocytes. J Cell Biol 191: 1351-1365. PubMed ID: 21187330

  • Graeme Davis University of California San Francisco
    Orr, B. O., Fetter, R. D. and Davis, G. W. (2022). Activation and expansion of presynaptic signaling foci drives presynaptic homeostatic plasticity. Neuron. PubMed ID: 36087584

    Johnson, A. E., Orr, B. O., Fetter, R. D., Moughamian, A. J., Primeaux, L. A., Geier, E. G., Yokoyama, J. S., Miller, B. L. and Davis, G. W. (2021). SVIP is a molecular determinant of lysosomal dynamic stability, neurodegeneration and lifespan. Nat Commun 12(1): 513. PubMed ID: 33479240

    Genc, O., An, J. Y., Fetter, R. D., Kulik, Y., Zunino, G., Sanders, S. J. and Davis, G. W. (2020). Homeostatic plasticity fails at the intersection of autism-gene mutations and a novel class of common genetic modifiers. Elife 9. PubMed ID: 32609087

    Harris, N., Fetter, R. D., Brasier, D. J., Tong, A. and Davis, G. W. (2018). Molecular interface of neuronal innate immunity, synaptic vesicle stabilization, and presynaptic homeostatic plasticity. Neuron. PubMed ID: 30344041

    Hauswirth, A. G., Ford, K. J., Wang, T., Fetter, R. D., Tong, A. and Davis, G. W. (2018). A postsynaptic PI3K-cII dependent signaling controller for presynaptic homeostatic plasticity. Elife 7. PubMed ID: 29303480

    Smart, A. D., Pache, R. A., Thomsen, N. D., Kortemme, T., Davis, G. W. and Wells, J. A. (2017). Engineering a light-activated caspase-3 for precise ablation of neurons in vivo. Proc Natl Acad Sci U S A 114(39): E8174-e8183. PubMed ID: 28893998

    Orr, B. O., Fetter, R. D. and Davis, G. W. (2017). Retrograde semaphorin-plexin signalling drives homeostatic synaptic plasticity. Nature. PubMed ID: 28953869

    Wang, T., Jones, R. T., Whippen, J. M. and Davis, G. W. (2016). alpha2delta-3 Is Required for Rapid Transsynaptic Homeostatic Signaling. Cell Rep 16: 2875-2888. PubMed ID: 27626659

    Harris, N., Braiser, D. J., Dickman, D. K., Fetter, R. D., Tong, A. and Davis, G. W. (2015). The innate immune receptor PGRP-LC controls presynaptic homeostatic plasticity. Neuron 88: 1157-1164. PubMed ID: 26687223

    Johnson, A. E., Shu, H., Hauswirth, A. G., Tong, A. and Davis, G. W. (2015). VCP-dependent muscle degeneration is linked to defects in a dynamic tubular lysosomal network. Elife 4. PubMed ID: 26167652

    Muller, M., Genc, O. and Davis, G. W. (2015). RIM-binding protein links synaptic homeostasis to the stabilization and replenishment of high release probability vesicles. Neuron 85: 1056-1069. PubMed ID: 25704950

    Gavino, M. A., Ford, K. J., Archila, S. and Davis, G. W. (2015). Homeostatic synaptic depression is achieved through a regulated decrease in presynaptic calcium channel abundance. Elife 4. PubMed ID: 25884248

  • Ilan Davis Department of Biochemistry, University of Oxford
    Thompson, M. K., Ceccarelli, A., Ish-Horowicz, D. and Davis, I. (2023). Dynamically regulated transcription factors are encoded by highly unstable mRNAs in the Drosophila larval brain. Rna. PubMed ID: 37041032

    Mitchell, J., Smith, C. S., Titlow, J., Otto, N., van Velde, P., Booth, M., Davis, I. and Waddell, S. (2021). Selective dendritic localization of mRNA in Drosophila mushroom body output neurons. Elife 10. PubMed ID: 33724180

    Samuels, T. J., Arava, Y., Jarvelin, A. I., Robertson, F., Lee, J. Y., Yang, L., Yang, C. P., Lee, T., Ish-Horowicz, D. and Davis, I. (2020). Neuronal upregulation of Prospero protein is driven by alternative mRNA polyadenylation and Syncrip-mediated mRNA stabilisation. Biol Open. PubMed ID: 32205310

    Titlow, J., Robertson, F., Jarvelin, A., Ish-Horowicz, D., Smith, C., Gratton, E. and Davis, I. (2020). Syncrip/hnRNP Q is required for activity-induced Msp300/Nesprin-1 expression and new synapse formation. J Cell Biol 219(3). PubMed ID: 32040548

    Samuels, T. J., Jarvelin, A. I., Ish-Horowicz, D. and Davis, I. (2020). Imp/IGF2BP levels modulate individual neural stem cell growth and division through myc mRNA stability. Elife 9. PubMed ID: 31934860

    Titlow, J. S., Yang, L., Parton, R. M., Palanca, A. and Davis, I. (2018). Super-resolution single molecule FISH at the Drosophila neuromuscular junction. Methods Mol Biol 1649: 163-175. PubMed ID: 29130196

    Yang, L., Titlow, J., Ennis, D., Smith, C., Mitchell, J., Young, F. L., Waddell, S., Ish-Horowicz, D. and Davis, I. (2017). Single molecule fluorescence in situ hybridisation for quantitating post-transcriptional regulation in Drosophila brains. Methods. PubMed ID: 28651965

    Davidson, A., Parton, R. M., Rabouille, C., Weil, T. T. and Davis, I. (2016). Localized Translation of gurken/TGF-alpha mRNA during Axis Specification Is Controlled by Access to Orb/CPEB on Processing Bodies. Cell Rep 14: 2451-2462. PubMed ID: 26947065

    Halstead, J. M., Lin, Y. Q., Durraine, L., Hamilton, R. S., Ball, G., Neely, G. G., Bellen, H. J. and Davis, I. (2014). Syncrip/hnRNP Q influences synaptic transmission and regulates BMP signaling at the Drosophila neuromuscular synapse. Biol Open 3: 839-849. PubMed ID: 25171887

    McDermott, S. M., Yang, L., Halstead, J. M., Hamilton, R. S., Meignin, C. and Davis, I. (2014). Drosophila Syncrip modulates the expression of mRNAs encoding key synaptic proteins required for morphology at the neuromuscular junction. RNA. PubMed ID: 25171822

  • Ron Davis Scripps Research Institute, Jupiter, Florida
    Noyes, N. C. and Davis, R. L. (2023). Innate and learned odor-guided behaviors utilize distinct molecular signaling pathways in a shared dopaminergic circuit. Cell Rep 42(2): 112026. PubMed ID: 36701232

    Zhang, X., Sabandal, J. M., Tsaprailis, G. and Davis, R. L. (2022). Active forgetting requires Sickie function in a dedicated dopamine circuit in Drosophila. Proc Natl Acad Sci U S A 119(38): e2204229119. PubMed ID: 36095217

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

    Sabandal, J. M., Berry, J. A. and Davis, R. L. (2021). Dopamine-based mechanism for transient forgetting. Nature. PubMed ID: 33473212

    Cervantes-Sandoval, I., Davis, R. L. and Berry, J. A. (2020). Rac1 Impairs Forgetting-Induced Cellular Plasticity in Mushroom Body Output Neurons. Front Cell Neurosci 14: 258. PubMed ID: 33061890

    Wang, X. and Davis, R. L. (2020). Early Mitochondrial Fragmentation and Dysfunction in a Drosophila Model for Alzheimer's Disease. Mol Neurobiol. PubMed ID: 32909149

    Guven-Ozkan, T., Busto, G. U., Jung, J. Y., Drago, I. and Davis, R. L. (2020). miR-92a Suppresses Mushroom Body-Dependent Memory Consolidation in Drosophila. eNeuro. PubMed ID: 32737186

    Noyes, N. C., Walkinshaw, E. and Davis, R. L. (2020). Ras acts as a molecular switch between two forms of consolidated memory in Drosophila. Proc Natl Acad Sci U S A 117(4): 2133-2139. PubMed ID: 31932418

    Zhang, X., Noyes, N. C., Zeng, J., Li, Y. and Davis, R. L. (2019). Aversive training induces both pre- and postsynaptic suppression in Drosophila. J Neurosci. PubMed ID: 31558620

    Pacifico, R., MacMullen, C. M., Walkinshaw, E., Zhang, X. and Davis, R. L. (2018). Brain transcriptome changes in the aging Drosophila melanogaster accompany olfactory memory performance deficits. PLoS One 13(12): e0209405. PubMed ID: 30576353

    Phan, A., Thomas, C. I., Chakraborty, M., Berry, J. A., Kamasawa, N. and Davis, R. L. (2018). Stromalin constrains memory scquisition by developmentally limiting synaptic vesicle pool size. Neuron. PubMed ID: 30503644

    Berry, J. A., Phan, A. and Davis, R. L. (2018). Dopamine neurons mediate learning and forgetting through bidirectional modulation of a memory trace. Cell Rep 25(3): 651-662.e655. PubMed ID: 30332645

    Crittenden, J. R., Skoulakis, E. M. C., Goldstein, E. S. and Davis, R. L. (2018). Drosophila mef2 is essential for normal mushroom body and wing development. Biol Open. PubMed ID: 30115617

  • Ken Dawson-Scully Biological Sciences, Florida Atlantic University, Boca Raton
    Bollinger, W. L., Sial, N. and Dawson-Scully, K. (2018). BK channels and a cGMP-dependent protein kinase (PKG) function through independent mechanisms to regulate the tolerance of synaptic transmission to acute oxidative stress at the Drosophila larval neuromuscular junction. J Neurogenet 32(3): 246-255. PubMed ID: 30484391

    Krill, J. L. and Dawson-Scully, K. (2016). cGMP-Dependent Protein Kinase Inhibition Extends the Upper Temperature Limit of Stimulus-Evoked Calcium Responses in Motoneuronal Boutons of Drosophila melanogaster Larvae. PLoS One 11: e0164114. PubMed ID: 27711243

    Caplan, S. L., Milton, S. L. and Dawson-Scully, K. (2013). A cGMP-dependent protein kinase (PKG) controls synaptic transmission tolerance to acute oxidative stress at the Drosophila larval neuromuscular junction. J Neurophysiol 109: 649-658. PubMed ID: 23136350

    Armstrong, G. A., Xiao, C., Krill, J. L., Seroude, L., Dawson-Scully, K. and Robertson, R. M. (2011). Glial Hsp70 protects K+ homeostasis in the Drosophila brain during repetitive anoxic depolarization. PLoS One 6: e28994. PubMed ID: 22174942

  • Péter Deák Institute of Biochemistry, Hungarian Academy of Sciences, Szeged
    Kovacs, L., Nagy, A., Pal, M. and Deak, P. (2020). Usp14 is required for spermatogenesis and ubiquitin stress responses in Drosophila melanogaster. J Cell Sci 133(2). PubMed ID: 31974276

    Nagy, A., Kovacs, L., Lipinszki, Z., Pal, M. and Deak, P. (2018). Developmental and tissue specific changes of ubiquitin forms in Drosophila melanogaster. PLoS One 13(12): e0209080. PubMed ID: 30543682

    Kovacs, L., Nagy, O., Pal, M., Udvardy, A., Popescu, O. and Deak, P. (2015). Role of the deubiquitylating enzyme DmUsp5 in coupling ubiquitin equilibrium to development and apoptosis in Drosophila melanogaster. PLoS One 10: e0120875. PubMed ID: 25806519

    Kokai, E., Paldy, F. S., Somogyi, K., Chougule, A., Pal, M., Kerekes, E., Deak, P., Friedrich, P., Dombradi, V. and Adam, G. (2012). CalpB modulates border cell migration in Drosophila egg chambers. BMC Dev Biol 12: 20. PubMed ID: 22827336

    Nagy, O., Pal, M., Udvardy, A., Shirras, C. A., Boros, I., Shirras, A. D. and Deak, P. (2012). lemmingA encodes the Apc11 subunit of the APC/C in Drosophila melanogaster that forms a ternary complex with the E2-C type ubiquitin conjugating enzyme, Vihar and Morula/Apc2. Cell Div 7: 9. PubMed ID: 22417125

    Lipinszki, Z., Kovacs, L., Deak, P. and Udvardy, A. (2012). Ubiquitylation of Drosophila p54/Rpn10/S5a regulates its interaction with the UBA-UBL polyubiquitin receptors. Biochemistry 51: 2461-2470. PubMed ID: 22364263

    Lipinszki, Z., Pal, M., Nagy, O., Deak, P., Hunyadi-Gulyas, E. and Udvardy, A. (2011). Overexpression of Dsk2/dUbqln results in severe developmental defects and lethality in Drosophila melanogaster that can be rescued by overexpression of the p54/Rpn10/S5a proteasomal subunit. FEBS J 278: 4833-4844. PubMed ID: 21973017

  • Benjamin de Bivort Department of Organismic and Evolutionary Biology, Harvard
    Smith, M. A., Honegger, K. S., Turner, G. and de Bivort, B. (2022). Idiosyncratic learning performance in flies. Biol Lett 18(2): 20210424. PubMed ID: 35104427 Sood, C., Justis, V. T., Doyle, S. E. and Siegrist, S. E. (2022). Notch signaling regulates neural stem cell quiescence entry and exit in Drosophila. Development 149(4). PubMed ID: 35112131

    Werkhoven, Z., Bravin, A., Skutt-Kakaria, K., Reimers, P., Pallares, L. F., Ayroles, J. and de Bivort, B. L. (2021). The structure of behavioral variation within a genotype. Elife 10. PubMed ID: 34664550

    Krams, I. A., Krama, T., Krams, R., Trakimas, G., Popovs, S., Jõers, P., Munkevics, M., Elferts, D., Rantala, M. J., Makņa, J. and de Bivort, B. L. (2021). Serotoninergic Modulation of Phototactic Variability Underpins a Bet-Hedging Strategy in Drosophila melanogaster. Front Behav Neurosci 15: 659331. PubMed ID: 33935664

    Akhund-Zade, J., Lall, S., Gajda, E., Yoon, D., Ayroles, J. F. and de Bivort, B. L. (2021). Genetic basis of offspring number-body weight tradeoff in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 33871609

    Versace, E., Caffini, M., Werkhoven, Z. and de Bivort, B. L. (2020). Individual, but not population asymmetries, are modulated by social environment and genotype in Drosophila melanogaster. Sci Rep 10(1): 4480. PubMed ID: 32161330

    Honegger, K. S., Smith, M. A., Churgin, M. A., Turner, G. C. and de Bivort, B. L. (2019). Idiosyncratic neural coding and neuromodulation of olfactory individuality in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 31455738

    Akhund-Zade, J., Ho, S., O'Leary, C. and de Bivort, B. (2019). The effect of environmental enrichment on behavioral variability depends on genotype, behavior, and type of enrichment. J Exp Biol. PubMed ID: 31413102

    Xu, C., Theisen, E., Maloney, R., Peng, J., Santiago, I., Yapp, C., Werkhoven, Z., Rumbaut, E., Shum, B., Tarnogorska, D., Borycz, J., Tan, L., Courgeon, M., Meinertzhagen, I. A., de Bivort, B., Drugowitsch, J. and Pecot, M. Y. (2019). Control of synaptic specificity by establishing a relative preference for synaptic partners. Neuron. PubMed ID: 31300277

    Isakov, A., Buchanan, S. M., Sullivan, B., Ramachandran, A., Chapman, J. K., Lu, E. S., Mahadevan, L. and de Bivort, B. (2016). Recovery of locomotion after injury in Drosophila depends on proprioception. J Exp Biol. PubMed ID: 26994176

    Kain, J. S., Zhang, S., Akhund-Zade, J., Samuel, A. D., Klein, M. and de Bivort, B. L. (2015). Variability in thermal and phototactic preferences in Drosophila may reflect an adaptive bet-hedging strategy. Evolution 69: 3171-3185. PubMed ID: 26531165

    Buchanan, S. M., Kain, J. S. and de Bivort, B. L. (2015). Neuronal control of locomotor handedness in Drosophila. Proc Natl Acad Sci U S A 112: 6700-6705. PubMed ID: 25953337

  • Marien de Bruyne Monash University, Melbourne
    Liu, Y. C., Pearce, M. W., Honda, T., Johnson, T. K., Charlu, S., Sharma, K. R., Imad, M., Burke, R. E., Zinsmaier, K. E., Ray, A., Dahanukar, A., de Bruyne, M. and Warr, C. G. (2014). The Drosophila melanogaster Phospholipid Flippase dATP8B Is Required for Odorant Receptor Function. PLoS Genet 10: e1004209. PubMed ID: 24651716

    Faucher, C. P., Hilker, M., de Bruyne, M. (2013) Interactions of carbon dioxide and food odours in Drosophila: olfactory hedonics and sensory neuron properties. PLoS One 8: e56361. Pubmed ID: 23457557

    Tunstall, N. E., Herr, A., de Bruyne, M. and Warr, C. G. (2012). A screen for genes expressed in the olfactory organs of Drosophila melanogaster identifies genes involved in olfactory behaviour. PLoS One 7: e35641. PubMed ID: 22530061

    Tom, W., de Bruyne, M., Haehnel, M., Carlson, J. R. and Ray, A. (2011). Disruption of olfactory receptor neuron patterning in Scutoid mutant Drosophila. Mol Cell Neurosci 46: 252-261. PubMed ID: 20875862

  • José Felix de Celis CBM Severo Ochoa, Madrid
    Ostale, C. M., Vega-Cuesta, P., Gonzalez, T., Lopez-Varea, A. and de Celis, J. F. (2023). RNAi screen in the Drosophila wing of genes encoding proteins related to cytoskeleton organization and cell division. Dev Biol 498: 61-76. PubMed ID: 37015290

    Ostala, C. M., Esteban, N., Lapez-Varea, A. and de Celis, J. F. (2021). Functional requirements of protein kinases and phosphatases in the development of the Drosophila melanogaster wing. G3 (Bethesda). PubMed ID: 34599799

    Lapez-Varea, A., Ostale, C. M., Vega-Cuesta, P., Ruiz-Gomez, A., Organista, M. F., Martan, M., Hevia, C. F., Molnar, C., de Celis, J., Culi, J., Esteban, N. and de Celis, J. F. (2021). Genome-wide Phenotypic RNAi Screen in the Drosophila Wing: Global Parameters. G3 (Bethesda). PubMed ID: 34599819

    Beatty, J. S., Molnar, C., Luque, C. M., de Celis, J. F. and Martin-Bermudo, M. D. (2021). EGFRAP encodes a new negative regulator of the EGFR acting in both normal and oncogenic EGFR/Ras-driven tissue morphogenesis. PLoS Genet 17(8): e1009738. PubMed ID: 34411095

    Vega-Cuesta, P., Ruiz-Gomez, A., Molnar, C., Organista, M. F., Resnik-Docampo, M., Falo-Sanjuan, J., Lopez-Varea, A. and de Celis, J. F. (2020). Ras2, the TC21/R-Ras2 Drosophila homologue, contributes to insulin signalling but is not required for organism viability. Dev Biol. PubMed ID: 32061885

    Molnar, C., Estrada, B. and de Celis, J. F. (2018). Tay bridge and ERK activity are required for motoneuron function in the Drosophila neural system. Genes Brain Behav. PubMed ID: 29524312

    Martin, M., Ostale, C. M. and de Celis, J. F. (2017). Patterning of the Drosophila L2 vein is driven by regulatory interactions between region-specific transcription factors expressed in response to Dpp signalling. Development. PubMed ID: 28760811

    Hevia, C. F., Lopez-Varea, A., Esteban, N. and de Celis, J. F. (2017). A search for genes mediating the growth promoting function of TGFbeta in the Drosophila melanogaster wing disc. Genetics [Epub ahead of print]. PubMed ID: 28315837

    Organista, M. F., Martin, M., de Celis, J. M., Barrio, R., Lopez-Varea, A., Esteban, N., Casado, M. and de Celis, J. F. (2015). The Spalt Transcription Factors Generate the Transcriptional Landscape of the Drosophila melanogaster Wing Pouch Central Region. PLoS Genet 11: e1005370. PubMed ID: 26241320

    Molnar, C. and de Celis, J. F. (2013). Tay Bridge Is a Negative Regulator of EGFR Signalling and Interacts with Erk and Mkp3 in theDrosophila melanogaster Wing. PLoS Genet 9: e1003982. PubMed ID: 24348264

    Hevia, C. F. and de Celis, J. F. (2013). Activation and function of TGFbeta signalling during Drosophila wing development and its interactions with the BMP pathway. Dev Biol. PubMed ID: 23485686

  • David Deitcher Department of Neurobiology and Behavior, Cornell University
    Dean, D., Weinstein, H., Amin, S., Karno, B., McAvoy, E., Hoy, R., Recknagel, A., Jarvis, C. and Deitcher, D. (2017). Extending julius seizure, a bang-sensitive gene, as a model for studying epileptogenesis: Cold shock, and a new insertional mutation. Fly (Austin): [Epub ahead of print] PubMed ID: 29125376

    Horne, M., Krebushevski, K., Wells, A., Tunio, N., Jarvis, C., Francisco, G., Geiss, J., Recknagel, A. and Deitcher, D. L. (2017). julius seizure, a Drosophila Mutant, Defines a Neuronal Population Underlying Epileptogenesis. Genetics. PubMed ID: 28082408

    Dean, D. M., Maroja, L. S., Cottrill, S., Bomkamp, B. E., Westervelt, K. A. and Deitcher, D. L. (2015). The wavy Mutation Maps to the Inositol 1,4,5-Trisphosphate 3-Kinase 2 (IP3K2) Gene of Drosophila and Interacts with IP3R to Affect Wing Development. G3 (Bethesda) 6(2): 299-310. PubMed ID: 26613949

    Lawton, K. J., Wassmer, T. L. and Deitcher, D. L. (2014). Conserved role of Drosophila melanogaster FoxP in motor coordination and courtship song. Behav Brain Res 268: 213-221. PubMed ID: 24747661

  • Andrés Dekanty Facultad de Bioquímica y Ciencias Biologicas, Universidad Nacional del Litoral, Santa Fe 3000, Argentina.
    Sanchez, J. A., Ingaramo, M. C., Gerve, M. P., Thomas, M. G., Boccaccio, G. L. and Dekanty, A. (2023). FOXO-mediated repression of Dicer1 regulates metabolism, stress resistance, and longevity in Drosophila. Proc Natl Acad Sci U S A 120(15): e2216539120. PubMed ID: 37014862

    Ingaramo, M. C., Sanchez, J. A., Perrimon, N. and Dekanty, A. (2020). Fat Body p53 Regulates Systemic Insulin Signaling and Autophagy under Nutrient Stress via Drosophila Upd2 Repression. Cell Rep 33(4): 108321. PubMed ID: 33113367

    Sanchez, J. A., Mesquita, D., Ingaramo, M. C., Ariel, F., Milan, M. and Dekanty, A. (2019). Eiger/TNFalpha-mediated Dilp8 and ROS production coordinate intra-organ growth in Drosophila. PLoS Genet 15(8): e1008133. PubMed ID: 31425511

  • Marion Delattre Department of Genetics and Evolution, University of Geneva
    Basquin, D., Spierer, A., Begeot, F., Koryakov, D. E., Todeschini, A. L., Ronsseray, S., Vieira, C., Spierer, P. and Delattre, M. (2014). The Drosophila Su(var)3-7 Gene Is Required for Oogenesis and Female Fertility, Genetically Interacts with piwi and aubergine, but Impacts Only Weakly Transposon Silencing. PLoS One 9: e96802. PubMed ID: 24820312

    Rebollo, R., Horard, B., Begeot, F., Delattre, M., Gilson, E. and Vieira, C. (2012). A snapshot of histone modifications within transposable elements in Drosophila wild type strains. PLoS One 7: e44253. PubMed ID: 22962605

    Deng, H., Cai, W., Wang, C., Lerach, S., Delattre, M., Girton, J., Johansen, J. and Johansen, K. M. (2010). JIL-1 and Su(var)3-7 interact genetically and counteract each other's effect on position-effect variegation in Drosophila. Genetics 185: 1183-1192. PubMed ID: 20457875

  • Christos Delidakis Forth Foundation for Research and Technology, Hellas
    Theodorou, V., Stefanaki, A., Drakos, M., Triantafyllou, D. and Delidakis, C. (2022). ASC proneural factors are necessary for chromatin remodeling during neuroectodermal to neuroblast fate transition to ensure the timely initiation of the neural stem cell program. BMC Biol 20(1): 107. PubMed ID: 35549704

    Voutyraki, C., Choromidis, A., Theodorou, V., Efraimoglou, C., Anagnostopoulos, G., Magadi, S. S., Grammenoudi, S., Zacharioudaki, E. and Delidakis, C. (2021). Repression of differentiation genes by Hes transcription factors fuels neural tumour growth in Drosophila. Int J Dev Biol. PubMed ID: 34881794

    Deliconstantinos, G., Kalodimou, K. and Delidakis, C. (2021). Translational Control of Serrate Expression in Drosophila Cells. In Vivo 35(2): 859-869. PubMed ID: 33622878

    Magadi, S. S., Voutyraki, C., Anagnostopoulos, G., Zacharioudaki, E., Poutakidou, I. K., Efraimoglou, C., Stapountzi, M., Theodorou, V., Nikolaou, C., Koumbanakis, K. A., Fullard, J. F. and Delidakis, C. (2020). Dissecting Hes-centred transcriptional networks in neural stem cell maintenance and tumorigenesis in Drosophila. Development 147(22). PubMed ID: 33229432

    Piwko, P., Vitsaki, I., Livadaras, I. and Delidakis, C. (2019). The role of insulators in transgene transvection in Drosophila. Genetics. PubMed ID: 30948430

    Alexiadis, A., Delidakis, C. and Kalantidis, K. (2017). Snipper, an Eri1 homologue, affects histone mRNA abundance and is crucial for normal Drosophila melanogaster development. FEBS Lett. PubMed ID: 28626879

    Kiparaki, M., Zarifi, I. and Delidakis, C. (2015). bHLH proteins involved in Drosophila neurogenesis are mutually regulated at the level of stability. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 25694512

    Kux, K., Kiparaki, M. and Delidakis, C. (2013). The two Tribolium E(spl) genes show evolutionarily conserved expression and function during embryonic neurogenesis. Mech Dev. PubMed ID: 23485410

    Zacharioudaki, E., Magadi, S. S. and Delidakis, C. (2012) bHLH-O proteins are crucial for Drosophila neuroblast self-renewal and mediate Notch-induced overproliferation. Development 139: 1258-1269. PubMed ID: 22357926

    Monastirioti, M., Giagtzoglou, N,. Koumbanakis, K.A., Zacharioudaki, E., Deligiannaki, M., Wech, I., Almeida, M., Preiss, A., Bray, S. and Delidakis, C. (2010). Drosophila Hey is a target of Notch in asymmetric divisions during embryonic and larval neurogenesis. Development 137: 191-201. PubMed ID: 20040486

  • Fabio Demontis Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis,
    Jiao, J., Curley, M., Graca, F. A., Robles-Murguia, M., Shirinifard, A., Finkelstein, D., Xu, B., Fan, Y. and Demontis, F. (2023). Modulation of protease expression by the transcription factor Ptx1/PITX regulates protein quality control during aging. Cell Rep 42(1): 111970. PubMed ID: 36640359

    Graca, F. A., Rai, M., Hunt, L. C., Stephan, A., Wang, Y. D., Gordon, B., Wang, R., Quarato, G., Xu, B., Fan, Y., Labelle, M. and Demontis, F. (2022). The myokine Fibcd1 is an endogenous determinant of myofiber size and mitigates cancer-induced myofiber atrophy. Nat Commun 13(1): 2370. PubMed ID: 35501350

    Graca, F. A., Sheffield, N., Puppa, M., Finkelstein, D., Hunt, L. C. and Demontis, F. (2021). A large-scale transgenic RNAi screen identifies transcription factors that modulate myofiber size in Drosophila. PLoS Genet 17(11): e1009926. PubMed ID: 34780463

    Hunt, L. C. and Demontis, F. (2021). Age-Related Increase in Lactate Dehydrogenase Activity in Skeletal Muscle Reduces Lifespan in Drosophila. J Gerontol A Biol Sci Med Sci. PubMed ID: 34477202

    Rai, M., Coleman, Z., Curley, M., Nityanandam, A., Platt, A., Robles-Murguia, M., Jiao, J., Finkelstein, D., Wang, Y. D., Xu, B., Fan, Y. and Demontis, F. (2021). Proteasome stress in skeletal muscle mounts a long-range protective response that delays retinal and brain aging. Cell Metab. PubMed ID: 33773104

    Jiao, J., Kavdia, K., Pagala, V., Palmer, L., Finkelstein, D., Fan, Y., Peng, J. and Demontis, F. (2021). An age-downregulated ribosomal RpS28 protein variant regulates the muscle proteome. G3 (Bethesda). PubMed ID: 33974070

    Hunt, L. C., Schadeberg, B., Stover, J., Haugen, B., Pagala, V., Wang, Y. D., Puglise, J., Barton, E. R., Peng, J. and Demontis, F. (2021). Antagonistic control of myofiber size and muscle protein quality control by the ubiquitin ligase UBR4 during aging. Nat Commun 12(1): 1418. PubMed ID: 33658508

    Robles-Murguia, M., Rao, D., Finkelstein, D., Xu, B., Fan, Y. and Demontis, F. (2020). Muscle-derived Dpp regulates feeding initiation via endocrine modulation of brain dopamine biosynthesis. Genes Dev 34(1-2): 37-52. PubMed ID: 31831628

    Hunt, L. C., Stover, J., Haugen, B., Shaw, T. I., Li, Y., Pagala, V. R., Finkelstein, D., Barton, E. R., Fan, Y., Labelle, M., Peng, J. and Demontis, F. (2019). A key role for the ubiquitin ligase UBR4 in myofiber hypertrophy in Drosophila and mice. Cell Rep 28(5): 1268-1281. PubMed ID: 31365869

    Hunt, L. C., Jiao, J., Wang, Y. D., Finkelstein, D., Rao, D., Curley, M., Robles-Murguia, M., Shirinifard, A., Pagala, V. R., Peng, J., Fan, Y. and Demontis, F. (2019). Circadian gene variants and the skeletal muscle circadian clock contribute to the evolutionary divergence in longevity across Drosophila populations. Genome Res 29(8): 1262-1276. PubMed ID: 31249065

    Hunt, L. C. and Demontis, F. (2013). Whole-mount immunostaining of Drosophila skeletal muscle. Nat Protoc 8: 2496-2501. PubMed ID: 24232251

    Demontis, F., Piccirillo, R., Goldberg, A. L. and Perrimon, N. (2013). Mechanisms of skeletal muscle aging: insights from Drosophila and mammalian models. Dis Model Mech 6: 1339-1352. PubMed ID: 24092876

  • Huai Deng Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor
    Neidviecky, E. and Deng, H. (2023). Determination of Complex Formation between Drosophila Nrf2 and GATA4 Factors at Selective Chromatin Loci Demonstrates Transcription Coactivation. Cells 12(6). PubMed ID: 36980279

    Carlson, J., Price, L., Cook, I. and Deng, H. (2021). Drosophila Keap1 xenobiotic response factor regulates developmental transcription through binding to chromatin. Dev Biol. PubMed ID: 34662537

    Si, H., Ma, P., Liang, Q., Yin, Y., Wang, P., Zhang, Q., Wang, S. and Deng, H. (2019). Overexpression of pink1 or parkin in indirect flight muscles promotes mitochondrial proteostasis and extends lifespan in Drosophila melanogaster. PLoS One 14(11): e0225214. PubMed ID: 31714929

    Carlson, J., Swisse, T., Smith, C. and Deng, H. (2019). Regulation of position effect variegation at pericentric heterochromatin by Drosophila Keap1-Nrf2 xenobiotic response factors. Genesis: e23290. PubMed ID: 30888733

    Huang, Y. C., Moreno, H., Row, S., Jia, D. and Deng, W. M. (2018). Germline silencing of UASt depends on the piRNA pathway. J Genet Genomics. PubMed ID: 29789221

    Shu, Z. and Deng, W.M. (2017). Differential regulation of Cyclin E by Yorkie-Scalloped signaling in organ development.G3 (Bethesda) [Epub ahead of print]. PubMed ID: 28143945

    Deng, H. and Kerppola, T. K. (2013). Regulation of Drosophila metamorphosis by xenobiotic response regulators. PLoS Genet 9: e1003263. PubMed ID: 23408904

    Wang, C., Cai, W., Li, Y., Deng, H., Bao, X., Girton, J., Johansen, J. and Johansen, K. M. (2011). The epigenetic H3S10 phosphorylation mark is required for counteracting heterochromatic spreading and gene silencing in Drosophila melanogaster. J Cell Sci 124: 4309-4317. PubMed ID: 22247192

    Deng, H., Bell, J. B. and Simmonds, A. J. (2010). Vestigial is required during late-stage muscle differentiation in Drosophila melanogaster embryos. Mol Biol Cell 21: 3304-3316. PubMed ID: 20685961

  • Wu-Min Deng Department of Biological Science at The Florida State University. Tallahassee
    Sun, J., Liu, W. K., Ellsworth, C., Sun, Q., Pan, Y. F., Huang, Y. C. and Deng, W. M. (2023). Integrating lipid metabolism, pheromone production and perception by Fruitless and Hepatocyte nuclear factor 4. bioRxiv. PubMed ID: 36865119

    Chatterjee, D., Cong, F., Wang, X. F., Machado Costa, C. A., Huang, Y. C. and Deng, W. M. (2023). Cell polarity opposes Jak/STAT-mediated Escargot activation that drives intratumor heterogeneity in a Drosophila tumor model. Cell Rep 42(2): 112061. PubMed ID: 36709425

    Chatterjee, D., Costa, C. A. M., Wang, X. F., Jevitt, A., Huang, Y. C. and Deng, W. M. (2022). Single-cell transcriptomics identifies Keap1-Nrf2 regulated collective invasion in a Drosophila tumor model. Elife 11. PubMed ID: 36321803

    Row, S., Huang, Y. C. and Deng, W. M. (2021). Developmental regulation of oocyte lipid intake through 'patent' follicular epithelium in Drosophila melanogaster. iScience 24(4): 102275. PubMed ID: 33817579

    Jevitt, A., Chatterjee, D., Xie, G., Wang, X. F., Otwell, T., Huang, Y. C. and Deng, W. M. (2020). A single-cell atlas of adult Drosophila ovary identifies transcriptional programs and somatic cell lineage regulating oogenesis. PLoS Biol 18(4): e3000538. PubMed ID: 32339165

    Lo, P. K., Huang, Y. C., Corcoran, D., Jiao, R. and Deng, W. M. (2019). Drosophila chromatin assembly factor 1 p105 and p180 subunits are required for follicle cell proliferation via inhibiting Notch signaling. J Cell Sci. PubMed ID: 30630896

    Shu, Z., Huang, Y. C., Palmer, W. H., Tamori, Y., Xie, G., Wang, H., Liu, N. and Deng, W. M. (2017). Systematic analysis reveals tumor-enhancing and -suppressing microRNAs in Drosophila epithelial tumors. Oncotarget 8(65): 108825-108839. PubMed ID: 29312571

    Xie, G., Chen, H., Jia, D., Shu, Z., Palmer, W.H., Huang, Y.C., Zeng, X., Hou, S.X., Jiao, R.2 and Deng, W.M. (2016). The SWI/SNF complex protein Snr1 is a tumor suppressor in Drosophila imaginal tissues. Cancer Res [Epub ahead of print]. PubMed ID: 27923836

    Tamori, Y., Suzuki, E. and Deng, W. M. (2016). Epithelial tumors originate in tumor hotspots, a tissue-intrinsic microenvironment. PLoS Biol 14: e1002537. PubMed ID: 27584724

    Jia, D., Bryant, J., Jevitt, A., Calvin, G. and Deng, W. M. (2016). The ecdysone and Notch pathways synergistically regulate Cut at the dorsal-ventral boundary in Drosophila wing discs. J Genet Genomics [Epub ahead of print]. PubMed ID: 27117286

    Lo, P. K., Huang, Y. C., Poulton, J. S., Leake, N., Palmer, W. H., Vera, D., Xie, G., Klusza, S. and Deng, W. M. (2016). RNA helicase Belle/DDX3 regulates transgene expression in Drosophila. Dev Biol [Epub ahead of print]. PubMed ID: 26900887

  • Barry Denholm Centre for Integrative Physiology, The University of Edinburgh
    Koehler, S., Huber, T. B. and Denholm, B. (2022). A protective role for Drosophila Filamin in nephrocytes via Yorkie mediated hypertrophy. Life Sci Alliance 5(12). PubMed ID: 35922155

    Beaven, R. and Denholm, B. (2018). Release and spread of Wingless is required to pattern the proximo-distal axis of Drosophila renal tubules. Elife 7. PubMed ID: 30095068

    Denholm, B. (2013) Shaping up for action: The path to physiological maturation in the renal tubules of Drosophila. Organogenesis 9. PubMed ID: 23445869

    Denholm, B., Hu, N., Fauquier, T., Caubit, X., Fasano, L. and Skaer, H. (2013). The tiptop/teashirt genes regulate cell differentiation and renal physiology in Drosophila. Development 140: 1100-1110. PubMed ID: 23404107

    Bunt, S., Denholm, B. and Skaer, H. (2011). Characterisation of the Drosophila procollagen lysyl hydroxylase, dPlod. Gene Expr Patterns 11: 72-78. PubMed ID: 20888931

  • Angela DePace Department of Systems Biology, Harvard Medical School, Boston
    Harden, T. T., Vincent, B. J. and DePace, A. H. (2023). Transcriptional activators in the early Drosophila embryo perform different kinetic roles. Cell Syst 14(4): 258-272.e254. PubMed ID: 37080162

    Lopez-Rivera, F., Foster Rhoades, O. K., Vincent, B. J., Pym, E. C. G., Bragdon, M. D. J., Estrada, J., DePace, A. H. and Wunderlich, Z. (2020). A Mutation in the Drosophila melanogaster eve Stripe 2 Minimal Enhancer Is Buffered by Flanking Sequences. G3 (Bethesda). PubMed ID: 33037064

    Scholes, C., Biette, K. M., Harden, T. T. and DePace, A. H. (2019). Signal integration by shadow enhancers and enhancer duplications varies across the Drosophila embryo. Cell Rep 26(9): 2407-2418.e2405. PubMed ID: 30811990

    Vincent, B. J., Staller, M. V., Lopez-Rivera, F., Bragdon, M. D. J., Pym, E. C. G., Biette, K. M., Wunderlich, Z., Harden, T. T., Estrada, J. and DePace, A. H. (2018). Hunchback is counter-repressed to regulate even-skipped stripe 2 expression in Drosophila embryos. PLoS Genet 14(9): e1007644. PubMed ID: 30192762

    Vincent, B. J., Staller, M. V., Lopez-Rivera, F., Bragdon, M. D. J., Pym, E. C. G., Biette, K. M., Wunderlich, Z., Harden, T. T., Estrada, J. and DePace, A. H. (2018). Hunchback is counter-repressed to regulate even-skipped stripe 2 expression in Drosophila embryos. PLoS Genet 14(9): e1007644. PubMed ID: 30192762

    Samee, M. A. H., Lydiard-Martin, T., Biette, K. M., Vincent, B. J., Bragdon, M. D., Eckenrode, K. B., Wunderlich, Z., Estrada, J., Sinha, S. and DePace, A. H. (2017). Quantitative measurement and thermodynamic modeling of fused enhancers support a two-tiered mechanism for interpreting regulatory DNA. Cell Rep 21(1): 236-245. PubMed ID: 28978476

    Wunderlich, Z., Bragdon, M.D., Vincent, B.J., White, J.A., Estrada, J. and DePace, A.H. (2015). Krüppel expression levels are maintained through compensatory evolution of shadow enhancers. Cell Rep [Epub ahead of print]. PubMed ID: 26344774

    Staller, M. V., Fowlkes, C. C., Bragdon, M. D., Wunderlich, Z., Estrada, J. and DePace, A. H. (2015). A gene expression atlas of a bicoid-depleted Drosophila embryo reveals early canalization of cell fate. Development 142: 587-596. PubMed ID: 25605785

    Staller, M. V., Vincent, B. J., Bragdon, M. D., Lydiard-Martin, T., Wunderlich, Z., Estrada, J. and DePace, A. H. (2015). Shadow enhancers enable Hunchback bifunctionality in the Drosophila embryo. Proc Natl Acad Sci U S A. PubMed ID: 25564665

  • Bart Deplancke the Laboratory of Systems Biology and Genetics, Swiss Institute of Bioinformatics, Lausanne
    Litovchenko, M., Meireles-Filho, A. C. A., Frochaux, M. V., Bevers, R. P. J., Prunotto, A., Anduaga, A. M., Hollis, B., Gardeux, V., Braman, V. S., Russeil, J. M. C., Kadener, S., Dal Peraro, M. and Deplancke, B. (2021). Extensive tissue-specific expression variation and novel regulators underlying circadian behavior. Sci Adv 7(5). PubMed ID: 33514540

    Mouchiroud, L., Sorrentino, V., Williams, E. G., Cornaglia, M., Frochaux, M. V., Lin, T., Nicolet-Dit-Felix, A. A., Krishnamani, G., Ouhmad, T., Gijs, M. A., Deplancke, B. and Auwerx, J. (2016). The Movement Tracker: A Flexible System for Automated Movement Analysis in Invertebrate Model Organisms. Curr Protoc Neurosci 77: 8 37 31-38 37 21. PubMed ID: 27696358

    Bou Sleiman, M. S., Osman, D., Massouras, A., Hoffmann, A. A., Lemaitre, B. and Deplancke, B. (2015). Genetic, molecular and physiological basis of variation in Drosophila gut immunocompetence. Nat Commun 6: 7829. PubMed ID: 26213329

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

  • Stefano De Renzis Developmental Biology, EMBL Heidelberg
    Gustafson, H. J., Claussen, N., De Renzis, S. and Streichan, S. J. (2022). Patterned mechanical feedback establishes a global myosin gradient. Nat Commun 13(1): 7050. PubMed ID: 36396633

    Akhmanova, M., Emtenani, S., Krueger, D., Gyoergy, A., Guarda, M., Vlasov, M., Vlasov, F., Akopian, A., Ratheesh, A., De Renzis, S. and Siekhaus, D. E. (2022). Cell division in tissues enables macrophage infiltration. Science 376(6591): 394-396. PubMed ID: 35446632

    Viswanathan, R., Hartmann, J., Pallares Cartes, C. and De Renzis, S. (2021). Desensitisation of Notch signalling through dynamic adaptation in the nucleus. Embo j: e107245. PubMed ID: 34396565

    Krueger, D., Pallares Cartes, C., Makaske, T. and De Renzis, S. (2020). betaH-spectrin is required for ratcheting apical pulsatile constrictions during tissue invagination. EMBO Rep: e49858. PubMed ID: 32588528

    Viswanathan, R., Necakov, A., Trylinski, M., Harish, R. K., Krueger, D., Esposito, E., Schweisguth, F., Neveu, P. and De Renzis, S. (2019). Optogenetic inhibition of Delta reveals digital Notch signalling output during tissue differentiation. EMBO Rep: e47999. PubMed ID: 31668010

    Krueger, D., Quinkler, T., Mortensen, S. A., Sachse, C. and De Renzis, S. (2019). Cross-linker-mediated regulation of actin network organization controls tissue morphogenesis. J Cell Biol. PubMed ID: 31253650

    Deneke, V. E., Puliafito, A., Krueger, D., Narla, A. V., De Simone, A., Primo, L., Vergassola, M., De Renzis, S. and Di Talia, S. (2019). Self-Organized Nuclear Positioning Synchronizes the Cell Cycle in Drosophila Embryos. Cell. PubMed ID: 30982601

    Krueger, D., Tardivo, P., Nguyen, C. and De Renzis, S. (2018). Downregulation of basal myosin-II is required for cell shape changes and tissue invagination. EMBO J 37(23). PubMed ID: 30442834

    Izquierdo, E., Quinkler, T. and De Renzis, S. (2018). Guided morphogenesis through optogenetic activation of Rho signalling during early Drosophila embryogenesis. Nat Commun 9(1): 2366. PubMed ID: 29915285

    Guglielmi, G. and De Renzis, S. (2017). Optogenetic inhibition of apical constriction during Drosophila embryonic development. Methods Cell Biol 139: 167-186. PubMed ID: 28215335

  • Girish Deshpande Princeton University, Princeton, NJ
    Colonnetta, M. M., Schedl, P. and Deshpande, G. (2023). Germline/soma distinction in Drosophila embryos requires regulators of zygotic genome activation. Elife 12. PubMed ID: 36598809

    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

    Colonnetta, M. M., Goyal, Y., Johnson, H. E., Syal, S., Schedl, P. and Deshpande, G. (2022). Preformation and epigenesis converge to specify primordial germ cell fate in the early Drosophila embryo. PLoS Genet 18(1): e1010002. PubMed ID: 34986144

    Colonnetta, M. M., Abrahante, J. E., Schedl, P., Gohl, D. M. and Deshpande, G. (2021). CLAMP regulates zygotic genome activation in Drosophila embryos. Genetics 219(2). PubMed ID: 34849887

    Sahu, A., Karmakar, S., Halder, S., Ghosh, G., Acharjee, S., Dasgupta, P., Ghosh, R., Deshpande, G. and Prasad, M. (2021). Germline soma communication mediated by gap junction proteins regulates epithelial morphogenesis. PLoS Genet 17(8): e1009685. PubMed ID: 34343194

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

    Syal, S., Ng, C., Kim, Y., Janbieh, J., Govind, S. and Deshpande, G. (2020). Reactive oxygen species signaling in primordial germ cell development in Drosophila embryos. Genesis: e23362. PubMed ID: 32302036

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

    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

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

    Shukla, V., Dhiman, N., Nayak, P., Dahanukar, N., Deshpande, G. and Ratnaparkhi, G. S. (2018). Stonewall and Brickwall: Two Partially Redundant Determinants Required for the Maintenance of Female Germline in Drosophila. G3 (Bethesda) 8(6): 2027-2041. PubMed ID: 29669801

  • Claude Desplan Department of Biology, New York University
    Ferreira, A. A. G. and Desplan, C. (2023). An Atlas of the Developing Drosophila Visual System Glia and Subcellular mRNA Localization of Transcripts in Single Cells. bioRxiv. PubMed ID: 37609218

    Chen, Y. D., Chen, Y. C., Rajesh, R., Shoji, N., Jacy, M., Lacin, H., Erclik, T. and Desplan, C. (2023). Using single-cell RNA sequencing to generate predictive cell-type-specific split-GAL4 reagents throughout development. Proc Natl Acad Sci U S A 120(32): e2307451120. PubMed ID: 37523539

    Ozel, M. N., Gibbs, C. S., Holguera, I., Soliman, M., Bonneau, R. and Desplan, C. (2022). Coordinated control of neuronal differentiation and wiring by sustained transcription factors. Science 378(6626): eadd1884. PubMed ID: 36480601

    Konstantinides, N., Holguera, I., Rossi, A. M., Escobar, A., Dudragne, L., Chen, Y. C., Tran, T. N., MartInez Jaimes, A. M., Ozel, M. N., Simon, F., Shao, Z., Tsankova, N. M., Fullard, J. F., Walldorf, U., Roussos, P. and Desplan, C. (2022). A complete temporal transcription factor series in the fly visual system. Nature 604(7905): 316-322. PubMed ID: 35388222

    Ozel, M. N., Simon, F., Jafari, S., Holguera, I., Chen, Y. C., Benhra, N., El-Danaf, R. N., Kapuralin, K., Malin, J. A., Konstantinides, N. and Desplan, C. (2020). Neuronal diversity and convergence in a visual system developmental atlas. Nature. PubMed ID: 33149298

    Rossi, A. M. and Desplan, C. (2020). Extrinsic activin signaling cooperates with an intrinsic temporal program to increase mushroom body neuronal diversity. Elife 9. PubMed ID: 32628110

    Wei, H., Kyung, H. Y., Kim, P. J. and Desplan, C. (2019). The diversity of lobula plate tangential cells (LPTCs) in the Drosophila motion vision system. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 31709462

    Courgeon, M. and Desplan, C. (2019). Coordination between stochastic and deterministic specification in the Drosophila visual system. Science 366(6463). PubMed ID: 31582524

    Ebadi, H., Perry, M., Short, K., Klemm, K., Desplan, C., Stadler, P. F. and Mehta, A. (2018). Patterning the insect eye: From stochastic to deterministic mechanisms. PLoS Comput Biol 14(11): e1006363. PubMed ID: 30439954

    Barnhart, E. L., Wang, I. E., Wei, H., Desplan, C. and Clandinin, T. R. (2018). Sequential nonlinear filtering of local motion cues by global motion circuits. Neuron 100(1): 229-243.e223. PubMed ID: 30220510

    Konstantinides, N., Kapuralin, K., Fadil, C., Barboza, L., Satija, R. and Desplan, C. (2018). Phenotypic convergence: Distinct transcription factors regulate common terminal features. Cell. PubMed ID: 29909983

  • Justin DiAngelo Biochemistry and Molecular Biology, Penn State Berks
    Mercier, J., Nagengast, A. A. and DiAngelo, J. R. (2023). The role of SR protein kinases in regulating lipid storage in the Drosophila fat body. Biochem Biophys Res Commun 649: 10-15. PubMed ID: 36738578

    Fruin, A. M., Leon, K. E. and DiAngelo, J. R. (2022). The ESCRT-III Protein Chmp1 Regulates Lipid Storage in the Drosophila Fat Body. Med Sci (Basel) 11(1). PubMed ID: 36649042

    Weidman, T., Nagengast, A. A. and DiAngelo, J. R. (2022). The splicing factor 9G8 regulates the expression of NADPH-producing enzyme genes in Drosophila. Biochem Biophys Res Commun 620: 92-97. PubMed ID: 35780586

    Shabar, H. and DiAngelo, J. R. (2022). The regulation of lipid and carbohydrate storage by the splicing factor gene snRNP-U1-70K in the Drosophila fat body. MicroPubl Biol 2022. PubMed ID: 35655607

    Nagle, C., Bhogal, J. K., Nagengast, A. A. and DiAngelo, J. R. (2022). Transportin-serine/arginine-rich (Tnpo-SR) proteins are necessary for proper lipid storage in the Drosophila fat body. Biochem Biophys Res Commun 596: 1-5. PubMed ID: 35104661

    Palermo, J., Keene, A. C. and DiAngelo, J. R. (2022). Expression of a constitutively active insulin receptor in Drosulfakinin (Dsk) neurons regulates metabolism and sleep in Drosophila. Biochem Biophys Rep 30: 101280. PubMed ID: 35600902

    Paul, M. M., Dannhauser, S., Morris, L., Mrestani, A., Hubsch, M., Gehring, J., Hatzopoulos, G. N., Pauli, M., Auger, G. M., Bornschein, G., Scholz, N., Ljaschenko, D., Müller, M., Sauer, M., Schmidt, H., Kittel, R. J., DiAntonio, A., Vakonakis, I., Heckmann, M. and Langenhan, T. (2022). The human cognition-enhancing CORD7 mutation increases active zone number and synaptic release. Brain. PubMed ID: 35022694

    Kolasa, A. M., Bhogal, J. K. and DiAngelo, J. R. (2021). The heterogeneous nuclear ribonucleoprotein (hnRNP) glorund functions in the Drosophila fat body to regulate lipid storage and transport. Biochem Biophys Rep 25: 100919. PubMed ID: 33537463

    Leon, K. E., Fruin, A. M., Nowotarski, S. L. and DiAngelo, J. R. (2019). The regulation of triglyceride storage by ornithine decarboxylase (Odc1) in Drosophila. Biochem Biophys Res Commun. PubMed ID: 31870547

    Bennick, R. A., Nagengast, A. A. and DiAngelo, J. R. (2019). The SR proteins SF2 and RBP1 regulate triglyceride storage in the fat body of Drosophila. Biochem Biophys Res Commun 516(3): 928-933. PubMed ID: 31277943

  • Aaron DiAntonio Washington University in St. Louis
    Lones, L. and DiAntonio, A. (2023). SIK3 and Wnk converge on Fray to regulate glial K+ buffering and seizure susceptibility. PLoS Genet 19(1): e1010581. PubMed ID: 36626385

    Brace, E. J., Essuman, K., Mao, X., Palucki, J., Sasaki, Y., Milbrandt, J. and DiAntonio, A. (2022). Distinct developmental and degenerative functions of SARM1 require NAD+ hydrolase activity. PLoS Genet 18(6): e1010246. PubMed ID: 35737728

    Hayne, M. and DiAntonio, A. (2022). Protein phosphatase 2A restrains DLK signaling to promote proper Drosophila synaptic development and mammalian cortical neuron survival. Neurobiol Dis 163: 105586. PubMed ID: 34923110

    Li, H., Lones, L. and DiAntonio, A. (2021). Bidirectional regulation of glial potassium buffering - glioprotection versus neuroprotection. Elife 10. PubMed ID: 33646119

    Li, H., Russo, A. and DiAntonio, A. (2019). SIK3 suppresses neuronal hyperexcitability by regulating the glial capacity to buffer K(+) and water. J Cell Biol. PubMed ID: 31645458

    Russo, A. and DiAntonio, A. (2019). Wnd/DLK is a critical target of FMRP responsible for neurodevelopmental and behavior defects in the Drosophila model of Fragile X syndrome. Cell Rep 28(10): 2581-2593.e2585. PubMed ID: 31484070

    Russo, A., Goel, P., Brace, E. J., Buser, C., Dickman, D. and DiAntonio, A. (2019). The E3 ligase Highwire promotes synaptic transmission by targeting the NAD-synthesizing enzyme dNmnat. EMBO Rep. PubMed ID: 30692130

    Karney-Grobe, S., Russo, A., Frey, E., Milbrandt, J. and DiAntonio, A. (2018). HSP90 is a chaperone for DLK and is required for axon injury signaling. Proc Natl Acad Sci U S A. PubMed ID: 30275300

    Walker, L. J., Summers, D. W., Sasaki, Y., Brace, E. J., Milbrandt, J. and DiAntonio, A. (2017). MAPK signaling promotes axonal degeneration by speeding the turnover of the axonal maintenance factor NMNAT2. Elife 6. PubMed ID: 28095293

    Hao, Y., Frey, E., Yoon, C., Wong, H., Nestorovski, D., Holzman, L. B., Giger, R. J., DiAntonio, A. and Collins, C. (2016). An evolutionarily conserved mechanism for cAMP elicited axonal regeneration involves direct activation of the dual leucine zipper kinase DLK. Elife 5 [Epub ahead of print]. PubMed ID: 27268300

    Bae, H., Chen, S., Roche, J.P., Ai, M., Wu, C., Diantonio, A. and Graf, E.R. (2016). Rab3-GEF controls active zone development at the Drosophila neuromuscular junction. eNeuro 3(2). PubMed ID: 27022630

    Valakh, V., Frey, E., Babetto, E., Walker, L. J. and DiAntonio, A. (2015). Cytoskeletal disruption activates the DLK/JNK pathway, which promotes axonal regeneration and mimics a preconditioning injury. Neurobiol Dis 77: 13-25. PubMed ID: 25726747

  • Fernando Díaz-Benjumea CBM Severo Ochoa, Madrid
    Diaz-de-la-Pena, L., Maestro-Paramio, L., Diaz-Benjumea, F. J. and Herrero, P. (2020). Temporal groups of lineage-related neurons have different neuropeptidergic fates and related functions in the Drosophila melanogaster CNS. Cell Tissue Res. PubMed ID: 32556724

    Blom-Dahl, D., Cordoba, S., Gabilondo, H., Carr-Baena, P., Diaz-Benjumea, F. J. and Estella, C. (2020). In vivo analysis of the evolutionary conserved BTD-box domain of Sp1 and Btd during Drosophila development. Dev Biol. PubMed ID: 32738261

    Alvarez, J. A. and Diaz-Benjumea, F. J. (2018). Origin and specification of type II neuroblasts in the Drosophila embryo. Development 145(7). PubMed ID: 29567672

    Alvarez-Rivero, J., Moris-Sanz, M., Estacio-Gomez, A., Montoliu-Nerin, M., Diaz-Benjumea, F. J. and Herrero, P. (2016). Variability in the number of abdominal leucokinergic neurons in adult Drosophila melanogaster. J Comp Neurol [Epub ahead of print]. PubMed ID: 27506156

    Moris-Sanz, M., Estacio-Gómez, A., Sánchez-Herrero, E and Díaz-Benjumea, F.J. (2015). The study of the Bithorax-complex genes in patterning CCAP neurons reveals a temporal control of neuronal differentiation by Abd-B. Biol Open [Epub ahead of print]. PubMed ID: 26276099

    Estacio-Gomez, A. and Diaz-Benjumea, F. J. (2013). Roles of Hox genes in the patterning of the central nervous system of Drosophila. Fly (Austin) 8. PubMed ID: 24406332

    Herrero, P., Estacio-Gomez, A., Moris-Sanz, M., Alvarez-Rivero, J. and Diaz-Benjumea, F. J. (2013). ORIGIN and specification of the brain leucokinergic neurons of Drosophila: Similarities to and differences from abdominal leucokinergic neurons. Dev Dyn. PubMed ID: 24155257

    Perea, D., Molohon, K., Edwards, K. and Diaz-Benjumea, F. J. (2013). Multiple roles of the gene zinc finger homeodomain-2 in the development of the Drosophila wing. Mech Dev. PubMed ID: 23811114

    Benito-Sipos, J., Estacio-Gomez, A., Moris-Sanz, M., Baumgardt, M., Thor, S. and Diaz-Benjumea, F. J. (2010). A genetic cascade involving klumpfuss, nab and castor specifies the abdominal leucokinergic neurons in the Drosophila CNS. Development 137: 3327-3336. PubMed ID: 20823069

    Losada-Perez, M., Gabilondo, H., del Saz, D., Baumgardt, M., Molina, I., Leon, Y., Monedero, I., Diaz-Benjumea, F., Torroja, L. and Benito-Sipos, J. (2010). Lineage-unrelated neurons generated in different temporal windows and expressing different combinatorial codes can converge in the activation of the same terminal differentiation gene. Mech Dev 127: 458-471. PubMed ID: 20732418

  • Michael Dickinson California Institute of Technology
    Namiki, S., Ros, I. G., Morrow, C., Rowell, W. J., Card, G. M., Korff, W. and Dickinson, M. H. (2022). A population of descending neurons that regulates the flight motor of Drosophila. Curr Biol 32(5): 1189-1196.e1186. PubMed ID: 35090590

    Behbahani, A. H., Palmer, E. H., Corfas, R. A. and Dickinson, M. H. (2021). Drosophila re-zero their path integrator at the center of a fictive food patch. Curr Biol. PubMed ID: 34450090

    Leitch, K. J., Ponce, F. V., Dickson, W. B., van Breugel, F. and Dickinson, M. H. (2021). The long-distance flight behavior of Drosophila supports an agent-based model for wind-assisted dispersal in insects. Proc Natl Acad Sci U S A 118(17). PubMed ID: 33879607

    Agrawal, S. and Dickinson, M. H. (2019). The effects of target contrast on Drosophila courtship. J Exp Biol. PubMed ID: 31315932

    Corfas, R. A., Sharma, T. and Dickinson, M. H. (2019). Diverse food-sensing neurons trigger idiothetic local search in Drosophila. Curr Biol 29(10): 1660-1668. PubMed ID: 31056390

    Warren, T. L., Giraldo, Y. M. and Dickinson, M. H. (2019). Celestial navigation in Drosophila. J Exp Biol 222(Pt Suppl 1). PubMed ID: 30728228

    van Breugel, F., Huda, A. and Dickinson, M. H. (2018). Distinct activity-gated pathways mediate attraction and aversion to CO2 in Drosophila. Nature. PubMed ID: 30464346

    Chen, C. L., Hermans, L., Viswanathan, M. C., Fortun, D., Aymanns, F., Unser, M., Cammarato, A., Dickinson, M. H. and Ramdya, P. (2018). Imaging neural activity in the ventral nerve cord of behaving adult Drosophila. Nat Commun 9(1): 4390. PubMed ID: 30348941

    Giraldo, Y. M., Leitch, K. J., Ros, I. G., Warren, T. L., Weir, P. T. and Dickinson, M. H. (2018). Sun navigation requires compass neurons in Drosophila. Curr Biol. PubMed ID: 30174187

    Warren, T. L., Weir, P. T. and Dickinson, M. H. (2018). Flying Drosophila maintain arbitrary but stable headings relative to the angle of polarized light. J Exp Biol. PubMed ID: 29593084

    Kim, I. S. and Dickinson, M. H. (2017). Idiothetic path Integration in the fruit fly Drosophila melanogaster. Curr Biol 27(15): 2227-2238.e2223. PubMed ID: 28736164

    Schnell, B., Ros, I. G. and Dickinson, M. H. (2017). A Descending Neuron Correlated with the Rapid Steering Maneuvers of Flying Drosophila. Curr Biol 27(8): 1200-1205. PubMed ID: 28392112

  • Dion Dickman USC Neuroscience, University of Southern California
    Han, Y., Goel, P., Chen, J., Perry, S., Tran, N., Nishimura, S., Sanjani, M., Chien, C. and Dickman, D. (2023). Excess glutamate release triggers subunit-specific homeostatic receptor scaling. Cell Rep 42(7): 112775. PubMed ID: 37436892

    He, K., Han, Y., Li, X., Hernandez, R. X., Riboul, D. V., Feghhi, T., Justs, K. A., Mahneva, O., Perry, S., Macleod, G. T. and Dickman, D. (2023). Physiologic and Nanoscale Distinctions Define Glutamatergic Synapses in Tonic vs Phasic Neurons. J Neurosci 43(25): 4598-4611. PubMed ID: 37221096

    Perry, S., Han, Y., Qiu, C., Chien, C., Goel, P., Nishimura, S., Sajnani, M., Schmid, A., Sigrist, S. J. and Dickman, D. (2022). A glutamate receptor C-tail recruits CaMKII to suppress retrograde homeostatic signaling. Nat Commun 13(1): 7656. PubMed ID: 36496500

    Muttathukunnel, P., Frei, P., Perry, S., Dickman, D. and Muller, M. (2022). Rapid homeostatic modulation of transsynaptic nanocolumn rings. Proc Natl Acad Sci U S A 119(45): e2119044119. PubMed ID: 36322725

    Chen, X. and Dickman, D. (2021). Tissue-Specific Ribosome Profiling in Drosophila. Methods Mol Biol 2252: 175-188. PubMed ID: 33765275

    Ramesh, N., Escher, M. J. F., Mampell, M. M., Bohme, M. A., Gotz, T. W. B., Goel, P., Matkovic, T., Petzoldt, A. G., Dickman, D. and Sigrist, S. J. (2021). Antagonistic interactions between two Neuroligins coordinate pre- and postsynaptic assembly. Curr Biol. PubMed ID: 33651992

    Goel, P., Nishimura, S., Chetlapalli, K., Li, X., Chen, C. and Dickman, D. (2020). Distinct Target-Specific Mechanisms Homeostatically Stabilize Transmission at Pre- and Post-synaptic Compartments. Front Cell Neurosci 14: 196. PubMed ID: 32676010

    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

    Kikuma, K., Li, X., Perry, S., Li, Q., Goel, P., Chen, C., Kim, D., Stavropoulos, N. and Dickman, D. (2019). Cul3 and insomniac are required for rapid ubiquitination of postsynaptic targets and retrograde homeostatic signaling. Nat Commun 10(1): 2998. PubMed ID: 31278365

    Bohme, M. A., McCarthy, A. W., Grasskamp, A. T., Beuschel, C. B., Goel, P., Jusyte, M., Laber, D., Huang, S., Rey, U., Petzoldt, A. G., Lehmann, M., Gottfert, F., Haghighi, P., Hell, S. W., Owald, D., Dickman, D., Sigrist, S. J. and Walter, A. M. (2019). Rapid active zone remodeling consolidates presynaptic potentiation. Nat Commun 10(1): 1085. PubMed ID: 30842428

    Russo, A., Goel, P., Brace, E. J., Buser, C., Dickman, D. and DiAntonio, A. (2019). The E3 ligase Highwire promotes synaptic transmission by targeting the NAD-synthesizing enzyme dNmnat. EMBO Rep. PubMed ID: 30692130

  • Barry Dickson University of Queensland, Australia
    Ehrhardt, E., Whitehead, S. C., Namiki, S., Minegishi, R., Siwanowicz, I., Feng, K., Otsuna, H., Meissner, G. W., Stern, D., Truman, J., Shepherd, D., Dickinson, M. H., Ito, K., Dickson, B. J., Cohen, I., Card, G. M. and Korff, W. (2023). Single-cell type analysis of wing premotor circuits in the ventral nerve cord of Drosophila melanogaster. bioRxiv. PubMed ID: 37398009

    Chen, C. L., Aymanns, F., Minegishi, R., Matsuda, V. D. V., Talabot, N., Gunel, S., Dickson, B. J. and Ramdya, P. (2023). Ascending neurons convey behavioral state to integrative sensory and action selection brain regions. Nat Neurosci 26(4): 682-695. PubMed ID: 36959417

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

    Sterne, G. R., Otsuna, H., Dickson, B. J. and Scott, K. (2021). Classification and genetic targeting of cell types in the primary taste and premotor center of the adult Drosophila brain. Elife 10. PubMed ID: 34473057

    Wang, K., Wang, F., Forknall, N., Yang, T., Patrick, C., Parekh, R. and Dickson, B. J. (2020). Neural circuit mechanisms of sexual receptivity in Drosophila females. Nature. PubMed ID: 33239786

    Feng, K., Sen, R., Minegishi, R., Dubbert, M., Bockemuhl, T., Buschges, A. and Dickson, B. J. (2020). Distributed control of motor circuits for backward walking in Drosophila. Nat Commun 11(1): 6166. PubMed ID: 33268800

    Wang, F., Wang, K., Forknall, N., Parekh, R. and Dickson, B. J. (2020). Circuit and Behavioral Mechanisms of Sexual Rejection by Drosophila Females. Curr Biol. PubMed ID: 32795445

    McKellar, C. E., Siwanowicz, I., Dickson, B. J. and Simpson, J. H. (2020). Controlling motor neurons of every muscle for fly proboscis reaching. Elife 9. PubMed ID: 32584254

    Wang, F., Wang, K., Forknall, N., Patrick, C., Yang, T., Parekh, R., Bock, D. and Dickson, B. J. (2020). Neural circuitry linking mating and egg laying in Drosophila females. Nature 579(7797): 101-105. PubMed ID: 32103180

    Sen, R., Wang, K. and Dickson, B. J. (2019). TwoLumps Ascending Neurons Mediate Touch-Evoked Reversal of Walking Direction in Drosophila. Curr Biol 29(24): 4337-4344.e4335. PubMed ID: 31813606

    Ding, Y., Lillvis, J. L., Cande, J., Berman, G. J., Arthur, B. J., Long, X., Xu, M., Dickson, B. J. and Stern, D. L. (2019). Neural evolution of context-dependent fly song. Curr Biol. PubMed ID: 30880014

    McKellar, C. E., Lillvis, J. L., Bath, D. E., Fitzgerald, J. E., Cannon, J. G., Simpson, J. H. and Dickson, B. J. (2019). Threshold-based ordering of sequential actions during Drosophila courtship. Curr Biol 29(3): 426-434.e426. PubMed ID: 30661796

    Ribeiro, I. M. A., Drews, M., Bahl, A., Machacek, C., Borst, A. and Dickson, B. J. (2018). Visual projection neurons mediating directed courtship in Drosophila. Cell 174(3): 607-621.e618. PubMed ID: 30033367

  • Herman Dierick Molecular and Human Genetics, Baylor College of Medicine
    Chowdhury, B., Wang, M., Gnerer, J. P. and Dierick, H. A. (2021). The Divider Assay is a high-throughput pipeline for aggression analysis in Drosophila. Commun Biol 4(1): 85. PubMed ID: 33469118

    Davis, S. M., Thomas, A. L., Liu, L., Campbell, I. M. and Dierick, H. A. (2017). Isolation of aggressive behavior mutants in Drosophila using a screen for wing damage. Genetics. PubMed ID: 29109180

    Davis, S. M., Thomas, A. L., Nomie, K. J., Huang, L. and Dierick, H. A. (2014). Tailless and Atrophin control Drosophila aggression by regulating neuropeptide signalling in the pars intercerebralis. Nat Commun 5: 3177. PubMed ID: 24495972

    Gendron, C. M., Kuo, T. H., Harvanek, Z. M., Chung, B. Y., Yew, J. Y., Dierick, H. A. and Pletcher, S. D. (2014). Drosophila life span and physiology are modulated by sexual perception and reward. Science 343: 544-548. PubMed ID: 24292624

    Fedina, T. Y., Kuo, T. H., Dreisewerd, K., Dierick, H. A., Yew, J. Y. and Pletcher, S. D. (2012). Dietary effects on cuticular hydrocarbons and sexual attractiveness in Drosophila. PLoS One 7: e49799. PubMed ID: 23227150

  • Patrick DiMario Department of Biological Sciences, Louisiana State University, Baton Rouge
    Baral, S. S., Lieux, M. E. and DiMario, P. J. (2020). Nucleolar stress in Drosophila neuroblasts, a model for human ribosomopathies. Biol Open. PubMed ID: 32184230

    Baral, S. S. and DiMario, P. J. (2019). The Nopp140 gene in Drosophila melanogaster displays length polymorphisms in its large repetitive second exon. Mol Genet Genomics. PubMed ID: 31006039

    Wang, Y. and DiMario, P. (2016). Loss of Drosophila nucleostemin 2 (NS2) blocks nucleolar release of the 60S subunit leading to ribosome stress. Chromosoma [Epub ahead of print]. PubMed ID: 27150106

    He, F., James, A., Raje, H., Ghaffari, H. and DiMario, P. (2014). Deletion of Drosophila Nopp140 induces subcellular ribosomopathies. Chromosoma. PubMed ID: 25384888

    James, A., Cindass, R., Jr., Mayer, D., Terhoeve, S., Mumphrey, C., Dimario, P. (2013) Nucleolar stress in Drosophila melanogaster: RNAi-mediated depletion of Nopp140. Nucleus 4. PubMed ID: 23412656

    He, F. and DiMario, P. J. (2011). Drosophila delta-1-pyrroline-5-carboxylate dehydrogenase (P5CDh) is required for proline breakdown and mitochondrial integrity-Establishing a fly model for human type II hyperprolinemia. Mitochondrion 11: 397-404. PubMed ID: 21168532

    Rosby, R., Cui, Z., Rogers, E., deLivron, M. A., Robinson, V. L. and DiMario, P. J. (2009). Knockdown of the Drosophila GTPase nucleostemin 1 impairs large ribosomal subunit biogenesis, cell growth, and midgut precursor cell maintenance. Mol Biol Cell 20: 4424-4434. PubMed ID: 19710426

  • Patrizio Dimitri Dottorato in Genetica e Biologia Molecolare, University of Rome
    Messina, G., Celauro, E., Marsano, R. M., Prozzillo, Y. and Dimitri, P. (2022). Epigenetic Silencing of P-Element Reporter Genes Induced by Transcriptionally Active Domains of Constitutive Heterochromatin in Drosophila melanogaster. Genes (Basel) 14(1). PubMed ID: 36672753

    Messina, G., Prozzillo, Y., Monache, F. D., Santopietro, M. V. and Dimitri, P. (2022). Evolutionary conserved relocation of chromatin remodeling complexes to the mitotic apparatus. BMC Biol 20(1): 172. PubMed ID: 35922843

    Prozzillo, Y., Cuticone, S., Ferreri, D., Fattorini, G., Messina, G. and Dimitri, P. (2021). In Vivo Silencing of Genes Coding for dTip60 Chromatin Remodeling Complex Subunits Affects Polytene Chromosome Organization and Proper Development in Drosophila melanogaster. Int J Mol Sci 22(9). PubMed ID: 33926075

    Dimitri, P.Caizzi, R., Moschetti, R., Piacentini, L., Fanti, L., Marsano, R. M. and Dimitri, P. (2016). Comparative genomic analyses provide new insights into the evolutionary dynamics of heterochromatin in Drosophila. PLoS Genet 12: e1006212. PubMed ID: 27513559

    Messina, G., Atterrato, M. T., Fanti, L., Giordano, E. and Dimitri, P. (2016). Expression of human Cfdp1 gene in Drosophila reveals new insights into the function of the evolutionarily conserved BCNT protein family. Sci Rep 6: 25511. PubMed ID: 27151176

    Moschetti, R., Celauro, E., Cruciani, F., Caizzi, R. and Dimitri, P. (2014). On the evolution of Yeti, a Drosophila melanogaster heterochromatin gene. PLoS One 9: e113010. PubMed ID: 25405891

    Messina, G., Damia, E., Fanti, L., Atterrato, M. T., Celauro, E., Mariotti, F. R., Accardo, M. C., Walther, M., Verni, F., Picchioni, D., Moschetti, R., Caizzi, R., Piacentini, L., Cenci, G., Giordano, E. and Dimitri, P. (2014). Yeti, a Drosophila melanogaster essential gene, encodes a protein required for chromatin organization. J Cell Sci. PubMed ID: 24652835

    Wang, S. H., Nan, R., Accardo, M. C., Sentmanat, M., Dimitri, P. and Elgin, S. C. (2014). A distinct type of heterochromatin at the telomeric region of the Drosophila melanogaster Y chromosome. PLoS One 9: e86451. PubMed ID: 24475122

    Cipressa, F., Romano, S., Centonze, S., zur Lage, P. I., Verni, F., Dimitri, P., Gatti, M. and Cenci, G. (2013). Effete, a Drosophila chromatin-associated ubiquitin-conjugating enzyme that affects telomeric and heterochromatic position effect variegation. Genetics 195: 147-158. PubMed ID: 23821599

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

  • Dessislava Dimova Dept. Molecular Biology & Biochemistry, Rutgers
    Lee, H., Ragusano, L., Martinez, A., Gill, J. and Dimova, D. K. (2012). A dual role for the dREAM/MMB complex in the regulation of differentiation-specific E2F/RB target genes. Mol. Cell Biol. 32(11): 2110-20. PubMed Citation: 22451490

    Lee, H., Ohno, K., Voskoboynik, Y., Ragusano, L., Martinez, A. and Dimova, D. K. (2010). Drosophila RB proteins repress differentiation-specific genes via two different mechanisms. Mol Cell Biol 30: 2563-2577. PubMed ID: 20176807

  • Steve DiNardo at University of Pennsylvania
    Raz, A. A., Vida, G. S., Stern, S. R., Mahadevaraju, S., Fingerhut, J. M., Viveiros, J. M., Pal, S., Grey, J. R., Grace, M. R., Berry, C. W., Li, H., Janssens, J., Saelens, W., Shao, Z., Hu, C., Yamashita, Y. M., Przytycka, T., Oliver, B., Brill, J. A., Krause, H., Matunis, E. L., White-Cooper, H., DiNardo, S. and Fuller, M. T. (2023). Emergent dynamics of adult stem cell lineages from single nucleus and single cell RNA-Seq of Drosophila testes. Elife 12. PubMed ID: 36795469

    Anllo, L. and DiNardo, S. (2022). Visceral mesoderm signaling regulates assembly position and function of the Drosophila testis niche. Dev Cell 57(8): 1009-1023.e1005. PubMed ID: 35390292

    Nelson, K. A., Warder, B. N., DiNardo, S. and Anllo, L. (2020). Dissection and Live-Imaging of the Late Embryonic Drosophila Gonad. J Vis Exp(164). PubMed ID: 33135688

    Ong, K., Collier, C. and DiNardo, S. (2019). Multiple feedback mechanisms fine-tune Rho signaling to regulate morphogenetic outcomes. J Cell Sci. PubMed ID: 30872456

    Lenhart, K. F., Capozzoli, B., Warrick, G. S. D. and DiNardo, S. (2018). Diminished Jak/STAT signaling causes early-onset aging defects in stem cell cytokinesis. Curr Biol. PubMed ID: 30612906

    Anllo, L., Plasschaert, L. W., Sui, J. and DiNardo, S. (2018). Live imaging reveals hub cell assembly and compaction dynamics during morphogenesis of the Drosophila testis niche. Dev Biol. PubMed ID: 30553808

    Ly, D., Resch, E., Ordiway, G. and DiNardo, S. (2017). Asymmetrically deployed actomyosin-based contractility generates a boundary between developing leg segments in Drosophila. Dev Biol 429(1): 165-176. PubMed ID: 28689737

    Wingert, L. and DiNardo, S. (2015). Traffic jam functions in a branched pathway from Notch activation to niche cell fate. Development 142: 2268-2277. PubMed ID: 26092848

    Lenhart, K.F. and DiNardo, S. (2015). Somatic cell encystment promotes abscission in germline stem cells following a regulated block in cytokinesis. Dev Cell [Epub ahead of print]. PubMed ID: 26143993

    Lawlor, K. T., Ly, D. C. and Dinardo, S. (2013). Drosophila Dachsous and Fat polarize actin-based protrusions over a restricted domain of the embryonic denticle field. Dev Biol. PubMed ID: 24051227

    Zheng, Q., Wang, Y., Vargas, E. and DiNardo, S. (2011). magu is required for germline stem cell self-renewal through BMP signaling in the Drosophila testis. Dev Biol 357: 202-210. PubMed ID: 21723859

    Donoughe, S. and DiNardo, S. (2011). dachsous and frizzled contribute separately to planar polarity in the Drosophila ventral epidermis. Development 138: 2751-2759. PubMed ID: 21613320

  • Andrew Dingwall Department of Microbiology and Immunology, Loyola University, Chicago
    Ford, D. J., Zraly, C. B., Perez, J. H. and Dingwall, A. K. (2020). The Drosophila MLR COMPASS-like complex regulates bantam miRNA expression differentially in the context of cell fate. Dev Biol 468(1-2): 41-53. PubMed ID: 32946789

    Zraly, C. B., Zakkar, A., Perez, J. H., Ng, J., White, K. P., Slattery, M. and Dingwall, A. K. (2020). The Drosophila MLR COMPASS complex is essential for programming cis-regulatory information and maintaining epigenetic memory during development. Nucleic Acids Res. PubMed ID: 32052053

    Chauhan, C., Zraly, C. B., Dingwall, A. K. (2013) The Drosophila COMPASS-like Cmi-Trr coactivator complex regulates dpp/BMP signaling in pattern formation. Dev Biol. PubMed ID: 23707261

    Curtis, B. J., Zraly, C. B. and Dingwall, A. K. (2012). Drosophila LSD1-CoREST demethylase complex regulates DPP/TGFbeta signaling during wing development. Genesis. PubMed ID: 22965777

    Chauhan, C., Zraly, C. B., Parilla, M., Diaz, M. O. and Dingwall, A. K. (2012). Histone recognition and nuclear receptor co-activator functions of Drosophila cara mitad, a homolog of the N-terminal portion of mammalian MLL2 and MLL3. Development 139: 1997-2008. PubMed ID: 22569554

    Zraly, C. B. and Dingwall, A. K. (2012). The chromatin remodeling and mRNA splicing functions of the Brahma (SWI/SNF) complex are mediated by the SNR1/SNF5 regulatory subunit. Nucleic Acids Res 40: 5975-5987. PubMed ID: 22467207

  • Marc Dionne Centre for Molecular & Cellular Biology of Inflammation, Kings College London
    Vincent, C. M., Beckwith, E. J., Simoes da Silva, C. J., Pearson, W. H., Kierdorf, K., Gilestro, G. F. and Dionne, M. S. (2022). Infection increases activity via Toll dependent and independent mechanisms in Drosophila melanogaster. PLoS Pathog 18(9): e1010826. PubMed ID: 36129961

    Vincent, C. M. and Dionne, M. S. (2021). Disparate regulation of IMD signaling drives sex differences in infection pathology in Drosophila melanogaster. Proc Natl Acad Sci U S A 118(32). PubMed ID: 34341118

    Vincent, C. M., Simoes da Silva, C. J., Wadhawan, A. and Dionne, M. S. (2020). Origins of Metabolic Pathology in Francisella-Infected Drosophila. Front Immunol 11: 1419. PubMed ID: 32733472

    Sharrock, J., Estacio-Gomez, A., Jacobson, J., Kierdorf, K., Southall, T. D. and Dionne, M. S. (2019). fs(1)h controls metabolic and immune function and enhances survival via AKT and FOXO in Drosophila. Dis Model Mech 12(4). PubMed ID: 30910908

    Woodcock, K.J., Kierdorf, K., Pouchelon, C.A., Vivancos, V., Dionne, M.S. and Geissmann, F. (2014). Macrophage-derived Upd3 cytokine causes impaired glucose homeostasis and reduced lifespan in Drosophila fed a lipid-rich diet. Immunity [Epub ahead of print]. Pubmed ID: 25601202

    Clark, R. I., Tan, S. W., Pean, C. B., Roostalu, U., Vivancos, V., Bronda, K., Pilatova, M., Fu, J., Walker, D. W., Berdeaux, R., Geissmann, F. and Dionne, M. S. (2013). MEF2 Is an In Vivo Immune-Metabolic Switch. Cell. PubMed ID: 24075010

    Pean, C. B. and Dionne, M. S. (2013). Intracellular infections in Drosophila melanogaster: host defense and mechanisms of pathogenesis. Dev Comp Immunol. PubMed ID: 23648644

    Pilatova, M. and Dionne, M. S. (2012). Burkholderia thailandensis is virulent in Drosophila melanogaster. PLoS One 7: e49745. PubMed ID: 23209596

    Clark, R. I., Woodcock, K. J., Geissmann, F., Trouillet, C. and Dionne, M. S. (2011). Multiple TGF-beta superfamily signals modulate the adult Drosophila immune response. Curr Biol 21: 1672-1677. PubMed ID: 21962711

  • Luisa Di Stefano Laboratoire de biologie cellulaire et moléculaire du contrôle de la prolifération (LBCMCP) - CNRS / Université Paul Sabatier, Toulouse, France
    Lepesant, J. M. J., Iampietro, C., Galeota, E., Auge, B., Aguirrenbengoa, M., Merce, C., Chaubet, C., Rocher, V., Haenlin, M., Waltzer, L., Pelizzola, M. and Di Stefano, L. (2019). A dual role of dLsd1 in oogenesis: regulating developmental genes and repressing transposons. Nucleic Acids Res. PubMed ID: 31799607

    Miles, W. O., Lepesant, J. M., Bourdeaux, J., Texier, M., Kerenyi, M. A., Nakakido, M., Hamamoto, R., Orkin, S. H., Dyson, N. J. and Di Stefano, L. (2015). The LSD1 Family of Histone Demethylases and the Pumilio Posttranscriptional Repressor Function in a Complex Regulatory Feedback Loop. Mol Cell Biol 35(24): 4199-4211. PubMed ID: 26438601

    Dietz, K. N., Di Stefano, L., Maher, R. C., Zhu, H., Macdonald, M. E., Gusella, J. F. and Walker, J. A. (2015). The Drosophila Huntington's disease gene ortholog dhtt influences chromatin regulation during development. Hum Mol Genet 24(2): 330-345. PubMed ID: 25168387

  • Stefano Di Talia
    Hayden, L., Chao, A., Deneke, V. E., Vergassola, M., Puliafito, A. and Di Talia, S. (2022). Cullin-5 mutants reveal collective sensing of the nucleocytoplasmic ratio in Drosophila embryogenesis. Curr Biol 32(9): 2084-2092 e2084. PubMed ID: 35334230

    Ferree, P. L., Xing, M., Zhang, J. Q. and Di Talia, S. (2022). Structure-function analysis of Cdc25(Twine) degradation at the Drosophila maternal-to-zygotic transition. Fly (Austin) 16(1): 111-117. PubMed ID: 35227166

    Deneke, V. E., Puliafito, A., Krueger, D., Narla, A. V., De Simone, A., Primo, L., Vergassola, M., De Renzis, S. and Di Talia, S. (2019). Self-Organized Nuclear Positioning Synchronizes the Cell Cycle in Drosophila Embryos. Cell 177(4): 925-941 e917. PubMed ID: 30982601

    Vergassola, M., Deneke, V. E. and Di Talia, S. (2018). Mitotic waves in the early embryogenesis of Drosophila: Bistability traded for speed. Proc Natl Acad Sci U S A 115(10): E2165-E2174. PubMed ID: 29449348

  • Anita Devineni Department of Biology, Emory University, Atlanta, GA
    Deere, J. U. and Devineni, A. V. (2022). Taste cues elicit prolonged modulation of feeding behavior in Drosophila. iScience 25(10): 105159. PubMed ID: 36204264

    Devineni, A. V. and Scaplen, K. M. (2021). Neural Circuits Underlying Behavioral Flexibility: Insights From Drosophila. Front Behav Neurosci 15: 821680. PubMed ID: 35069145

    Devineni, A. V., Deere, J. U., Sun, B. and Axel, R. (2021). Individual bitter-sensing neurons in Drosophila exhibit both ON and OFF responses that influence synaptic plasticity. Curr Biol 31(24): 5533-5546 e5537. PubMed ID: 34731675

    Devineni, A. V., Sun, B., Zhukovskaya, A. and Axel, R. (2019). Acetic acid activates distinct taste pathways in Drosophila to elicit opposing, state-dependent feeding responses. Elife 8. PubMed ID: 31205005

  • Leonard Dobens University of Missouri, Kansas City
    Das, R., Sebo, Z., Pence, L. and Dobens, L. L. (2014). Drosophila Tribbles antagonizes insulin signaling-mediated growth and metabolism via interactions with Akt kinase. PLoS One 9: e109530. PubMed ID: 25329475

    Dobens, A. C. and Dobens, L. L. (2013). FijiWings: An Open Source Toolkit for Semi-Automated Morphometric Analysis of Insect Wings. G3 (Bethesda). PubMed ID: 23797110

    Masoner, V., Das, R., Pence, L., Anand, G., Laferriere, H., Zars, T., Bouyain, S., Dobens, L. L. (2013) The kinase domain of Drosophila Tribbles is required for turnover of fly C/EBP during cell migration. Dev Biol. PubMed ID: 23305818

    Dobens, L. L., Jr. and Bouyain, S. (2012). Developmental roles of tribbles protein family members. Dev Dyn 241: 1239-1248. PubMed ID: 22711497

    Levine, B., Hackney, J. F., Bergen, A., Dobens, L., 3rd, Truesdale, A. and Dobens, L. (2010). Opposing interactions between Drosophila cut and the C/EBP encoded by slow border cells direct apical constriction and epithelial invagination. Dev Biol 344: 196-209. PubMed ID: 20450903

  • Chris Doe University of Oregon, Institute of Neuroscience
    Munroe, J. A., Syed, M. H. and Doe, C. Q. (2022). Imp is required for timely exit from quiescence in Drosophila type II neuroblasts. PLoS One 17(12): e0272177. PubMed ID: 36520944

    Linskens, A., Doe, C. and Lee, K. (2022). Developmental origin of the Pair1 descending interneuron. MicroPubl Biol 2022. PubMed ID: 36606082

    Seroka, A., Lai, S. L. and Doe, C. Q. (2022). Transcriptional profiling from whole embryos to single neuroblast lineages in Drosophila. Dev Biol 489: 21-33. PubMed ID: 35660371

    Lee, K. M., Linskens, A. M. and Doe, C. Q. (2022). Hunchback activates Bicoid in Pair1 neurons to regulate synapse number and locomotor circuit function. Curr Biol 32(11): 2430-2441. PubMed ID: 35512697

    Seroka, A., Lai, S. L. and Doe, C. Q. (2022). Transcriptional profiling from whole embryos to single neuroblast lineages in Drosophila. Dev Biol 489: 21-33. PubMed ID: 35660371

    Carreira-Rosario, A., York, R. A., Choi, M., Doe, C. Q. and Clandinin, T. R. (2021). Mechanosensory input during circuit formation shapes Drosophila motor behavior through patterned spontaneous network activity. Curr Biol. PubMed ID: 34478644

    Lee, K. and Doe, C. Q. (2021). A locomotor neural circuit persists and functions similarly in larvae and adult Drosophila. Elife 10. PubMed ID: 34259633

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

    Ackerman, S. D., Perez-Catalan, N. A., Freeman, M. R. and Doe, C. Q. (2021). Astrocytes close a motor circuit critical period. Nature 592(7854): 414-420. PubMed ID: 33828296

    Seroka, A., Yazejian, R. M., Lai, S. L. and Doe, C. Q. (2020). A novel temporal identity window generates alternating Eve(+)/Nkx6(+) motor neuron subtypes in a single progenitor lineage. Neural Dev 15(1): 9. PubMed ID: 32723364

    Zarin, A. A., Mark, B., Cardona, A., Litwin-Kumar, A. and Doe, C. Q. (2019). A multilayer circuit architecture for the generation of distinct locomotor behaviors in Drosophila. Elife 8. PubMed ID: 31868582

    Sales, E. C., Heckman, E. L., Warren, T. L. and Doe, C. Q. (2019). Regulation of subcellular dendritic synapse specificity by axon guidance cues. Elife 8. PubMed ID: 31012844

    Seroka, A. Q. and Doe, C. Q. (2019). The Hunchback temporal transcription factor determines motor neuron axon and dendrite targeting in Drosophila. Development. PubMed ID: 30890568

    Sullivan, L. F., Warren, T. L. and Doe, C. Q. (2019). Temporal identity establishes columnar neuron morphology, connectivity, and function in a Drosophila navigation circuit. Elife 8. PubMed ID: 30706848

    Sen, S. Q., Chanchani, S., Southall, T. D. and Doe, C. Q. (2019). Neuroblast-specific open chromatin allows the temporal transcription factor, Hunchback, to bind neuroblast-specific loci. Elife 8. PubMed ID: 30694180

  • Tomas Dolezal Department of Molecular Biology, Laboratory of Drosophila model for human diseases, Faculty of Science University of South Bohemia, Ceske Budejovice, Czech Republic
    Thakkar, N., Giesecke, A., Bazalova, O., Martinek, J., Smykal, V., Stanewsky, R. and Dolezel, D. (2022). Evolution of casein kinase 1 and functional analysis of new doubletime mutants in Drosophila. Front Physiol 13: 1062632. PubMed ID: 36589447

    Kotwica-Rolinska, J., Chodakova, L., Smykal, V., Damulewicz, M., Provaznik, J., Wu, B. C., Hejnikova, M., Chvalova, D. and Dolezel, D. (2022). Loss of Timeless Underlies an Evolutionary Transition within the Circadian Clock. Mol Biol Evol 39(1). PubMed ID: 34893879

    Bajgar, A. and Dolezal, T. (2018). Extracellular adenosine modulates host-pathogen interactions through regulation of systemic metabolism during immune response in Drosophila. PLoS Pathog 14(4): e1007022. PubMed ID: 29702691

    Fenckova, M., Hobizalova, R., Fric, Z. F. and Dolezal, T. (2011). Functional characterization of ecto-5'-nucleotidases and apyrases in Drosophila melanogaster. Insect Biochem Mol Biol 41: 956-967. PubMed ID: 21996016

    Novakova, M. and Dolezal, T. (2011). Expression of Drosophila adenosine deaminase in immune cells during inflammatory response. PLoS One 6: e17741. PubMed ID: 21412432


  • Patrick Dolph Molecular & Cellular Biology Graduate Program, Dartmouth University
    Li, X., Abou Tayoun, A., Song, Z., Dau, A., Rien, D., Jaciuch, D., Dongre, S., Blanchard, F., Nikolaev, A., Zheng, L., Bollepalli, M. K., Chu, B., Hardie, R. C., Dolph, P. J. and Juusola, M. (2019). Ca(2+)-activated K(+) channels reduce network excitability, improving adaptability and energetics for transmitting and perceiving sensory information. J Neurosci. PubMed ID: 31350259

    Murali, A., Maue, R. A. and Dolph, P. J. (2014). Reversible symptoms and clearance of mutant prion protein in an inducible model of a genetic prion disease in Drosophila melanogaster. Neurobiol Dis. PubMed ID: 24686303

    Abou Tayoun, A. N., Pikielny, C. and Dolph, P. J. (2012). Roles of the Drosophila SK channel (dSK) in courtship memory. PLoS One 7: e34665. PubMed ID: 22509342

    Kinser, R. D. and Dolph, P. J. (2012). Cathepsin proteases mediate photoreceptor cell degeneration in Drosophila. Neurobiol Dis 46: 655-662. PubMed ID: 22426392

  • Maria Dominguez Developmental Neurobiology, CSIC-Universidad Miguel Hernández, Instituto Neurociencias, Alicante, Spain
    Reiff, T., Antonello, Z. A., Ballesta-Illan, E., Mira, L., Sala, S., Navarro, M., Martinez, L. M. and Dominguez, M. (2019). Notch and EGFR regulate apoptosis in progenitor cells to ensure gut homeostasis in Drosophila. EMBO J: e101346. PubMed ID: 31566767

    Gutierrez-Perez, I., Rowley, M. J., Lyu, X., Valadez-Graham, V., Vallejo, D. M., Ballesta-Illan, E., Lopez-Atalaya, J. P., Kremsky, I., Caparros, E., Corces, V. G. and Dominguez, M. (2019). Ecdysone-Induced 3D Chromatin Reorganization Involves Active Enhancers Bound by Pipsqueak and Polycomb. Cell Rep 28(10): 2715-2727.e2715. PubMed ID: 31484080

    Reiff, T., Jacobson, J., Cognigni, P., Antonello, Z., Ballesta, E., Tan, K.J., Yew, J.Y., Dominguez, M. and Miguel-Aliaga, I. (2015). Endocrine remodelling of the adult intestine sustains reproduction in Drosophila. Elife 4. PubMed ID: 26216039

    Antonello, Z. A., Reiff, T., Ballesta-Illan, E. and Dominguez, M. (2015). Robust intestinal homeostasis relies on cellular plasticity in enteroblasts mediated by miR-8-Escargot switch. EMBO J [Epub ahead of print]. PubMed

    Antonello, Z.A., Reiff, T., Ballesta-Illan, E. and Dominguez, M. (2015). Robust intestinal homeostasis relies on cellular plasticity in enteroblasts mediated by miR-8-Escargot switch. EMBO J [Epub ahead of print]. PubMed ID: 26077448

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

    Dominguez, M. (2014). Oncogenic programmes and Notch activity: An 'organized crime'? Semin Cell Dev Biol. PubMed ID: 24780858

    Morante, J., Vallejo, D. M., Desplan, C. and Dominguez, M. (2013). Conserved miR-8/miR-200 Defines a Glial Niche that Controls Neuroepithelial Expansion and Neuroblast Transition. Dev Cell. PubMed ID: 24139822

    Da Ros, V. G., Gutierrez-Perez, I., Ferres-Marco, D., Dominguez, M. (2013) Dampening the Signals Transduced through Hedgehog via MicroRNA miR-7 Facilitates Notch-Induced Tumourigenesis. PLoS Biol 11: e1001554. PubMed ID: 23667323

    Garelli, A., Gontijo, A. M., Miguela, V., Caparros, E. and Dominguez, M. (2012). Imaginal discs secrete insulin-like peptide 8 to mediate plasticity of growth and maturation. Science 336: 579-582. PubMed ID: 22556250

  • Nathalie Dostatni Institut Curie, Paris
    Fernandes, G., Tran, H., Andrieu, M., Diaw, Y., Perez Romero, C., Fradin, C., Coppey, M., Walczak, A. M. and Dostatni, N. (2022). Synthetic reconstruction of the hunchback promoter specifies the role of Bicoid, Zelda and Hunchback in the dynamics of its transcription. Elife 11. PubMed ID: 35363606

    Desponds, J., Tran, H., Ferraro, T., Lucas, T., Perez Romero, C., Guillou, A., Fradin, C., Coppey, M., Dostatni, N. and Walczak, A. M. (2016). Precision of Readout at the hunchback Gene: Analyzing Short Transcription Time Traces in Living Fly Embryos. PLoS Comput Biol 12(12): e1005256. PubMed ID: 27942043

    Roelens, B., Clemot, M., Leroux-Coyau, M., Klapholz, B. and Dostatni, N. (2017). Maintenance of Heterochromatin by the Large Subunit of the CAF-1 Replication-Coupled Histone Chaperone Requires Its Interaction with HP1a Through a Conserved Motif. Genetics 205(1): 125-137. PubMed ID: 27838630

    Ferraro, T., Esposito, E., Mancini, L., Ng, S., Lucas, T., Coppey, M., Dostatni, N., Walczak, A. M., Levine, M. and Lagha, M. (2016). Transcriptional Memory in the Drosophila Embryo. Curr Biol 26(2): 212-218. PubMed ID: 26748851

    Lucas, T., Ferraro, T., Roelens, B., De Las Heras Chanes, J., Walczak, A. M., Coppey, M. and Dostatni, N. (2013). Live imaging of bicoid-dependent transcription in Drosophila embryos. Curr Biol 23(21): 2135-2139. PubMed ID: 24139736

  • Ke Dong Department of Entomology, Michigan State University, East Lansing
    Wang, Q., Xu, P., Andreazza, F., Liu, Y., Nomura, Y., Duran, P., Jiang, L., Chen, M., Takamatsu, G., Ihara, M., Matsuda, K., Isaacs, R., Oliveira, E. E., Du, Y. and Dong, K. (2021). Identification of multiple odorant receptors essential for pyrethrum repellency in Drosophila melanogaster. PLoS Genet 17(7): e1009677. PubMed ID: 34237075

    Wang, L., Du, Y., Nomura, Y. and Dong, K. (2015). Distinct modulating effects of TipE-homologs 2-4 on Drosophila sodium channel splice variants. Insect Biochem Mol Biol. PubMed ID: 25744892

    Rinkevich, F. D., Du, Y., Tolinski, J., Ueda, A., Wu, C. F., Zhorov, B. S. and Dong, K. (2015). Distinct roles of the DmNa and DSC1 channels in the action of DDT and pyrethroids. Neurotoxicology [Epub ahead of print]. PubMed ID: 25687544

    Zhang, T., Wang, Z., Wang, L., Luo, N., Jiang, L., Liu, Z., Wu, C. F., Dong, K. (2013) Role of the DSC1 Channel in Regulating Neuronal Excitability in Drosophila melanogaster: Extending Nervous System Stability under Stress. PLoS Genet 9: e1003327. PubMed ID: 23505382

    Wang, S., Yang, J., Tsai, A., Kuca, T., Sanny, J., Lee, J., Dong, K., Harden, N. and Krieger, C. (2011). Drosophila adducin regulates Dlg phosphorylation and targeting of Dlg to the synapse and epithelial membrane. Dev Biol 357: 392-403. PubMed ID: 21791202

    Du, Y., Garden, D. P., Wang, L., Zhorov, B. S. and Dong, K. (2011). Identification of new batrachotoxin-sensing residues in segment IIIS6 of the sodium channel. J Biol Chem 286: 13151-13160. PubMed ID: 21303907

    Shao, Y. M., Dong, K., Tang, Z. H. and Zhang, C. X. (2009). Molecular characterization of a sodium channel gene from the Silkworm Bombyx mori. Insect Biochem Mol Biol 39: 145-151. PubMed ID: 18996195

  • Jeffrey Donlea ​Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA
    Weiss, J. T. and Donlea, J. M. (2021). Sleep deprivation results in diverse patterns of synaptic scaling across the Drosophila mushroom bodies. Curr Biol. PubMed ID: 34107302

    Omoto, J. J., Nguyen, B. M., Kandimalla, P., Lovick, J. K., Donlea, J. M. and Hartenstein, V. (2018). Neuronal Constituents and Putative Interactions Within the Drosophila Ellipsoid Body Neuropil. Front Neural Circuits 12: 103. PubMed ID: 30546298

    Donlea, J. M., Pimentel, D. and Miesenbock, G. (2014). Neuronal machinery of sleep homeostasis in Drosophila. Neuron 81(4): 860-872. PubMed ID: 24559676

  • Gerald Dorn Division of Biology and Biomedical Sciences, Washington University St. Louis
    Bhandari, P., Song, M. and Dorn, G. W. (2014). Dissociation of mitochondrial from sarcoplasmic reticular stress in Drosophila cardiomyopathy induced by molecularly distinct mitochondrial fusion defects. J Mol Cell Cardiol 80C: 71-80. PubMed ID: 25555803

    Bhandari, P., Song, M., Chen, Y., Burelle, Y. and Dorn, G. W. (2014). Mitochondrial contagion induced by Parkin deficiency in Drosophila hearts and its containment by suppressing mitofusin. Circ Res 114: 257-265. PubMed ID: 24192653

    Chen, Y., Sparks, M., Bhandari, P., Matkovich, S. J. and Dorn, G. W. (2014). Mitochondrial genome linearization is a causative factor for cardiomyopathy in mice and Drosophila. Antioxid Redox Signal 21: 1949-1959. PubMed ID: 23909626

  • Silke Dorner Max F. Perutz Laboratories, Vienna
    Antic, S., Wolfinger, M. T., Skucha, A., Hosiner, S. and Dorner, S. (2015). General and miRNA-mediated mRNA degradation occurs on ribosome complexes in Drosophila cells. Mol Cell Biol. PubMed ID: 25918245

    Barisic-Jager, E., Krecioch, I., Hosiner, S., Antic, S. and Dorner, S. (2013). HPat a decapping activator interacting with the miRNA effector complex. PLoS One 8: e71860. PubMed ID: 23977167

  • Dale Dorsett
    Pherson, M., Misulovin, Z., Gause, M. and Dorsett, D. (2019). Cohesin occupancy and composition at enhancers and promoters are linked to DNA replication origin proximity in Drosophila. Genome Res. PubMed ID: 30796039

    Swain, A., Misulovin, Z., Pherson, M., Gause, M., Mihindukulasuriya, K., Rickels, R. A., Shilatifard, A. and Dorsett, D. (2016). Drosophila TDP-43 RNA-Binding Protein Facilitates Association of Sister Chromatid Cohesion Proteins with Genes, Enhancers and Polycomb Response Elements. PLoS Genet 12: e1006331. PubMed ID: 27662615

    Wu, Y., Gause, M., Xu, D., Misulovin, Z., Schaaf, C. A., Mosarla, R. C., Mannino, E., Shannon, M., Jones, E., Shi, M., Chen, W. F., Katz, O. L., Sehgal, A., Jongens, T. A., Krantz, I. D. and Dorsett, D. (2015). Drosophila Nipped-B mutants model Cornelia de Lange Syndrome in growth and behavior. PLoS Genet 11: e1005655. PubMed ID: 26544867

    Dorsett, D. (2013). What fruit flies can tell us about human birth defects. Mo Med 110: 309-313. PubMed ID: 24003648

    Schaaf, C. A., Misulovin, Z., Gause, M., Koenig, A. and Dorsett, D. (2013). The Drosophila Enhancer of Split Gene Complex: Architecture and Coordinate Regulation by Notch, Cohesin and Polycomb Group Proteins. G3 (Bethesda). PubMed ID: 23979932

    Schaaf, C. A., Misulovin, Z., Gause, M., Koenig, A., Gohara, D. W., Watson, A. and Dorsett, D. (2013). Cohesin and polycomb proteins functionally interact to control transcription at silenced and active genes. PLoS Genet 9: e1003560. PubMed ID: 23818863

    Schaaf, C. A., Kwak, H., Koenig, A., Misulovin, Z., Gohara, D. W., Watson, A., Zhou, Y., Lis, J. T. and Dorsett, D. (2013). Genome-wide control of RNA polymerase II activity by cohesin. PLoS Genet 9: e1003382. PubMed ID: 23555293

    Fay, A., Misulovin, Z., Li, J., Schaaf, C. A., Gause, M., Gilmour, D. S. and Dorsett, D. (2011). Cohesin selectively binds and regulates genes with paused RNA polymerase. Curr Biol 21: 1624-1634. PubMed ID: 21962715

    Cunningham, M. D., Gause, M., et al. (2012). Wap1 antagonizes cohesin binding and promotes Polycomb-group silencing in Drosophila. Development 139(22): 4172-9. PubMed Citation: 23034634

  • Steve Dorus Department of Biology, Center for Reproductive Evolution, Syracuse University, Syracuse, NY
    McCullough, E. L., Whittington, E., Singh, A., Pitnick, S., Wolfner, M. F. and Dorus, S. (2022). The life history of Drosophila sperm involves molecular continuity between male and female reproductive tracts. Proc Natl Acad Sci U S A 119(11): e2119899119. PubMed ID: 35254899

    McDonough-Goldstein, C. E., Pitnick, S. and Dorus, S. (2022). Drosophila female reproductive glands contribute to mating plug composition and the timing of sperm ejection. Proc Biol Sci 289(1968): 20212213. PubMed ID: 35105240

    McDonough-Goldstein, C. E., Whittington, E., McCullough, E. L., Buel, S. M., Erdman, S., Pitnick, S. and Dorus, S. (2021). Pronounced Postmating Response in the Drosophila Female Reproductive Tract Fluid Proteome. Mol Cell Proteomics 20: 100156. PubMed ID: 34597791

  • Angela Douglas Department of Entomology, Cornell

    McMullen, J. G., Peters-Schulze, G., Cai, J., Patterson, A. D. and Douglas, A. E. (2020). How gut microbiome interactions affect nutritional traits of Drosophila melanogaster. J Exp Biol 223(Pt 19). PubMed ID: 33051361

    Kang, D. and Douglas, A. E. (2020). Functional traits of the gut microbiome correlated with host lipid content in a natural population of Drosophila melanogaster. Biol Lett 16(2): 20190803. PubMed ID: 32097599

    Fromont, C., Adair, K. L. and Douglas, A. E. (2019). Correlation and causation between the microbiome, Wolbachia and host functional traits in natural populations of drosophilid flies. Mol Ecol. PubMed ID: 30714238

    Bost, A., Franzenburg, S., Adair, K. L., Martinson, V. G., Loeb, G. and Douglas, A. E. (2017). How gut transcriptional function of Drosophila melanogaster varies with the presence and composition of the gut microbiota. Mol Ecol. PubMed ID: 29113026

    Kim, G., Huang, J. H., McMullen, J. G., Newell, P. D. and Douglas, A. E. (2017). Physiological responses of insects to microbial fermentation products: insights from the interactions between Drosophila and acetic acid. J Insect Physiol. PubMed ID: 28522417

    Dobson, A. J., Chaston, J. M. and Douglas, A. E. (2016). The Drosophila transcriptional network is structured by microbiota. BMC Genomics 17(1): 975. PubMed ID: 27887564

    Chaston, J. M., Dobson, A. J., Newell, P. D. and Douglas, A. E. (2015). Host genetic control of the microbiota mediates Drosophila nutritional phenotype. Appl Environ Microbiol [Epub ahead of print]. PubMed ID: 26567306

    Huang, J.H. and Douglas, A.E. (2015). Consumption of dietary sugar by gut bacteria determines Drosophila lipid content. Biol Lett 11(9). PubMed ID: 26382071

    Wong, A.C., Luo, Y., Jing, X., Franzenburg, S., Bost, A. and Douglas, A.E. (2015). The host as driver of the microbiota in the gut and external environment of Drosophila melanogaster. Appl Environ Microbiol [Epub ahead of print]. PubMed ID: 26150460

    Jing, X., White, T. A., Yang, X. and Douglas, A. E. (2015). The molecular correlates of organ loss: the case of insect Malpighian tubules. Biol Lett 11. PubMed ID: 25972400


  • Julian Dow University of Glasgow
    Xu, J., Liu, Y., Li, H., Tarashansky, A. J., Kalicki, C. H., Hung, R. J., Hu, Y., Comjean, A., Kolluru, S. S., Wang, B., Quake, S. R., Luo, L., McMahon, A. P., Dow, J. A. T. and Perrimon, N. (2022). Transcriptional and functional motifs defining renal function revealed by single-nucleus RNA sequencing. Proc Natl Acad Sci U S A 119(25): e2203179119. PubMed ID: 35696569

    Kanoh, H., Iwashita, S., Kuraishi, T., Goto, A., Fuse, N., Ueno, H., Nimura, M., Oyama, T., Tang, C., Watanabe, R., Hori, A., Momiuchi, Y., Ishikawa, H., Suzuki, H., Nabe, K., Takagaki, T., Fukuzaki, M., Tong, L. L., Yamada, S., Oshima, Y., Aigaki, T., Dow, J. A. T., Davies, S. A. and Kurata, S. (2021). cGMP signaling pathway that modulates NF-κB activation in innate immune responses. iScience 24(12): 103473. PubMed ID: 34988396

    Koyama, T., Terhzaz, S., Naseem, M. T., Nagy, S., Rewitz, K., Dow, J. A. T., Davies, S. A. and Halberg, K. V. (2021). A nutrient-responsive hormonal circuit mediates an inter-tissue program regulating metabolic homeostasis in adult Drosophila. Nat Commun 12(1): 5178. PubMed ID: 34462441

    Cabrero, P., Terhzaz, S., Dornan, A. J., Ghimire, S., Holmes, H. L., Turin, D. R., Romero, M. F., Davies, S. A. and Dow, J. A. T. (2020). Specialized stellate cells offer a privileged route for rapid water flux in Drosophila renal tubule. Proc Natl Acad Sci U S A 117(3): 1779-1787. PubMed ID: 31907321

    Ghimirie, S., Terhzaz, S., Cabrero, P., Romero, M. F., Davies, S. and Dow, J. A. T. (2019). Targeted renal knockdown of Na(+)/H(+) exchanger regulatory factor Sip1 produces uric acid nephrolithiasis in Drosophila. Am J Physiol Renal Physiol. PubMed ID: 31364377

    Martinez-Corrales, G., Cabrero, P., Dow, J. A. T., Terhzaz, S. and Davies, S. A. (2019). Novel roles for GATAe in growth, maintenance and proliferation of cell populations in the Drosophila renal tubule. Development 146(9). PubMed ID: 31036543

    Overend, G., Luo, Y., Henderson, L., Douglas, A.E., Davies, S.A. and Dow, J.A. (2016). Molecular mechanism and functional significance of acid generation in the Drosophila midgut. Sci Rep 6: 27242. PubMed ID: 27250760

    Halberg, K.A., Rainey, S.M., Veland, I.R., Neuert, H., Dornan, A.J., Klämbt, C., Davies, S.A. and Dow, J.A. (2016). The cell adhesion molecule Fasciclin2 regulates brush border length and organization in Drosophila renal tubules. Nat Commun 7: 11266. PubMed ID: 27072072

    Cannell, E., Dornan, A. J., Halberg, K. A., Terhzaz, S., Dow, J. A. and Davies, S. A. (2016). The corticotropin-releasing factor-like diuretic hormone 44 (DH) and kinin neuropeptides modulate desiccation and starvation tolerance in Drosophila melanogaster. Peptides [Epub ahead of print]. PubMed ID: 26896569

    Landry, G. M., Hirata, T., Anderson, J. B., Cabrero, P., Gallo, C. J., Dow, J. A. and Romero, M. F. (2015). Sulfate and thiosulfate inhibit oxalate transport via a dPrestin(mSlc26a6)-dependent mechanism in an insect model of calcium oxalate nephrolithiasis. Am J Physiol Renal Physiol: ajprenal 00406 02015. PubMed ID: 26538444

    Brinzer, R. A., Henderson, L., Marchiondo, A. A., Woods, D. J., Davies, S. A. and Dow, J. A. (2015). Metabolomic profiling of permethrin-treated Drosophila melanogaster identifies a role for tryptophan catabolism in insecticide survival. Insect Biochem Mol Biol. PubMed ID: 26474926

  • Damian Dowling Monash University, Melbourne
    Dobson, A. J., Voigt, S., Kumpitsch, L., Langer, L., Voigt, E., Ibrahim, R., Dowling, D. K. and Reinhardt, K. (2023). Mitonuclear interactions shape both direct and parental effects of diet on fitness and involve a SNP in mitoribosomal 16s rRNA. PLoS Biol 21(8): e3002218. PubMed ID: 37603597

    Brand, J. A., Yee, W. K. W., Aitkenhead, I. J., Martin, J. M., Polverino, G., Chown, S. L., Wong, B. B. M. and Dowling, D. K. (2023). Temperature change exerts sex-specific effects on behavioural variation. Proc Biol Sci 290(2002): 20230110. PubMed ID: 37403505

    Layh, S., Nagarajan-Radha, V., Lemos, B. and Dowling, D. K. (2023). Y chromosome-linked variation affects locomotor activity in male Drosophila melanogaster and is robust to differences in thermal environment. Heredity (Edinb). PubMed ID: 36914794

    Koch, R. E. and Dowling, D. K. (2022). Effects of mitochondrial haplotype on pre-copulatory mating success in male fruit flies (Drosophila melanogaster). J Evol Biol 35(10): 1396-1402. PubMed ID: 35988150

    Keaney, T. A., Wong, H. W. S., Dowling, D. K., Jones, T. M. and Holman, L. (2020). Sibling rivalry versus mother's curse: can kin competition facilitate a response to selection on male mitochondria? Proc Biol Sci 287(1930): 20200575. PubMed ID: 32605521

    Vaught, R. C., Voigt, S., Dobler, R., Clancy, D. J., Reinhardt, K. and Dowling, D. K. (2020). Interactions between cytoplasmic and nuclear genomes confer sex-specific effects on lifespan in Drosophila melanogaster. J Evol Biol. PubMed ID: 32053259

    Nagarajan-Radha, V., Aitkenhead, I., Clancy, D. J., Chown, S. L. and Dowling, D. K. (2020). Sex-specific effects of mitochondrial haplotype on metabolic rate in Drosophila melanogaster support predictions of the Mother's Curse hypothesis. Philos Trans R Soc Lond B Biol Sci 375(1790): 20190178. PubMed ID: 31787038

    Nagarajan-Radha, V., Rapkin, J., Hunt, J. and Dowling, D. K. (2019). Interactions between mitochondrial haplotype and dietary macronutrient ratios confer sex-specific effects on longevity in Drosophila melanogaster. J Gerontol A Biol Sci Med Sci. PubMed ID: 31044222

    Nystrand, M., Cassidy, E. J. and Dowling, D. K. (2018). The effects of a bacterial challenge on reproductive success of fruit flies evolved under low or high sexual selection. Ecol Evol 8(18): 9341-9352. PubMed ID: 30377505

    Lajbner, Z., Pnini, R., Camus, M. F., Miller, J. and Dowling, D. K. (2018). Experimental evidence that thermal selection shapes mitochondrial genome evolution. Sci Rep 8(1): 9500. PubMed ID: 29934612

    Camus, M. F. and Dowling, D. K. (2018). Mitochondrial genetic effects on reproductive success: signatures of positive intrasexual, but negative intersexual pleiotropy. Proc Biol Sci 285(1879). PubMed ID: 29794041

    Wolff, J. N., Gemmell, N. J., Tompkins, D. M. and Dowling, D. K. (2017). Introduction of a male-harming mitochondrial haplotype via 'Trojan Females' achieves population suppression in fruit flies. Elife 6. PubMed ID: 28467301

    Nystrand, M., Cassidy, E. J. and Dowling, D. K. (2016). Transgenerational plasticity following a dual pathogen and stress challenge in fruit flies. BMC Evol Biol 16: 171. PubMed ID: 27567640

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

  • Jacqueline Dresch Clark University
    Dresch, J. M., Conrad, R. D., Klonaros, D. and Drewell, R. A. (2023). Investigating the sequence landscape in the Drosophila initiator core promoter element using an enhanced MARZ algorithm. PeerJ 11: e15597. PubMed ID: 37366427

    Dresch, J. M., Zellers, R. G., Bork, D. K. and Drewell, R. A. (2016). Nucleotide Interdependency in Transcription Factor Binding Sites in the Drosophila Genome. Gene Regul Syst Bio 10: 21-33. PubMed ID: 27330274

    Pettie, K. P., Dresch, J. M. and Drewell, R. A. (2016). Spatial distribution of predicted transcription factor binding sites in Drosophila ChIP peaks. Mech Dev. PubMed ID: 27264535

    Zellers, R. G., Drewell, R. A. and Dresch, J. M. (2015). MARZ: an algorithm to combinatorially analyze gapped n -mer models of transcription factor binding. BMC Bioinformatics 16: 30. PubMed ID: 25637281

    Drewell, R. A., Nevarez, M. J., Kurata, J. S., Winkler, L. N., Li, L. and Dresch, J. M. (2013). Deciphering the combinatorial architecture of a Drosophila homeotic gene enhancer. Mech Dev. PubMed ID: 24514265

  • Daniela Drummond-Barbosa School of Medicine, Johns Hopkins University
    Gandara, A. C. P. and Drummond-Barbosa, D. (2023). Chronic exposure to warm temperature causes low sperm abundance and quality in Drosophila melanogaster. Sci Rep 13(1): 12331. PubMed ID: 37518578

    Gandara, A. C. P. and Drummond-Barbosa, D. (2022). Warm and cold temperatures have distinct germline stem cell lineage effects during Drosophila oogenesis. Development 149(5). PubMed ID: 35156684

    Weaver, L. N. and Drummond-Barbosa, D. (2021). Hormone receptor 4 is required in muscles and distinct ovarian cell types to regulate specific steps of Drosophila oogenesis. Development 148(5). PubMed ID: 33547134

    Ma, T., Matsuoka, S. and Drummond-Barbosa, D. (2020). RNAi-based screens uncover a potential new role for the orphan neuropeptide receptor Moody in Drosophila female germline stem cell maintenance. PLoS One 15(12): e0243756. PubMed ID: 33307547

    Weaver, L. N. and Drummond-Barbosa, D. (2020). The Nuclear Receptor Seven Up Regulates Genes Involved in Immunity and Xenobiotic Response in the Adult Drosophila Female Fat Body. G3 (Bethesda). PubMed ID: 33087412

    Weaver, L. N., Ma, T. and Drummond-Barbosa, D. (2020). Analysis of Gal4 Expression Patterns in Adult Drosophila Females. G3 (Bethesda). PubMed ID: 32917721

    Weaver, L. N. and Drummond-Barbosa, D. (2019). The nuclear receptor seven up functions in adipocytes and oenocytes to control distinct steps of Drosophila oogenesis. Dev Biol. PubMed ID: 31470019

    Weaver, L. N. and Drummond-Barbosa, D. (2018). Maintenance of proper germline stem cell number requires adipocyte collagen in adult Drosophila females. Genetics. PubMed ID: 29884747

    Armstrong, A. R. and Drummond-Barbosa, D. (2018). Insulin signaling acts in adult adipocytes via GSK-3beta and independently of FOXO to control Drosophila female germline stem cell numbers. Dev Biol. PubMed ID: 29729259

    Matsuoka, S., Armstrong, A., Sampson, L. L., Laws, K. M. and Drummond-Barbosa, D. (2017). Adipocyte metabolic pathways regulated by diet control the female germline stem cell Lineage in Drosophila. Genetics [Epub ahead of print]. PubMed ID: 28396508

    Hsu, H. J. and Drummond-Barbosa, D. (2017). A visual screen for diet-regulated proteins in the Drosophila ovary using GFP protein trap lines. Gene Expr Patterns 23-24: 13-21. PubMed ID: 28093350

    Laws, K. M. and Drummond-Barbosa, D. (2016). AMP-activated protein kinase has diet-dependent and -independent roles in Drosophila oogenesis. Dev Biol. PubMed ID: 27729213

  • Wei Du Department for Cancer Research, University of Chicago
    Sheng, Z. and Du, W. (2020). NatB regulates Rb mutant cell death and tumor growth by modulating EGFR/MAPK signaling through the N-end rule pathways. PLoS Genet 16(6): e1008863. PubMed ID: 32559195

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

    Tanaka-Matakatsu, M., Miller, J. and Du, W. (2014). The homeodomain of Eyeless regulates cell growth and antagonizes the paired domain-dependent retinal differentiation function. Protein Cell. PubMed ID: 25234589

    Tanaka-Matakatsu, M., Miller, J., Borger, D., Tang, W. J. and Du, W. (2014). Daughterless homodimer synergizes with eyeless to induce atonal expression and retinal neuron differentiation. Dev Biol. PubMed ID: 24886829

    Gordon, G. M., Zhang, T., Zhao, J., Du, W. (2013) Deregulated G1/S control and energy stress contribute to the synthetic lethal interactions between inactivation of RB and TSC1/TSC2. J Cell Sci. PubMed ID: 23447678

    Sukhanova, M. J., Steele, L. J., Zhang, T., Gordon, G. M. and Du, W. (2011). RBF and Rno promote photoreceptor differentiation onset through modulating EGFR signaling in the Drosophila developing eye. Dev Biol 359: 190-198. PubMed ID: 21920355

    Tanaka-Matakatsu, M., Xu, J., Cheng, L. and Du, W. (2009). Regulation of apoptosis of rbf mutant cells during Drosophila development. Dev Biol 326: 347-356. PubMed ID: 19100727

  • Ranhui Duan Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.
    Xue, J., Zhu, Y., Wei, L., Huang, H., Li, G., Huang, W., Zhu, H. and Duan, R. (2022). Loss of Drosophila NUS1 results in cholesterol accumulation and Parkinson's disease-related neurodegeneration. FASEB J 36(7): e22411. PubMed ID: 35695805

    Deng, J., Guan, X. X., Zhu, Y. B., Deng, H. T., Li, G. X., Guo, Y. C., Jin, P., Duan, R. H. and Huang, W. (2022). Reducing the Excess Activin Signaling Rescues Muscle Degeneration in Myotonic Dystrophy Type 2 Drosophila Model. J Pers Med 12(3). PubMed ID: 35330385

    Zhao, J., Xue, J., Zhu, T., He, H., Kang, H., Jiang, X., Huang, W. and Duan, R. (2021). Dysregulated CRMP Mediates Circadian Deficits in a Drosophila Model of Fragile X Syndrome. Neurosci Bull 37(7): 973-984. PubMed ID: 33856646

  • Joshua Dubnau Cold Spring Harbor Laboratory
    Chang, Y. H. and Dubnau, J. (2023). Endogenous retroviruses and TDP-43 proteinopathy form a sustaining feedback driving intercellular spread of Drosophila neurodegeneration. Nat Commun 14(1): 966. PubMed ID: 36810738

    Azpurua, J., El-Karim, E. G., Tranquille, M. and Dubnau, J. (2021). A behavioral screen for mediators of age-dependent TDP-43 neurodegeneration identifies SF2/SRSF1 among a group of potent suppressors in both neurons and glia. PLoS Genet 17(11): e1009882. PubMed ID: 34723963

    Keegan, R. M., Talbot, L. R., Chang, Y. H., Metzger, M. J. and Dubnau, J. (2021). Intercellular viral spread and intracellular transposition of Drosophila gypsy. PLoS Genet 17(4): e1009535. PubMed ID: 33886543

    Chang, Y. H. and Dubnau, J. (2019). The gypsy endogenous retrovirus drives non-cell-autonomous propagation in a Drosophila TDP-43 model of neurodegeneration. Curr Biol. PubMed ID: 31495585

    Chang, Y. H., Keegan, R. M., Prazak, L. and Dubnau, J. (2019). Cellular labeling of endogenous retrovirus replication (CLEVR) reveals de novo insertions of the gypsy retrotransposable element in cell culture and in both neurons and glial cells of aging fruit flies. PLoS Biol 17(5): e3000278. PubMed ID: 31095565

    Shih, M. M., Davis, F. P., Henry, G. L. and Dubnau, J. (2018). Nuclear transcriptomes of the seven neuronal cell types that constitute the Drosophila mushroom bodies. G3 (Bethesda). PubMed ID: 30397017

    Krug, L., Chatterjee, N., Borges-Monroy, R., Hearn, S., Liao, W. W., Morrill, K., Prazak, L., Rozhkov, N., Theodorou, D., Hammell, M. and Dubnau, J. (2017). Retrotransposon activation contributes to neurodegeneration in a Drosophila TDP-43 model of ALS. PLoS Genet 13(3): e1006635. PubMed ID: 28301478

    Beshel, J., Dubnau, J. and Zhong, Y. (2017). A leptin analog locally produced in the brain acts via a conserved neural circuit to modulate obesity-linked behaviors in Drosophila. Cell Metab 25(1): 208-217. PubMed ID: 28076762

    Cressy, M., Valente, D., Altick, A., Kockenmesiter, E., Honegger, K., Qin, H., Mitra, P. P. and Dubnau, J. (2014). Laboratory evolution of adenylyl cyclase independent learning in Drosophila and missing heritability. Genes Brain Behav. PubMed ID: 24888634

    Li, W., Prazak, L., Chatterjee, N., Gruninger, S., Krug, L., Theodorou, D., Dubnau, J. (2013) Activation of transposable elements during aging and neuronal decline in Drosophila. Nat Neurosci. PubMed ID: 23563579

    Li, W., Cressy, M., Qin, H., Fulga, T., Van Vactor, D., Dubnau, J. (2013) MicroRNA-276a Functions in Ellipsoid Body and Mushroom Body Neurons for Naive and Conditioned Olfactory Avoidance in Drosophila. J Neurosci 33: 5821-5833. PubMed ID: 23536094

  • Edward Dubrovsky Biological Sciences, Fordham University, Bronx, NY
    Migunova, E., Rajamani, S., Bonanni, S., Wang, F., Zhou, C. and Dubrovsky, E. B. (2023). Cardiac RNase Z edited via CRISPR-Cas9 drives heart hypertrophy in Drosophila. PLoS One 18(5): e0286214. PubMed ID: 37228086

    Migunova, E., Theophilopoulos, J., Mercadante, M., Men, J., Zhou, C. and Dubrovsky, E. B. (2021). ELAC2/RNaseZ-linked cardiac hypertrophy in Drosophila melanogaster. Dis Model Mech 14(8). PubMed ID: 34338278

    Xie, X. and Dubrovsky, E. B. (2015). Knockout of Drosophila RNase ZL impairs mitochondrial transcript processing, respiration and cell cycle progression. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 26553808

    Bernardo, T. J., Dubrovskaya, V. A., Xie, X. and Dubrovsky, E. B. (2014). A view through a chromatin loop: insights into the ecdysone activation of early genes in Drosophila. Nucleic Acids Res. PubMed ID: 25143532

    Xie, X., Dubrovskaya, V., Yacoub, N., Walska, J., Gleason, T., Reid, K. and Dubrovsky, E. B. (2013). Developmental roles of Drosophila tRNA processing endonuclease RNase ZL as revealed with a conditional rescue system. Dev Biol 381: 324-340. PubMed ID: 23867108

    Bernardo, T. J. and Dubrovsky, E. B. (2012). The Drosophila juvenile hormone receptor candidates methoprene-tolerant (MET) and germ cell-expressed (GCE) utilize a conserved LIXXL motif to bind the FTZ-F1 nuclear receptor. J Biol Chem 287: 7821-7833. PubMed ID: 22249180

  • Carsten Duch Freie Universität Berlin
    Hurkey, S., Niemeyer, N., Schleimer, J. H., Ryglewski, S., Schreiber, S. and Duch, C. (2023). Gap junctions desynchronize a neural circuit to stabilize insect flight. Nature 618(7963): 118-125. PubMed ID: 37225999

    Krick, N., Ryglewski, S., Pichler, A., Bikbaev, A., Gotz, T., Kobler, O., Heine, M., Thomas, U. and Duch, C. (2021). Separation of presynaptic Ca(v)2 and Ca(v)1 channel function in synaptic vesicle exo- and endocytosis by the membrane anchored Ca(2+) pump PMCA. Proc Natl Acad Sci U S A 118(28). PubMed ID: 34244444

    Werner, J., Arian, J., Bernhardt, I., Ryglewski, S. and Duch, C. (2020). Differential localization of voltage-gated potassium channels during Drosophila metamorphosis. J Neurogenet: 1-18. PubMed ID: 31997675

    Kadas, D., Duch, C. and Consoulas, C. (2019). Postnatal increases in axonal conduction velocity of an identified Drosophila interneuron require fast sodium, L-type calcium and Shaker potassium channels. eNeuro. PubMed ID: 31253715

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

    Kadas, D., Klein, A., Krick, N., Worrell, J. W., Ryglewski, S. and Duch, C. (2017). Dendritic and axonal L-type calcium channels cooperate to enhance motoneuron firing output during Drosophila larval locomotion. J Neurosci 37(45): 10971-10982. PubMed ID: 28986465

    Ryglewski, S., Vonhoff, F., Scheckel, K. and Duch, C. (2017). Intra-neuronal Competition for Synaptic Partners Conserves the Amount of Dendritic Building Material. Neuron 93(3): 632-645.e636. PubMed ID: 28132832

    Williams, A.A., White, R., Siniard, A., Corneveaux, J., Huentelman, M. and Duch, C. (2016). MECP2 impairs neuronal structure by regulating KIBRA. Neurobiol Dis [Epub ahead of print]. PubMed ID: 27015692

    Kadas, D., Ryglewski, S. and Duch, C. (2015). Transient BK outward current enhances motoneurone firing rates during drosophila larval locomotion. J Physiol [Epub ahead of print]. PubMed ID: 26332699

    Ryglewski, S., Kadas, D., Hutchinson, K., Schuetzler, N., Vonhoff, F. and Duch, C. (2014). Dendrites are dispensable for basic motoneuron function but essential for fine tuning of behavior. Proc Natl Acad Sci U S A. PubMed ID: 25453076

  • Molly Duman-Scheel Medical and Molecular Genetics, Indiana University School of Medicine, South Bend
    Sarro, J., Andrews, E., Sun, L., Behura, S. K., Tan, J. C., Zeng, E., Severson, D. W. and Duman-Scheel, M. (2013). A requirement for commissureless2 function during dipteran insect nerve cord development. Dev Dyn. PubMed ID: 24026811

    Behura, S. K., Haugen, M., Flannery, E., Sarro, J., Tessier, C. R., Severson, D. W. and Duman-Scheel, M. (2011). Comparative genomic analysis of Drosophila melanogaster and vector mosquito developmental genes. PLoS One 6: e21504. PubMed ID: 21754989

    VanZomeren-Dohm, A., Sarro, J., Flannery, E. and Duman-Scheel, M. (2011). The Drosophila Netrin receptor frazzled/DCC functions as an invasive tumor suppressor. BMC Dev Biol 11: 41. PubMed ID: 21672235

  • Ian Duncan Department of Biology, Washington University in Saint Louis
    Duncan, D. M., Kiefel, P. and Duncan, I. (2017). Mutants for Drosophila Isocitrate dehydrogenase 3b are defective in mitochondrial function and larval cell death. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 28104670

    Mou, X., Duncan, D. M., Baehrecke, E. H. and Duncan, I. (2012). Control of target gene specificity during metamorphosis by the steroid response gene E93. Proc. Natl. Acad. Sci. 109(8): 2949-54. PubMed ID: 22308414

  • Jean-Maurice Dura Centre national de la recherche scientifique, Paris
    Boulanger, A., Thinat, C., Züchner, S., Fradkin, L. G., Lortat-Jacob, H. and Dura, J. M. (2021). Axonal chemokine-like Orion induces astrocyte infiltration and engulfment during mushroom body neuronal remodeling. Nat Commun 12(1): 1849. PubMed ID: 33758182

    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

    Boulanger, A. and Dura, J. M. (2014). Nuclear receptors and Drosophila neuronal remodelling. Biochim Biophys Acta. PubMed ID: 24882358

    Zirin, J., Cheng, D., Dhanyasi, N., Cho, J., Dura, J. M., Vijayraghavan, K. and Perrimon, N. (2013). Ecdysone signaling at metamorphosis triggers apoptosis of Drosophila abdominal muscles. Dev Biol. PubMed ID: 24051228

    Boulanger, A., Clouet-Redt, C., Farge, M., Flandre, A., Guignard, T., Fernando, C., Juge, F. and Dura, J. M. (2011). ftz-f1 and Hr39 opposing roles on EcR expression during Drosophila mushroom body neuron remodeling. Nat Neurosci 14: 37-44. PubMed ID: 21131955

    Boulanger, A., Farge, M., Ramanoudjame, C., Wharton, K. and Dura, J. M. (2012). Drosophila motor neuron retraction during metamorphosis is mediated by inputs from TGF-beta/BMP signaling and orphan nuclear receptors. PLoS One 7: e40255. PubMed ID: 22792255

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

  • Bénédicte Durand Institut NeuroMyoGène, Villeurbanne
    Lapart, J. A., Billon, A., Duteyrat, J. L., Thomas, J. and Durand, B. (2020). Role of DZIP1-CBY-FAM92 transition zone complex in the basal body to membrane attachment and ciliary budding. Biochem Soc Trans. PubMed ID: 32491167

    Lapart, J. A., Gottardo, M., Cortier, E., Duteyrat, J. L., Augiere, C., Mange, A., Jerber, J., Solassol, J., Gopalakrishnan, J., Thomas, J. and Durand, B. (2019). Dzip1 and Fam92 form a ciliary transition zone complex with cell type specific roles in Drosophila. Elife 8. PubMed ID: 31821146

    Augiere, C., Lapart, J. A., Duteyrat, J. L., Cortier, E., Maire, C., Thomas, J. and Durand, B. (2019). salto/CG13164 is required for sperm head morphogenesis in Drosophila. Mol Biol Cell: mbcE18070429. PubMed ID: 30601696

    Vieillard, J., Paschaki, M., Duteyrat, J. L., Augiere, C., Cortier, E., Lapart, J. A., Thomas, J. and Durand, B. (2016). Transition zone assembly and its contribution to axoneme formation in Drosophila male germ cells. J Cell Biol 214: 875-889. PubMed ID: 27646273

    Jana, S. C., Bettencourt-Dias, M., Durand, B. and Megraw, T. L. (2016). Drosophila melanogaster as a model for basal body research. Cilia 5: 22. PubMed ID: 27382461

    Vieillard, J., Duteyrat, J. L., Cortier, E. and Durand, B. (2015). Imaging cilia in Drosophila melanogaster. Methods Cell Biol 127: 279-302. PubMed ID: 25837397

    Soulavie, F., Piepenbrock, D., Thomas, J., Vieillard, J., Duteyrat, J. L., Cortier, E., Laurencon, A., Gopfert, M. C. and Durand, B. (2014). hemingway is required for sperm flagella assembly and ciliary motility in Drosophila. Mol Biol Cell 25: 1276-1286. PubMed ID: 24554765

  • Robert Duronio Department of Biology, University of North Carolina at Chapel Hill
    McPherson, J. E., Grossmann, L. C., Salzler, H. R., Armstrong, R. L., Kwon, E., Matera, A. G., McKay, D. J. and Duronio, R. J. (2023) Krupp, S., Tam, O., Gale Hammell, M. and Dubnau, J. (2023). TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1. bioRxiv. PubMed ID: 37205372

    Salzler, H. R., Vandadi, V., McMichael, B. D., Brown, J. C., Boerma, S. A., Leatham-Jensen, M. P., Adams, K. M., Meers, M. P., Simon, J. M., Duronio, R. J., McKay, D. J. and Matera, A. G. (2023). Distinct roles for canonical and variant histone H3 lysine-36 in Polycomb silencing. Sci Adv 9(9): eadf2451. PubMed ID: 36857457

    Andreyeva, E. N., Emelyanov, A. V., Nevil, M., Sun, L., Vershilova, E., Hill, C. A., Keogh, M. C., Duronio, R. J., Skoultchi, A. I. and Fyodorov, D. V. (2022). Drosophila SUMM4 complex couples insulator function and DNA replication control. Elife 11. PubMed ID: 36458689

    McPherson, J. E., Grossmann, L. C., Armstrong, R. L., Kwon, E., Salzler, H. R., Matera, A. G., McKay, D. J. and Duronio, R. J. (2023). Reduced histone gene copy number disrupts Drosophila Polycomb function. bioRxiv. PubMed ID: 37034607

    Crain, A. T., Klusza, S., Armstrong, R. L., Santa Rosa, P., Temple, B. R. S., Strahl, B. D., McKay, D. J., Matera, A. G. and Duronio, R. J. (2022). Distinct developmental phenotypes result from mutation of Set8/KMT5A and histone H4 lysine 20 in Drosophila melanogaster. Genetics. PubMed ID: 35404465

    Kemp, J. P., Jr., Yang, X. C., Dominski, Z., Marzluff, W. F. and Duronio, R. J. (2021). Superresolution light microscopy of the Drosophila histone locus body reveals a core-shell organization associated with expression of replication-dependent histone genes. Mol Biol Cell 32(9): 942-955. PubMed ID: 33788585

    Hur, W., Kemp, J. P., Jr., Tarzia, M., Deneke, V. E., Marzluff, W. F., Duronio, R. J. and Di Talia, S. (2020). CDK-Regulated Phase Separation Seeded by Histone Genes Ensures Precise Growth and Function of Histone Locus Bodies. Dev Cell. PubMed ID: 32579968

    Koreski, K. P., Rieder, L. E., McLain, L. M., Chaubal, A., Marzluff, W. F. and Duronio, R. J. (2020). Drosophila Histone Locus Body assembly and function involves multiple interactions. Mol Biol Cell: mbcE20030176. PubMed ID: 32401666

    Armstrong, R. L., Das, S., Hill, C. A., Duronio, R. J. and Nordman, J. T. (2020). Rif1 Functions in a Tissue-Specific Manner To Control Replication Timing Through Its PP1-binding Motif. Genetics. PubMed ID: 32144132

    Armstrong, R. L. and Duronio, R. J. (2019). Phasing in heterochromatin during development. Genes Dev 33(7-8): 379-381. PubMed ID: 30936191

    Armstrong, R. L., Penke, T. J. R., Chao, S. K., Gentile, G. M., Strahl, B. D., Matera, A. G., McKay, D. J. and Duronio, R. J. (2019). H3K9 Promotes under-replication of pericentromeric heterochromatin in Drosophila salivary gland Polytene Chromosomes. Genes (Basel) 10(2). PubMed ID: 30700014

  • Monica Dus Biological Sciences, University of Michigan, Ann Arbor
    Sung, H., Vaziri, A., Wilinski, D., Woerner, R. K. R., Freddolino, P. L. and Dus, M. (2023). Nutrigenomic regulation of sensory plasticity. Elife 12. PubMed ID: 36951889

    Sung, H., Vaziri, A., Wilinski, D., Woerner, R. K. R., Freddolino, P. L. and Dus, M. (2023). Nutrigenomic regulation of sensory plasticity. Elife 12. PubMed ID: 36951889

    Pardo-Garcia, T. R., Gu, K., Woerner, R. K. R. and Dus, M. (2023). Food memory circuits regulate eating and energy balance. Curr Biol 33(2): 215-227. PubMed ID: 36528025

    Vaziri, A., Khabiri, M., Genaw, B. T., May, C. E., Freddolino, P. L. and Dus, M. (2020). Persistent epigenetic reprogramming of sweet taste by diet. Sci Adv 6(46). PubMed ID: 33177090

    Wilinski, D., Winzeler, J., Duren, W., Persons, J. L., Holme, K. J., Mosquera, J., Khabiri, M., Kinchen, J. M., Freddolino, P. L., Karnovsky, A. and Dus, M. (2019). Rapid metabolic shifts occur during the transition between hunger and satiety in Drosophila melanogaster. Nat Commun 10(1): 4052. PubMed ID: 31492856

    May, C. E., Vaziri, A., Lin, Y. Q., Grushko, O., Khabiri, M., Wang, Q. P., Holme, K. J., Pletcher, S. D., Freddolino, P. L., Neely, G. G. and Dus, M. (2019). High Dietary Sugar Reshapes Sweet Taste to Promote Feeding Behavior in Drosophila melanogaster. Cell Rep 27(6): 1675-1685 e1677. PubMed ID: 31067455

    Park, J. Y., Dus, M., Kim, S., Abu, F., Kanai, M. I., Rudy, B. and Suh, G. S. B. (2016). Drosophila SLC5A11 Mediates Hunger by Regulating K(+) Channel Activity. Curr Biol 26(18): 2550. PubMed ID: 27676293

  • Atanu Duttaroy Department of Biology, Howard University, Washington, DC
    Roy, S. D., Nagarajan, S., Jalal, M. S., Basar, M. A. and Duttaroy, A. (2023). New mutant alleles for Spargel/dPGC-1 highlights the function of Spargel RRM domain in oogenesis and expands the role of Spargel in embryogenesis and intracellular transport. G3 (Bethesda). PubMed ID: 37369430

    Basar, M. A., Williamson, K., Roy, S. D., Finger, D. S., Ables, E. T. and Duttaroy, A. (2019). Spargel/dPGC-1 is essential for oogenesis and nutrient-mediated ovarian growth in Drosophila. Dev Biol. PubMed ID: 31251895

    Bailey, D., Basar, M. A., Nag, S., Bondhu, N., Teng, S. and Duttaroy, A. (2017). The essential requirement of an animal heme peroxidase protein during the wing maturation process in Drosophila. BMC Dev Biol 17(1): 1. PubMed ID: 28077066

    Mukherjee, S., Basar, M. A., Davis, C. and Duttaroy, A. (2014). Emerging functional similarities and divergences between Drosophila Spargel/dPGC-1 and mammalian PGC-1 protein. Front Genet 5: 216. PubMed ID: 25071841

    Mukherjee, S. and Duttaroy, A. (2013). Spargel/ dPGC-1 is a new downstream effector in the Insulin-TOR Signaling pathway in Drosophila. Genetics. PubMed ID: 23934892

    Armstrong, N., Ramamoorthy, M., Lyon, D., Jones, K., Duttaroy, A. (2013) Mechanism of silver nanoparticles action on insect pigmentation reveals intervention of copper homeostasis. PLoS One 8: e53186. PubMed ID: 23308159

    Vrailas-Mortimer, A., del Rivero, T., Mukherjee, S., Nag, S., Gaitanidis, A., Kadas, D., Consoulas, C., Duttaroy, A. and Sanyal, S. (2011). A muscle-specific p38 MAPK/Mef2/MnSOD pathway regulates stress, motor function, and life span in Drosophila. Dev Cell 21: 783-795. PubMed ID: 22014527

    Mukherjee, S., Forde, R., Belton, A. and Duttaroy, A. (2011). SOD2, the principal scavenger of mitochondrial superoxide, is dispensable for embryogenesis and imaginal tissue development but essential for adult survival. Fly (Austin) 5: 39-46. PubMed ID: 21212740

  • Ian Dworkin McMaster University, Hamilton Ontario
    Pelletier, K., Pitchers, W. R., Mammel, A., Northrop-Albrecht, E., Marquez, E. J., Moscarella, R. A., Houle, D. and Dworkin, I. (2023). Complexities of recapitulating polygenic effects in natural populations: replication of genetic effects on wing shape in artificially selected and wild caught populations of Drosophila melanogaster. Genetics. PubMed ID: 36961731

    Scott, A. M., Dworkin, I. and Dukas, R. (2021). Evolution of sociability by artificial selection. Evolution. PubMed ID: 34605553

    Wilson, A. E., Siddiqui, A. and Dworkin, I. (2021). Spatial heterogeneity in resources alters selective dynamics in Drosophila melanogaster. Evolution. PubMed ID: 33963761

    Chandler, C. H., Mammel, A. and Dworkin, I. (2020). Sexual Selection Does Not Increase the Rate of Compensatory Adaptation to a Mutation Influencing a Secondary Sexual Trait in Drosophila melanogaster. G3 (Bethesda). PubMed ID: 32122961

    Parigi, A., Porter, C., Cermak, M., Pitchers, W. R. and Dworkin, I. (2019). The behavioral repertoire of Drosophila melanogaster in the presence of two predator species that differ in hunting mode. PLoS One 14(5): e0216860. PubMed ID: 31150415

    Testa, N. D. and Dworkin, I. (2016). The sex-limited effects of mutations in the EGFR and TGF-beta signaling pathways on shape and size sexual dimorphism and allometry in the Drosophila wing. Dev Genes Evol. PubMed ID: 27038022

    Chandler, C. H., Chari, S., Tack, D. and Dworkin, I. (2014). Causes and Consequences of Genetic Background Effects Illuminated by Integrative Genomic Analysis. Genetics. PubMed ID: 24504186

    Chari, S. and Dworkin, I. (2013). The conditional nature of genetic interactions: the consequences of wild-type backgrounds on mutational interactions in a genome-wide modifier screen. PLoS Genet 9: e1003661. PubMed ID: 23935530

    Pitchers, W., Pool, J. E., Dworkin, I. (2013) Altitudinal clinal variation in wing size and shape in african Drosophila melanogaster: one cline or many? Evolution 67: 438-452. PubMed ID: 23356616

    Debat, V., Debelle, A. and Dworkin, I. (2009). Plasticity, canalization, and developmental stability of the Drosophila wing: joint effects of mutations and developmental temperature. Evolution 63: 2864-2876. PubMed ID: 19624729

  • Nicholas Dyson Department of Medicine, Harvard Medical School
    Guarner, A., Morris, R., Korenjak, M., Boukhali, M., Zappia, M. P., Van Rechem, C., Whetstine, J. R., Ramaswamy, S., Zou, L., Frolov, M. V., Haas, W. and Dyson, N. J. (2017). E2F/DP prevents cell-cycle progression in endocycling fat body cells by suppressing dATM expression. Dev Cell 43(6): 689-703.e685. PubMed ID: 29233476

    Miles, W. O., Korenjak, M., Griffiths, L. M., Dyer, M. A., Provero, P. and Dyson, N. J. (2014). Post-transcriptional gene expression control by NANOS is up-regulated and functionally important in pRb-deficient cells. EMBO J 33: 2201-2215. PubMed ID: 25100735

    Herr, A., Longworth, M., Ji, J. Y., Korenjak, M., Macalpine, D. M., Dyson, N. J. (2012) Identification of E2F target genes that are rate limiting for dE2F1-dependent cell proliferation. Dev Dyn 241: 1695-1707. PubMed ID: 22972499

    Nicolay, B. N., Gameiro, P. A., Tschop, K., Korenjak, M., Heilmann, A. M., Asara, J. M., Stephanopoulos, G., Iliopoulos, O., Dyson, N. J. (2013) Loss of RBF1 changes glutamine catabolism. Genes Dev. PubMed ID: 23322302

    Ji, J. Y., Miles, W. O., Korenjak, M., Zheng, Y. and Dyson, N. J. (2012). In Vivo Regulation of E2F1 by Polycomb Group Genes in Drosophila. G3 (Bethesda) 2: 1651-1660. PubMed ID: 23275887

    Korenjak, M., Anderssen, E., Ramaswamy, S., Whetstine, J. R. and Dyson, N. J. (2012). RBF binding to both canonical E2F targets and noncanonical targets depends on functional dE2F/dDP complexes. Mol Cell Biol 32: 4375-4387. PubMed ID: 22927638

    Nicolay, B. N. and Dyson, N. J. (2012). It's all in the timing: too much E2F is a bad thing. PLoS Genet 8: e1002909. PubMed ID: 22916039


  • Return to The Interactive Fly