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


  • Mart Saarma Academy of Finland, Helsinki
    Lindstrom, R., Lindholm, P., Kallijarvi, J., Palgi, M., Saarma, M. and Heino, T. I. (2016). Exploring the Conserved role of MANF in the unfolded protein response in Drosophila melanogaster. PLoS One 11: e0151550. PubMed ID: 26975047

    Kallijarvi, J., Stratoulias, V., Virtanen, K., Hietakangas, V., Heino, T. I. and Saarma, M. (2012). Characterization of Drosophila GDNF Receptor-Like and Evidence for Its Evolutionarily Conserved Interaction with Neural Cell Adhesion Molecule (NCAM)/FasII. PLoS One 7: e51997. PubMed ID: 23284846

    Lindholm, P. and Saarma, M. (2010). Novel CDNF/MANF family of neurotrophic factors. Dev Neurobiol 70: 360-371. PubMed ID: 20186704

  • Silke Sachse Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Jena
    Das Chakraborty, S. and Sachse, S. (2021). Olfactory processing in the lateral horn of Drosophila. Cell Tissue Res 383(1): 113-123. PubMed ID: 33475851

    Grabe, V., Schubert, M., Strube-Bloss, M., Reinert, A., Trautheim, S., Lavista-Llanos, S., Fiala, A., Hansson, B. S. and Sachse, S. (2019). Odor-induced multi-level inhibitory maps in Drosophila. eNeuro. PubMed ID: 31888962

    Mohamed, A. A. M., Hansson, B. S. and Sachse, S. (2019). Third-order neurons in the lateral horn enhance bilateral contrast of odor inputs through contralateral inhibition in Drosophila. Front Physiol 10: 851. PubMed ID: 31354516

    Mohamed, A. A. M., Retzke, T., Das Chakraborty, S., Fabian, B., Hansson, B. S., Knaden, M. and Sachse, S. (2019). Odor mixtures of opposing valence unveil inter-glomerular crosstalk in the Drosophila antennal lobe. Nat Commun 10(1): 1201. PubMed ID: 30867415

    Das, S., Trona, F., Khallaf, M. A., Schuh, E., Knaden, M., Hansson, B. S. and Sachse, S. (2017). Electrical synapses mediate synergism between pheromone and food odors in Drosophila melanogaster. Proc Natl Acad Sci U S A 114(46): E9962-e9971. PubMed ID: 29087946

    Strube-Bloss, M. F., Grabe, V., Hansson, B. S. and Sachse, S. (2017). Calcium imaging revealed no modulatory effect on odor-evoked responses of the Drosophila antennal lobe by two populations of inhibitory local interneurons. Sci Rep 7(1): 7854. PubMed ID: 28798324

    Seki, Y., Dweck, H. K. M., Rybak, J., Wicher, D., Sachse, S. and Hansson, B. S. (2017). Olfactory coding from the periphery to higher brain centers in the Drosophila brain. BMC Biol 15(1): 56. PubMed ID: 28666437

    Grabe, V., Baschwitz, A., Dweck, H. K., Lavista-Llanos, S., Hansson, B. S. and Sachse, S. (2016). Elucidating the neuronal architecture of olfactory glomeruli in the Drosophila antennal lobe. Cell Rep 16: 3401-3413. PubMed ID: 27653699

    Lebreton, S., Trona, F., Borrero-Echeverry, F., Bilz, F., Grabe, V., Becher, P. G., Carlsson, M. A., Nassel, D. R., Hansson, B. S., Sachse, S. and Witzgall, P. (2015). Feeding regulates sex pheromone attraction and courtship in Drosophila females. Sci Rep 5: 13132. PubMed ID: 26255707

    Dweck, H.K., Ebrahim, S.A., Thoma, M., Mohamed, A.A., Keesey, I.W., Trona, F., Lavista-Llanos, S., Svatoš, A., Sachse, S., Knaden, M. and Hansson, B.S. (2015). Pheromones mediating copulation and attraction in Drosophila. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 25964351

    Strutz, A., Soelter, J., Baschwitz, A., Farhan, A., Grabe, V., Rybak, J., Knaden, M., Schmuker, M., Hansson, B. S. and Sachse, S. (2014). Decoding odor quality and intensity in the Drosophila brain. Elife 3. PubMed ID: 25512254

  • Enrique Saez Department of Chemical Physiology, Scripps Research Institute, La Jolla, CA (see also Enrique Saez)
    Top, D., Harms, E., Syed, S., Adams, E. L. and Saez, L. (2016). GSK-3 and CK2 kinases converge on Timeless to regulate the master clock. Cell Rep. PubMed ID: 27346344

    Jang, A. R., Moravcevic, K., Saez, L., Young, M. W. and Sehgal, A. (2015). Drosophila TIM binds importin alpha1, and acts as an adapter to transport PER to the nucleus. PLoS Genet 11: e1004974. PubMed ID: 25674790

    Axelrod, S., Saez, L. and Young, M. W. (2015). Studying circadian rhythm and sleep using genetic screens in Drosophila. Methods Enzymol 551: 3-27. PubMed ID: 25662449

  • Robert Saint Flinders University, Adelaide
    Jefferies, G., Somers, J., Lohrey, I., Chaturvedi, V., Calabria, J., Marshall, O. J., Southall, T. D., Saint, R. and Murray, M. J. (2020). Maintenance of Cell Fate by the Polycomb Group Gene Sex Combs Extra Enables a Partial Epithelial Mesenchymal Transition in Drosophila. G3 (Bethesda). PubMed ID: 33051260

    Khan, M., Shaukat, Z., Saint, R. and Gregory, S. L. (2018). Chromosomal instability causes sensitivity to protein folding stress and ATP depletion. Biol Open 7(10). PubMed ID: 30327366

    Golenkina, S., Chaturvedi, V., Saint, R. and Murray, M. J. (2018). Frazzled can act through distinct molecular pathways in epithelial cells to regulate motility, apical constriction, and localisation of E-Cadherin. PLoS One 13(3): e0194003. PubMed ID: 29518139

    Hussain, R., Shaukat, Z., Khan, M., Saint, R. and Gregory, S. L. (2017). Phosphoenolpyruvate carboxykinase maintains glycolysis-driven growth in Drosophila tumors. Sci Rep 7(1): 11531. PubMed ID: 28912546

    Liu, D., Shaukat, Z., Xu, T., Denton, D., Saint, R. and Gregory, S. (2016). Autophagy regulates the survival of cells with chromosomal instability. Oncotarget [Epub ahead of print]. PubMed ID: 27590505

    Shaukat, Z., Liu, D., Choo, A., Hussain, R., O'Keefe, L., Richards, R., Saint, R. and Gregory, S. L. (2015). Chromosomal instability causes sensitivity to metabolic stress. Oncogene 34: 4044-4055. PubMed ID: 25347746

    Wong, H. W., Shaukat, Z., Wang, J., Saint, R. and Gregory, S. L. (2014). JNK signaling is needed to tolerate chromosomal instability. Cell Cycle 13: 622-631. PubMed ID: 24335260

    Pert, M., Gan, M., Saint, R. and Murray, M.J. (2015). Netrins and Frazzled/DCC promote the migration and mesenchymal to epithelial transition of Drosophila midgut cells. Biol Open [Epub ahead of print]. PubMed ID: 25617422

    Wong, H. W., Shaukat, Z., Wang, J., Saint, R. and Gregory, S. L. (2013). JNK signaling is needed to tolerate chromosomal instability. Cell Cycle 13. PubMed ID: 24335260

    Manhire-Heath, R., Golenkina, S., Saint, R. and Murray, M. J. (2013). Netrin-dependent downregulation of Frazzled/DCC is required for the dissociation of the peripodial epithelium in Drosophila. Nat Commun 4: 2790. PubMed ID: 24225841

    Murray, M. J., Ng, M. M., Fraval, H., Tan, J., Liu, W., Smallhorn, M., Brill, J. A., Field, S. J. and Saint, R. (2012). Regulation of Drosophila mesoderm migration by phosphoinositides and the PH domain of the Rho GTP exchange factor Pebble. Dev Biol 372: 17-27. PubMed ID: 23000359

  • Kuniaki Saito Keio University School of Medicine, Tokyo
    Iwasaki, Y. W., Murano, K., Ishizu, H., Shibuya, A., Iyoda, Y., Siomi, M. C., Siomi, H. and Saito, K. (2016). Piwi modulates chromatin accessibility by regulating multiple factors including Histone H1 to repress transposons. Mol Cell [Epub ahead of print]. PubMed ID: 27425411

    Ohtani, H., Iwasaki, Y. W., Shibuya, A., Siomi, H., Siomi, M. C. and Saito, K. (2013). DmGTSF1 is necessary for Piwi-piRISC-mediated transcriptional transposon silencing in the Drosophila ovary. Genes Dev 27: 1656-1661. PubMed ID: 23913921

    Saito, K. (2013) The epigenetic regulation of transposable elements by PIWI-interacting RNAs in Drosophila. Genes Genet Syst 88: 9-17. PubMed ID: 23676706

    Saito, K., Ishizu, H., Komai, M., Kotani, H., Kawamura, Y., Nishida, K. M., Siomi, H. and Siomi, M. C. (2010). Roles for the Yb body components Armitage and Yb in primary piRNA biogenesis in Drosophila. Genes Dev 24: 2493-2498. PubMed ID: 20966047

  • Minoru Saitoe Tokyo Metropolitan Institute for Neuroscience Fuchu, Japan
    Ueno, K., Morstein, J., Ofusa, K., Naganos, S., Suzuki-Sawano, E., Minegishi, S., Rezgui, S. P., Kitagishi, H., Michel, B. W., Chang, C. J., Horiuchi, J. and Saitoe, M. (2020). Carbon Monoxide, a Retrograde Messenger Generated in Postsynaptic Mushroom Body Neurons, Evokes Noncanonical Dopamine Release. J Neurosci 40(18): 3533-3548. PubMed ID: 32253360

    Matsuno, M., Horiuchi, J., Ofusa, K., Masuda, T. and Saitoe, M. (2019). Inhibiting glutamate activity during consolidation suppresses age-related long-term memory impairment in Drosophila. iScience 15: 55-65. PubMed ID: 31030182

    Miyashita, T., Kikuchi, E., Horiuchi, J. and Saitoe, M. (2018). Long-term memory engram cells are established by c-Fos/CREB transcriptional cycling. Cell Rep 25(10): 2716-2728.e2713. PubMed ID: 30517860

    Sato, S., Ueno, K., Saitoe, M. and Sakai, T. (2018). Synaptic depression induced by postsynaptic cAMP production in the Drosophila mushroom body calyx. J Physiol. PubMed ID: 29659025

    Suzuki-Sawano, E., Ueno, K., Naganos, S., Sawano, Y., Horiuchi, J. and Saitoe, M. (2017). A Drosophila ex vivo model of olfactory appetitive learning. Sci Rep 7(1): 17725. PubMed ID: 29255174

    Ueno, K., Suzuki, E., Naganos, S., Ofusa, K., Horiuchi, J. and Saitoe, M. (2017). Coincident postsynaptic activity gates presynaptic dopamine release to induce plasticity in Drosophila mushroom bodies. Elife 6 [Epub ahead of print]. PubMed ID: 28117664

    Naganos, S., Ueno, K., Horiuchi, J. and Saitoe, M. (2016). Learning defects in Drosophila growth restricted chico mutants are caused by attenuated adenylyl cyclase activity. Mol Brain 9: 37. PubMed ID: 27048332

    Matsuno, M., Horiuchi, J., Yuasa, Y., Ofusa, K., Miyashita, T., Masuda, T., and Saitoe, M. (2015). Long-term memory formation in Drosophila requires training-dependent glial transcription. J Neurosci 35: 5557-5565. PubMed ID: 25855172

    Yamazaki, D., Horiuchi, J., Ueno, K., Ueno, T., Saeki, S., Matsuno, M., Naganos, S., Miyashita, T., Hirano, Y., Nishikawa, H., Taoka, M., Yamauchi, Y., Isobe, T., Honda, Y., Kodama, T., Masuda, T. and Saitoe, M. (2014). Glial dysfunction causes age-related memory impairment in Drosophila. Neuron 84: 753-763. PubMed ID: 25447741

    Hirano, Y. and Saitoe, M. (2013). Hunger and memory; CRTC coordinates long-term memory with the physiological state, hunger. Commun Integr Biol 6: e25152. PubMed ID: 24265850

    Hirano, Y., Masuda, T., Naganos, S., Matsuno, M., Ueno, K., Miyashita, T., Horiuchi, J., Saitoe, M. (2013) Fasting launches CRTC to facilitate long-term memory formation in Drosophila. Science 339: 443-446. PubMed ID: 23349290

  • Takaomi Sakai School of Science and Engineering Department of Biological Sciences, Tokyo Metropolitan University
    Suzuki, Y., Kurata, Y. and Sakai, T. (2022). Dorsal-lateral clock neurons modulate consolidation and maintenance of long-term memory in Drosophila. Genes Cells 27(4): 266-279. PubMed ID: 35094465

    Inami, S., Sato, T., Kurata, Y., Suzuki, Y., Kitamoto, T. and Sakai, T. (2021). Consolidation and maintenance of long-term memory involve dual functions of the developmental regulator Apterous in clock neurons and mushroom bodies in the Drosophila brain. PLoS Biol 19(12): e3001459. PubMed ID: 34860826

    Inami, S., Sato, S., Kondo, S., Tanimoto, H., Kitamoto, T. and Sakai, T. (2020). Environmental light is required for maintenance of long-term memory in Drosophila. J Neurosci 40(7): 1427-1439. PubMed ID: 31932417

    Watanabe, K., Suzuki, Y., Inami, S., Ohashi, H. and Sakai, T. (2018). Light is required for proper female mate choice between winged and wingless males in Drosophila. Genes Genet Syst. PubMed ID: 29998908

    Sato, S., Ueno, K., Saitoe, M. and Sakai, T. (2018). Synaptic depression induced by postsynaptic cAMP production in the Drosophila mushroom body calyx. J Physiol. PubMed ID: 29659025

    Ohashi, H. and Sakai, T. (2018). Leucokinin signaling regulates hunger-driven reduction of behavioral responses to noxious heat in Drosophila. Biochem Biophys Res Commun 499(2): 221-226. PubMed ID: 29559237

    Shimada, N., Inami, S., Sato, S., Kitamoto, T. and Sakai, T. (2016). Modulation of light-driven arousal by LIM-homeodomain transcription factor Apterous in large PDF-positive lateral neurons of the Drosophila brain. Sci Rep 6: 37255. PubMed ID: 27853240

    Mabuchi, I., Shimada, N., Sato, S., Ienaga, K., Inami, S. and Sakai, T. (2016). Mushroom body signaling is required for locomotor activity rhythms in Drosophila. Neurosci Res. PubMed ID: 27106579

    Watanabe, K. and Sakai, T. (2016). Knockout mutations of insulin-like peptide genes enhance sexual receptivity in Drosophila virgin females. Genes Genet Syst 90: 237-241. PubMed ID: 26617266

    Sakai, T., Watanabe, K., Ohashi, H., Sato, S., Inami, S., Shimada, N. and Kitamoto, T. (2014). Insulin-producing cells regulate the sexual receptivity through the painless TRP channel in Drosophila virgin females. PLoS One 9: e88175. PubMed ID: 24505416

  • Iris Salecker Francis Crick Institute, London
    Apitz, H. and Salecker, I. (2018). Spatio-temporal relays control layer identity of direction-selective neuron subtypes in Drosophila. Nat Commun 9(1): 2295. PubMed ID: 29895891

    Apitz, H. and Salecker, I. (2016). Retinal determination genes coordinate neuroepithelial specification and neurogenesis modes in the Drosophila optic lobe. Development 143: 2431-2442. PubMed ID: 27381228

    Apitz, H. and Salecker, I. (2014). A region-specific neurogenesis mode requires migratory progenitors in the Drosophila visual system. Nat Neurosci [Epub ahead of print]. PubMed ID: 25501037

    Richier, B. and Salecker, I. (2014). Versatile genetic paintbrushes: Brainbow technologies. Wiley Interdiscip Rev Dev Biol. PubMed ID: 25491327

    Apitz, H. and Salecker, I. (2014). A challenge of numbers and diversity: neurogenesis in the Drosophila optic lobe. J Neurogenet: 1-35. PubMed ID: 24912777

    Shimosako, N., Hadjieconomou, D. and Salecker, I. (2014). Flybow to dissect circuit assembly in the Drosophila brain. Methods Mol Biol 1082: 57-69. PubMed ID: 24048926

    Oyallon, J., Apitz, H., Miguel-Aliaga, I., Timofeev, K., Ferreira, L. and Salecker, I. (2012). Regulation of locomotion and motoneuron trajectory selection and targeting by the Drosophila homolog of Olig family transcription factors. Dev Biol 369: 261-276. PubMed ID: 22796650

    Timofeev, K., Joly, W., Hadjieconomou, D. and Salecker, I. (2012). Localized netrins act as positional cues to control layer-specific targeting of photoreceptor axons in Drosophila. Neuron 75: 80-93. PubMed ID: 22794263

    Hadjieconomou, D., Timofeev, K. and Salecker, I. (2011). A step-by-step guide to visual circuit assembly in Drosophila. Curr Opin Neurobiol 21: 76-84. PubMed ID: 20800474

  • Maria Saleh Department of Virology, Institut Pasteur
    Mongelli, V., Lequime, S., Kousathanas, A., Gausson, V., Blanc, H., Nigg, J., Quintana-Murci, L., Elena, S. F. and Saleh, M. C. (2022). Innate immune pathways act synergistically to constrain RNA virus evolution in Drosophila melanogaster. Nat Ecol Evol. PubMed ID: 35273366

    Mondotte, J. A., Gausson, V., Frangeul, L., Suzuki, Y., Vazeille, M., Mongelli, V., Blanc, H., Failloux, A. B. and Saleh, M. C. (2020). Evidence For Long-Lasting Transgenerational Antiviral Immunity in Insects. Cell Rep 33(11): 108506. PubMed ID: 33326778

    Torri, A., Mongelli, V., Mondotte, J. A. and Saleh, M. C. (2020). Viral Infection and Stress Affect Protein Levels of Dicer 2 and Argonaute 2 in Drosophila melanogaster. Front Immunol 11: 362. PubMed ID: 32194567

    Mondotte, J. A., Gausson, V., Frangeul, L., Blanc, H., Lambrechts, L. and Saleh, M. C. (2018). Immune priming and clearance of orally acquired RNA viruses in Drosophila. Nat Microbiol. PubMed ID: 30374170

    Petit, M., Mongelli, V., Frangeul, L., Blanc, H., Jiggins, F. and Saleh, M. C. (2016). piRNA pathway is not required for antiviral defense in Drosophila melanogaster. Proc Natl Acad Sci U S A. PubMed ID: 27357659

    Karlikow, M., Goic, B., Mongelli, V., Salles, A., Schmitt, C., Bonne, I., Zurzolo, C. and Saleh, M. C. (2016). Drosophila cells use nanotube-like structures to transfer dsRNA and RNAi machinery between cells. Sci Rep 6: 27085. PubMed ID: 27255932

    van Mierlo, J. T., Overheul, G. J., Obadia, B., van Cleef, K. W., Webster, C. L., Saleh, M. C., Obbard, D. J. and van Rij, R. P. (2014). Novel Drosophila viruses encode host-specific suppressors of RNAi. PLoS Pathog 10: e1004256. PubMed ID: 25032815

  • Julia Saltz BioSciences, Rice University, Houston, Texas
    Girardeau, A. R., Foley, B. R. and Saltz, J. B. (2021). Comparing single- and mixed-species groups in fruit flies: differences in group dynamics, but not group formation. J Hered. PubMed ID: 34453172

    Wice, E. W. and Saltz, J. B. (2021). Selection on heritable social network positions is context-dependent in Drosophila melanogaster. Nat Commun 12(1): 3357. PubMed ID: 34099680

    Jaffe, A., Burns, M. P. and Saltz, J. B. (2020). Genotype-by-genotype epistasis for exploratory behaviour in D. simulans. Proc Biol Sci 287(1928): 20200057. PubMed ID: 32517624

    Burns, M. P., Cavallaro, F. D. and Saltz, J. B. (2020). Does Divergence in Habitat Breadth Associate with Species Differences in Decision Making in Drosophila Sechellia and Drosophila Simulans? Genes (Basel) 11(5). PubMed ID: 32397481

  • Paul Salvaterra City of Hope, Duarte, CA
    Ling, D., Magallanes, M. and Salvaterra, P. M. (2014). Accumulation of amyloid-like Abeta1-42 in autophagy-endosomal-lysosomal (AEL) vesicles: Potential implications for plaque biogenesis. ASN Neuro. PubMed ID: 24521233

    Ling, D. and Salvaterra, P. M. (2011). Brain aging and Abeta(1)(-)(4)(2) neurotoxicity converge via deterioration in autophagy-lysosomal system: a conditional Drosophila model linking Alzheimer's neurodegeneration with aging. Acta Neuropathol 121: 183-191. PubMed ID: 21076961

    Baumann, O., Salvaterra, P. M. and Takeyasu, K. (2010). Developmental changes in beta-subunit composition of Na,K-ATPase in the Drosophila eye. Cell Tissue Res 340: 215-228. PubMed ID: 20336468

  • Helen Salz Department of Genetics and Genome Sciences, Case Western Reserve, Cleveland
    Smolko, A. E., Shapiro-Kulnane, L. and Salz, H. K. (2020). An autoregulatory switch in sex-specific phf7 transcription causes loss of sexual identity and tumors in the Drosophila female germline. Development 147(17). PubMed ID: 32816970

    Smolko, A. E., Shapiro-Kulnane, L. and Salz, H. K. (2018). The H3K9 methyltransferase SETDB1 maintains female identity in Drosophila germ cells. Nat Commun 9(1): 4155. PubMed ID: 30297796

    Salz, H. K. (2012). Sex, stem cells and tumors in the Drosophila ovary. Fly (Austin) 7. PubMed ID: 23208193

    Chau, J., Kulnane, L. S. and Salz, H. K. (2012). Sex-lethal enables germline stem cell differentiation by down-regulating Nanos protein levels during Drosophila oogenesis. Proc Natl Acad Sci U S A 109: 9465-9470. PubMed ID: 22645327

    Johnson, M. L., Nagengast, A. A. and Salz, H. K. (2010). PPS, a large multidomain protein, functions with sex-lethal to regulate alternative splicing in Drosophila. PLoS Genet 6: e1000872. PubMed ID: 20221253

  • Adi Salzberg Faculty of Medicine, Technion, Haifa
    Avetisyan, A., Glatt, Y., Cohen, M., Timerman, Y., Aspis, N., Nachman, A., Halachmi, N., Preger-Ben Noon, E. and Salzberg, A. (2021). Delilah, prospero, and D-Pax2 constitute a gene regulatory network essential for the development of functional proprioceptors. Elife 10. PubMed ID: 34964712

    Avetisyan, A. and Salzberg, A. (2019). Accurate elimination of superfluous attachment cells is critical for the construction of functional multicellular proprioceptors in Drosophila. Cell Death Differ. PubMed ID: 30622305

    Greenblatt Ben-El, R. T., Hassan, A. and Salzberg, A. (2017). Loss of thrombospondin reveals a possible role for the extracellular matrix in chordotonal cap cell elongation. Int J Dev Biol 61(3-4-5): 311-318. PubMed ID: 28621428

    Halachmi, N., Nachman, A. and Salzberg, A. (2016). A newly identified type of attachment cell is critical for normal patterning of chordotonal neurons. Dev Biol. PubMed ID: 26794680

    Nachman, A., Halachmi, N., Matia, N., Manzur, D. and Salzberg, A. (2015). Deconstructing the complexity of regulating common properties in different cell types: lessons from the delilah gene. Dev Biol 403: 180-191. PubMed ID: 25989022

    Zohar-Stoopel, A., Gonen, N., Mahroum, M., Ben-Zvi, D. S., Toledano, H. and Salzberg, A. (2013). Homothorax plays autonomous and non-autonomous roles in proximodistal axis formation and migration of the Drosophila renal tubules. Dev Dyn. PubMed ID: 23821438

    Pechkovsky, A., Lahav, M., Bitman, E., Salzberg, A., Kleinberger, T. (2013) E4orf4 induces PP2A- and Src-dependent cell death in Drosophila melanogaster and at the same time inhibits classic apoptosis pathways. Proc Natl Acad Sci U S A. PubMed ID: 23613593

    Halachmi, N., Nachman, A. and Salzberg, A. (2012). Visualization of proprioceptors in Drosophila larvae and pupae. J Vis Exp: e3846. PubMed ID: 22733157

    Egoz-Matia, N., Nachman, A., Halachmi, N., Toder, M., Klein, Y. and Salzberg, A. (2011). Spatial regulation of cell adhesion in the Drosophila wing is mediated by Delilah, a potent activator of betaPS integrin expression. Dev Biol 351: 99-109. PubMed ID: 21215259

  • Christos Samakovlis The Wenner-Gren Institute Developmental Biology, Stockholm University
    Yao, L., Wang, S., Orzechowski-Westholm, J., 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. PubMed ID: 28760809

    Hosono, C., Matsuda, R., Adryan, B. and Samakovlis, C. (2015). Transient junction anisotropies orient annular cell polarization in the Drosophila airway tubes. Nat Cell Biol 17: 1569-1576. PubMed ID: 26551273

    Matsuda, R., Hosono, C., Saigo, K. and Samakovlis, C. (2015). The intersection of the extrinsic Hedgehog and WNT/Wingless signals with the intrinsic Hox code underpins branching pattern and tube shape diversity in the drosophila Airways. PLoS Genet 11: e1004929. PubMed ID: 25615601

    Tsarouhas, V., Yao, L. and Samakovlis, C. (2014). Src-kinases and ERK activate distinct responses to Stitcher receptor tyrosine kinase signaling during wound healing in Drosophila. J Cell Sci. PubMed ID: 24522188

    O'Farrell, F., Wang, S., Katheder, N., Rusten, T. E. and Samakovlis, C. (2013). Two-tiered control of epithelial growth and autophagy by the insulin receptor and the ret-like receptor, stitcher. PLoS Biol 11: e1001612. PubMed ID: 23935447

    Tiklova, K., Tsarouhas, V. and Samakovlis, C. (2013). Control of airway tube diameter and integrity by secreted chitin-binding proteins in Drosophila. PLoS One 8: e67415. PubMed ID: 23826295

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

    Tiklova, K., Senti, K. A., Wang, S., Graslund, A. and Samakovlis, C. (2010). Epithelial septate junction assembly relies on melanotransferrin iron binding and endocytosis in Drosophila. Nat Cell Biol 12: 1071-1077. PubMed ID: 20935638

    Wagner, N., Weyhersmuller, A., Blauth, A., Schuhmann, T., Heckmann, M., Krohne, G. and Samakovlis, C. (2010). The Drosophila LEM-domain protein MAN1 antagonizes BMP signaling at the neuromuscular junction and the wing crossveins. Dev Biol 339: 1-13. PubMed ID: 20036230

  • Maria Samsonova Polytechnical University, St. Petersburg
    Surkova, S., Sokolkova, A., Kozlov, K., Nuzhdin, S. V. and Samsonova, M. (2019). Quantitative analysis reveals genotype- and domain- specific differences between mRNA and protein expression of segmentation genes in Drosophila. Dev Biol 448(1): 48-58. PubMed ID: 30629954

    Surkova, S., Sokolkova, A., Kozlov, K., Nuzhdin, S. V. and Samsonova, M. (2019). Quantitative analysis reveals genotype- and domain- specific differences between mRNA and protein expression of segmentation genes in Drosophila. Dev Biol. PubMed ID: 30629954

    Gursky, V. V., Kozlov, K. N., Kulakovskiy, I. V., Zubair, A., Marjoram, P., Lawrie, D. S., Nuzhdin, S. V. and Samsonova, M. G. (2017). Translating natural genetic variation to gene expression in a computational model of the Drosophila gap gene regulatory network. PLoS One 12(9): e0184657. PubMed ID: 28898266

    Kozlov, K., Gursky, V., Kulakovskiy, I. and Samsonova, M. (2014). Sequence-based model of gap gene regulatory network. BMC Genomics 15 Suppl 12: S6. PubMed ID: 25564104

    Surkova, S., Myasnikova, E., Kozlov, K. N., Pisarev, A., Reinitz, J. and Samsonova, M. (2013). Quantitative imaging of gene expression in Drosophila embryos. Cold Spring Harb Protoc 2013. PubMed ID: 23734022

    Surkova, S., Golubkova, E., Manu, Panok, L., Mamon, L., Reinitz, J. and Samsonova, M. (2013). Quantitative dynamics and increased variability of segmentation gene expression in the Drosophila Kruppel and knirps mutants. Dev Biol. PubMed ID: 23333947

    Janssens, H., Crombach, A., Richard Wotton, K., Cicin-Sain, D., Surkova, S., Lu Lim, C., Samsonova, M., Akam, M. and Jaeger, J. (2013). Lack of tailless Leads to an Increase in Expression Variability in Drosophila Embryos. Dev Biol. PubMed ID: 23333944

    Kozlov, K., Surkova, S., Myasnikova, E., Reinitz, J. and Samsonova, M. (2012). Modeling of gap gene expression in Drosophila Kruppel mutants. PLoS Comput Biol 8: e1002635. PubMed ID: 22927803

  • Aravinthan Samuel Dept. of Physics & Center for Brain Science, Harvard
    Hernandez-Nunez, L., Chen, A., Budelli, G., Berck, M. E., Richter, V., Rist, A., Thum, A. S., Cardona, A., Klein, M., Garrity, P. and Samuel, A. D. T. (2021). Synchronous and opponent thermosensors use flexible cross-inhibition to orchestrate thermal homeostasis. Sci Adv 7(35). PubMed ID: 34452914

    Vogt, K., Zimmerman, D. M., Schlichting, M., Hernandez-Nunez, L., Qin, S., Malacon, K., Rosbash, M., Pehlevan, C., Cardona, A. and Samuel, A. D. T. (2021). Internal state configures olfactory behavior and early sensory processing in Drosophila larvae. Sci Adv 7(1). PubMed ID: 33523854

    Si, G., Kanwal, J. K., Hu, Y., Tabone, C. J., Baron, J., Berck, M., Vignoud, G. and Samuel, A. D. T. (2019). Structured odorant response patterns across a complete olfactory receptor neuron population. Neuron. PubMed ID: 30683545

    He, L., Si, G., Huang, J., Samuel, A. D. T. and Perrimon, N. (2018). Mechanical regulation of stem-cell differentiation by the stretch-activated Piezo channel. Nature 555(7694): 103-106. PubMed ID: 29414942

    Berck, M.E., Khandelwal, A., Claus, L., Hernandez-Nunez, L., Si, G., Tabone, C.J., Li, F., Truman, J.W., Fetter, R.D., Louis, M., Samuel, A.D. and Cardona, A. (2016). The wiring diagram of a glomerular olfactory system. Elife [Epub ahead of print]. PubMed ID: 27177418

    van Giesen, L., Hernandez-Nunez, L., Delasoie-Baranek, S., Colombo, M., Renaud, P., Bruggmann, R., Benton, R., Samuel, A.D. and Sprecher, S.G. (2016). Multimodal stimulus coding by a gustatory sensory neuron in Drosophila larvae. Nat Commun 7: 10687. PubMed ID: 26864722

    Hernandez-Nunez, L., Belina, J., Klein, M., Si, G., Claus, L., Carlson, J. R. and Samuel, A. D. (2015). Reverse-correlation analysis of navigation dynamics in larva using optogenetics. Elife 4. PubMed ID: 25942453

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

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

    Gershow, M., Berck, M., Mathew, D., Luo, L., Kane, E. A., Carlson, J. R. and Samuel, A. D. (2012). Controlling airborne cues to study small animal navigation. Nat Methods 9: 290-296. PubMed ID: 22245808

  • Aziz Sancar University of North Carolina at Chapel Hill School of Medicine
    Deger, N., Cao, X., Selby, C. P., Gulec, S., Kawara, H., Dewey, E. B., Wang, L., Yang, Y., Archibald, S., Selcuk, B., Adebali, O., Sekelsky, J., Sancar, A. and Liu, Z. (2022). CSB-independent, XPC-dependent transcription-coupled repair in Drosophila. Proc Natl Acad Sci U S A 119(9). PubMed ID: 35217627

    Deger, N., Yang, Y., Lindsey-Boltz, L. A., Sancar, A. and Selby, C. P. (2019). Drosophila, which lacks canonical transcription-coupled repair proteins, performs transcription-coupled repair. J Biol Chem. PubMed ID: 31624146

    Ozturk, N., Selby, C. P., Zhong, D. and Sancar, A. (2013). Mechanism of Photosignaling by Drosophila Cryptochrome:Role of the Redox Status of the Flavin Chromophore. J Biol Chem. PubMed ID: 24379403

    Ozturk, N., Vanvickle-Chavez, S. J., Akileswaran, L., Van Gelder, R. N., Sancar, A. (2013) Ramshackle (Brwd3) promotes light-induced ubiquitylation of Drosophila Cryptochrome by DDB1-CUL4-ROC1 E3 ligase complex. Proc Natl Acad Sci U S A. PubMed ID: 23479607

    Selby, C. P. and Sancar, A. (2012). The second chromophore in Drosophila photolyase/cryptochrome family photoreceptors. Biochemistry 51: 167-171. PubMed ID: 22175817

    Ozturk, N., Selby, C. P., Annayev, Y., Zhong, D. and Sancar, A. (2011). Reaction mechanism of Drosophila cryptochrome. Proc Natl Acad Sci U S A 108: 516-521. PubMed ID: 21187431

    Li, J., Liu, Z., Tan, C., Guo, X., Wang, L., Sancar, A. and Zhong, D. (2010). Dynamics and mechanism of repair of ultraviolet-induced (6-4) photoproduct by photolyase. Nature 466: 887-890. PubMed ID: 20657578

  • Ernesto Sánchez-Herrero Centro de Biología Molecular 'Severo Ochoa', Madrid
    Romero-Pozuelo, J., Foronda, D., Martin, P., Hudry, B., Merabet, S., Graba, Y. and Sanchez-Herrero, E. (2019). Cooperation of axial and sex specific information controls Drosophila female genitalia growth by regulating the Decapentaplegic pathway. Dev Biol. PubMed ID: 31251896

    De Las Heras, J. M., Garcia-Cortes, C., Foronda, D., Pastor-Pareja, J. C., Shashidhara, L. S. and Sanchez-Herrero, E. (2018). The Drosophila Hox gene Ultrabithorax controls appendage shape by regulating extracellular matrix dynamics. Development. PubMed ID: 29853618

    Singh, S., Sanchez-Herrero, E. and Shashidhara, L. S. (2015). Critical role for Fat/Hippo and IIS/Akt pathways downstream of Ultrabithorax during haltere specification in Drosophila. Mech Dev. PubMed ID: 26299254

    Foronda, D., Curt, J. R., Prieto, N., Martin, P. and Sanchez-Herrero, E. (2015). The elimination of an adult segment by the Hox gene Abdominal-B. Mech Dev [Epub ahead of print]. PubMed ID: 26259679

    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

    de Navas, L., Foronda, D., Del Saz, D. and Sanchez-Herrero, E. (2014). A genetic strategy to obtain p-Gal4 elements in the Drosophila Hox genes. Methods Mol Biol 1196: 49-57. PubMed ID: 25151157

    Garaulet, D. L., Castellanos, M. C., Bejarano, F., Sanfilippo, P., Tyler, D. M., Allan, D. W., Sanchez-Herrero, E. and Lai, E. C. (2014). Homeotic Function of Drosophila Bithorax-Complex miRNAs Mediates Fertility by Restricting Multiple Hox Genes and TALE Cofactors in the CNS. Dev Cell. PubMed ID: 24909902

    Guarner, A., Manjon, C., Edwards, K., Steller, H., Suzanne, M. and Sanchez-Herrero, E. (2013). The zinc finger homeodomain-2 gene of Drosophila controls Notch targets and regulates apoptosis in the tarsal segments. Dev Biol. PubMed ID: 24144920

    Curt, J. R., de Navas, L. F., Sanchez-Herrero, E. (2013) Differential activity of Drosophila hox genes induces Myosin expression and can maintain compartment boundaries. PLoS One 8: e57159. Pubmed ID: 23451173

    Foronda, D., Martín, P., Sánnchez-Herrero, E. (2012). Drosophila Hox and sex-determination genes control segment elimination through EGFR and extramacrochetae activity. PLoS Genet. 8(8): e1002874. PubMed Citation: 22912593

  • Natalia Sanchez-Soriano Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool
    Ros, O., Barrecheguren, P. J., Cotrufo, T., Schaettin, M., Rosello-Busquets, C., Vilchez-Acosta, A., Hernaiz-Llorens, M., Martinez-Marmol, R., Ulloa, F., Stoeckli, E. T., Araujo, S. J. and Soriano, E. (2018). A conserved role for Syntaxin-1 in pre- and post-commissural midline axonal guidance in fly, chick, and mouse. PLoS Genet 14(6): e1007432. PubMed ID: 29912942

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

    Zschatzsch, M., Oliva, C., Langen, M., De Geest, N., Ozel, M. N., Williamson, W. R., Lemon, W. C., Soldano, A., Munck, S., Hiesinger, P. R., Sanchez-Soriano, N. and Hassan, B. A. (2014). Regulation of branching dynamics by axon-intrinsic asymmetries in Tyrosine Kinase Receptor signaling. Elife 3: e01699. PubMed ID: 24755286

    Stephan, D., Sanchez-Soriano, N., Loschek, L. F., Gerhards, R., Gutmann, S., Storchova, Z., Prokop, A. and Kadow, I. C. (2012). Drosophila Psidin regulates olfactory neuron number and axon targeting through two distinct molecular mechanisms. J Neurosci 32: 16080-16094. PubMed ID: 23152593

  • Tzu-Kang Sang Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
    Chang, Y. C., Peng, Y. X., Yu, B. H., Chang, H. C., Liang, P. S., Huang, T. Y., Shih, C. J., Chu, L. A. and Sang, T. K. (2021). VCP maintains nuclear size by regulating the DNA damage-associated MDC1-p53-autophagy axis in Drosophila. Nat Commun 12(1): 4258. PubMed ID: 34253734

    Liang, C. J., Chang, Y. C., Chang, H. C., Wang, C. K., Hung, Y. C., Lin, Y. E., Chan, C. C., Chen, C. H., Chang, H. Y. and Sang, T. K. (2014). Derlin-1 Regulates Mutant VCP-Linked Pathogenesis and Endoplasmic Reticulum Stress-Induced Apoptosis. PLoS Genet 10: e1004675. PubMed ID: 25255315

    Santhanam, A., Peng, W. H., Yu, Y. T., Sang, T. K., Chen, G. C. and Meng, T. C. (2014). Ecdysone-induced receptor tyrosine phosphatase PTP52F regulates Drosophila midgut histolysis by enhancement of autophagy and apoptosis. Mol Cell Biol 34: 1594-1606. PubMed ID: 24550005

    Chan, H. T., Lee, T. R., Huang, S. H., Lee, H. Y., Sang, T. K., Chan, H. L. and Lyu, P. C. (2012). Proteomic analysis of a drosophila IBMPFD model reveals potential pathogenic mechanisms. Mol Biosyst 8: 1730-1741. PubMed ID: 22481368

  • Benedicte Sanson Department of Physiology, Development and Neuroscience, University of Cambridge
    Finegan, T. M., Hervieux, N., Nestor-Bergmann, A., Fletcher, A. G., Blanchard, G. B. and Sanson, B. (2019). The tricellular vertex-specific adhesion molecule Sidekick facilitates polarised cell intercalation during Drosophila axis extension. PLoS Biol 17(12): e3000522. PubMed ID: 31805038

    Johansson, J., Naszai, M., Hodder, M. C., Pickering, K. A., Miller, B. W., Ridgway, R. A., Yu, Y., Peschard, P., Brachmann, S., Campbell, A. D., Cordero, J. B. and Sansom, O. J. (2019). RAL GTPases drive intestinal stem cell function and regeneration through internalization of WNT signalosomes. Cell Stem Cell 24(4): 592-607.e597. PubMed ID: 30853556

    Scarpa, E., Finet, C., Blanchard, G. B. and Sanson, B. (2018). Actomyosin-driven tension at compartmental boundaries orients cell division independently of cell geometry in vivo. Dev Cell. PubMed ID: 30503752

    Urbano, J. M., Naylor, H. W., Scarpa, E., Muresan, L. and Sanson, B. (2018). Suppression of epithelial folding at actomyosin-enriched compartment boundaries downstream of Wingless signalling in Drosophila. Development 145(8). PubMed ID: 29691225

    Tetley, R. J., Blanchard, G. B., Fletcher, A. G., Adams, R. J. and Sanson, B. (2016). Unipolar distributions of junctional Myosin II identify cell stripe boundaries that drive cell intercalation throughout axis extension. Elife 5 [Epub ahead of print]. PubMed ID: 27183005

    Lye, C. M., Blanchard, G. B., Naylor, H. W., Muresan, L., Huisken, J., Adams, R. J. and Sanson, B. (2015). Mechanical coupling between endoderm invagination and axis extension in Drosophila. PLoS Biol 13: e1002292. PubMed ID: 26544693

    Lye, C. M., Naylor, H. W. and Sanson, B. (2014). Subcellular localisations of the CPTI collection of YFP-tagged proteins in Drosophila embryos. Development 141: 4006-4017. PubMed ID: 25294944

    Lowe, N., et al. (2014). Analysis of the expression patterns, subcellular localisations and interaction partners of Drosophila proteins using a pigP protein trap library. Development 141: 3994-4005. PubMed ID: 25294943

    Lye, C. M. and Sanson, B. (2011). Tension and epithelial morphogenesis in Drosophila early embryos. Curr Top Dev Biol 95: 145-187. PubMed ID: 21501751

  • Subhabrata Sanyal Senior Scientist, Neuroscience at CalicoLabs, South San Francisco, California
    Ryan, S. M., Almassey, M., Burch, A. M., Ngo, G., Martin, J. M., Myers, D., Compton, D., Archie, S., Cross, M., Naeger, L., Salzman, A., Virola-Iarussi, A., Barbee, S. A., Mortimer, N. T., Sanyal, S. and Vrailas-Mortimer, A. D. (2021). Drosophila p38 MAPK interacts with BAG-3/starvin to regulate age-dependent protein homeostasis. Aging Cell 20(11): e13481. PubMed ID: 34674371

    Privman Champaloux, E., Donelson, N., Pyakurel, P., Wolin, D., Ostendorf, L., Denno, M., Borman, R., Burke, C., Short-Miller, J. C., Yoder, M. R., Copeland, J. M., Sanyal, S. and Jill Venton, B. (2020). Ring Finger Protein 11 (RNF11) Modulates Dopamine Release in Drosophila. Neuroscience. PubMed ID: 33176188

    Donelson, N. C. and Sanyal, S. (2015). Use of Drosophila in the investigation of sleep disorders. Exp Neurol [Epub ahead of print]. PubMed ID: 26160555

    Gokhale, A., Mullin, A. P., Zlatic, S. A., Easley, C. A. t., Merritt, M. E., Raj, N., Larimore, J., Gordon, D. E., Peden, A. A., Sanyal, S. and Faundez, V. (2015). The N-ethylmaleimide-sensitive factor and dysbindin interact to modulate synaptic plasticity. J Neurosci 35: 7643-7653. PubMed ID: 25972187

    Mullin, A. P., Sadanandappa, M. K., Ma, W., Dickman, D. K., VijayRaghavan, K., Ramaswami, M., Sanyal, S. and Faundez, V. (2015). Gene dosage in the dysbindin schizophrenia susceptibility network differentially affect synaptic function and plasticity. J Neurosci 35: 325-338. PubMed ID: 25568125

    Vrailas-Mortimer, A. D., Ryan, S. M., Avey, M. J., Mortimer, N. T., Dowse, H. and Sanyal, S. (2014). p38 MAP Kinase Regulates Circadian Rhythms in Drosophila. J Biol Rhythms. PubMed ID: 25403440

    Freeman, A. A., Syed, S. and Sanyal, S. (2013). Modeling the genetic basis for human sleep disorders in Drosophila. Commun Integr Biol 6: e22733. PubMed ID: 23802043

    Freeman, A. A., Mandilaras, K., Missirlis, F., Sanyal, S. (2013) An emerging role for Cullin-3 mediated ubiquitination in sleep and circadian rhythm: Insights from Drosophila. Fly (Austin) 7. PubMed ID: 23455037

    Vonhoff, F., Kuehn, C., Blumenstock, S., Sanyal, S. and Duch, C. (2013). Temporal coherency between receptor expression, neural activity and AP-1-dependent transcription regulates Drosophila motoneuron dendrite development. Development 140: 606-616. PubMed ID: 23293292

    Timmerman, C. and Sanyal, S. (2012). Behavioral and electrophysiological outcomes of tissue-specific Smn knockdown in Drosophila melanogaster. Brain Res 1489: 66-80. PubMed ID: 23103409

  • Alberto Sanz University of Tampere, Finland
    Scialo, F., Sriram, A., Stefanatos, R., Spriggs, R. V., Loh, S. H. Y., Martins, L. M. and Sanz, A. (2020). Mitochondrial complex I derived ROS regulate stress adaptation in Drosophila melanogaster. Redox Biol: 101450. PubMed ID: 32146156

    Scialo, F., Sriram, A., Naudí, A., Ayala, V., Jové, M., Pamplona, R. and Sanz, A. (2015). Target of rapamycin activation predicts lifespan in fruit flies. Cell Cycle [Epub ahead of print]. PubMed ID: 26259964

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

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

    Stefanatos, R., Sriram, A., Kiviranta, E., Mohan, A., Ayala, V., Jacobs, H. T., Pamplona, R. and Sanz, A. (2012). dj-1beta regulates oxidative stress, insulin-like signaling and development in Drosophila melanogaster. Cell Cycle 11: 3876-3886. PubMed ID: 22983063

  • Surajit Sarkar Neurobiology and Developmental Genetics, University of Delhi South Campus
    Tandon, S. and Sarkar, S. (2021). The S6k/4E-BP mediated growth promoting sub-pathway of insulin signalling cascade is essential to restrict pathogenesis of poly(Q) disorders in Drosophila. Life Sci 275: 119358. PubMed ID: 33744321

    Nisha and Sarkar, S. (2021). Downregulation of glob1 suppresses pathogenesis of human neuronal tauopathies in Drosophila by regulating tau phosphorylation and ROS generation. Neurochem Int 146: 105040. PubMed ID: 33865914

    Pragati and Sarkar, S. (2021). Shaggy functions downstream of dMyc and their concurrent downregulation confers additive rescue against tau toxicity in

    Aqsa and Sarkar, S. (2020). Age dependent trans-cellular propagation of human tau aggregates in Drosophila disease models. Brain Res: 147207. PubMed ID: 33212022

    Pragati, Chanu, S. I. and Sarkar, S. (2019). Reduced expression of dMyc mitigates Tau(V337M) mediated neurotoxicity by preventing the Tau hyperphosphorylation and inducing autophagy in Drosophila. Neurosci Lett: 134622. PubMed ID: 31715291

    Nisha, Aggarwal, P. and Sarkar, S. (2019). Adequate expression of Globin1 is required for development and maintenance of nervous system in Drosophila. Mol Cell Neurosci 100: 103398. PubMed ID: 31472221

    Raj, K. and Sarkar, S. (2018). Tissue-Specific Upregulation of Drosophila Insulin Receptor (InR) Mitigates Poly(Q)-Mediated Neurotoxicity by Restoration of Cellular Transcription Machinery. Mol Neurobiol. PubMed ID: 29881950

    Yadav, R., Nisha and Sarkar, S. (2018). Drosophila globin1 is required for maintenance of the integrity of F-actin based cytoskeleton during development. Exp Cell Res. PubMed ID: 29524391

    Chanu, S. I. and Sarkar, S. (2017). Targeted downregulation of dMyc restricts neurofibrillary tangles mediated pathogenesis of human neuronal tauopathies in Drosophila. Biochim Biophys Acta [Epub ahead of print]. PubMed ID: 28529046

    Yadav, R. and Sarkar, S. (2016). Drosophila glob1 is required for the maintenance of cytoskeletal integrity during oogenesis. Dev Dyn [Epub ahead of print]. PubMed ID: 27503269

    Chanu, S. I. and Sarkar, S. (2016). Targeted Downregulation of dMyc Suppresses Pathogenesis of Human Neuronal Tauopathies in Drosophila by Limiting Heterochromatin Relaxation and Tau Hyperphosphorylation. Mol Neurobiol. PubMed ID: 27000837

  • Miklós Sass Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest
    Billes, V., Kovacs, T., Manzeger, A., Lorincz, P., Szincsak, S., Regos, A., Kulcsar, P. I., Korcsmaros, T., Lukacsovich, T., Hoffmann, G., Erdelyi, M., Mihaly, J., Takacs-Vellai, K., Sass, M. and Vellai, T. (2018). Developmentally regulated autophagy is required for eye formation in Drosophila. Autophagy. PubMed ID: 29940806

    Maruzs, T., Lorincz, P., Szatmári, Z., Széplaki, S., Sándor, Z., Lakatos, Z., Puska, G., Juhász, G. and Sass, M. (2015). Retromer ensures the degradation of autophagic cargo via maintaining lysosome function in Drosophila. Traffic [Epub ahead of print]. PubMed ID: 26172538

    Kis, V, Barti, B., Lippai, M. and Sass, M. (2015). Specialized cortex glial cells accumulate lipid droplets in Drosophila melanogaster. PLoS One 10: e0131250. PubMed ID: 26148013

    Lorincz, P., Lakatos, Z., Maruzs, T., Szatmari, Z., Kis, V. and Sass, M. (2014). Atg6/UVRAG/Vps34-Containing Lipid Kinase Complex Is Required for Receptor Downregulation through Endolysosomal Degradation and Epithelial Polarity during Drosophila Wing Development. Biomed Res Int 2014: 851349. PubMed ID: 25006588

    Szatmari, Z., Kis, V., Lippai, M., Hegedus, K., Farago, T., Lorincz, P., Tanaka, T., Juhasz, G. and Sass, M. (2013). Rab11 facilitates crosstalk between autophagy and endosomal pathway through regulation of Hook localization. Mol Biol Cell. PubMed ID: 24356450

    Low, P., Varga, A., Pircs, K., Nagy, P., Szatmari, Z., Sass, M. and Juhasz, G. (2013). Impaired proteasomal degradation enhances autophagy via hypoxia signaling in Drosophila. BMC Cell Biol 14: 29. PubMed ID: 23800266

  • Makoto Sato Kanazawa University
    Hayashi, T., Tomomizu, T., Sushida, T., Akiyama, M., Ei, S. I. and Sato, M. (2022). Tiling mechanisms of the Drosophila compound eye through geometrical tessellation. Curr Biol. PubMed ID: 35390281

    Yasugi, T. and Sato, M. (2022). Mathematical modeling of Notch dynamics in Drosophila neural development. Fly (Austin) 16(1): 24-36. PubMed ID: 34609265

    Wang, M., Han, X., Liu, C., Takayama, R., Yasugi, T., Ei, S. I., Nagayama, M., Tanaka, Y. and Sato, M. (2021). Intracellular trafficking of Notch orchestrates temporal dynamics of Notch activity in the fly brain. Nat Commun 12(1): 2083. PubMed ID: 33828096

    Han, X., Wang, M., Liu, C., Trush, O., Takayama, R., Akiyama, T., Naito, T., Tomomizu, T., Imamura, K. and Sato, M. (2020). DWnt4 and DWnt10 Regulate Morphogenesis and Arrangement of Columnar Units via Fz2/PCP Signaling in the Drosophila Brain. Cell Rep 33(4): 108305. PubMed ID: 33113378

    Liu, C., Trush, O., Han, X., Wang, M., Takayama, R., Yasugi, T., Hayashi, T. and Sato, M. (2020). Dscam1 establishes the columnar units through lineage-dependent repulsion between sister neurons in the fly brain. Nat Commun 11(1): 4067. PubMed ID: 32792493

    Suzuki, T., Liu, C., Kato, S., Nishimura, K., Takechi, H., Yasugi, T., Takayama, R., Hakeda-Suzuki, S., Suzuki, T. and Sato, M. (2018). Netrin signaling defines the regional border in the Drosophila visual center. iScience 8: 148-160. PubMed ID: 30316037

    Suzuki, T., Trush, O., Yasugi, T., Takayama, R. and Sato, M. (2016). Wnt signaling specifies anteroposterior progenitor zone identity in the Drosophila visual center. J Neurosci 36: 6503-6513. PubMed ID: 27307238

    Suzuki, T., Hasegawa, E., Nakai, Y., Kaido, M., Takayama, R. and Sato, M. (2016). Formation of neuronal circuits by interactions between neuronal populations derived from different origins in the Drosophila visual center. Cell Rep [Epub ahead of print]. PubMed ID: 27068458

    Suzuki, T. and Sato, M. (2014). Neurogenesis and neuronal circuit formation in the Drosophila visual center. Dev Growth Differ. PubMed ID: 25200311

    Suzuki, T., Kaido, M., Takayama, R., Sato, M. (2013) A temporal mechanism that produces neuronal diversity in the Drosophila visual center. Dev Biol. PubMed ID: 23665475

  • Akiko Satoh Graduate School of Integrated Arts and Sciences, Hiroshima University
    Fujii, S., Kurokawa, K., Tago, T., Inaba, R., Takiguchi, A., Nakano, A., Satoh, T. and Satoh, A. K. (2020). Sec71 separates Golgi stacks in Drosophila S2 cells. J Cell Sci 133(24). PubMed ID: 33262309

    Hiramatsu, N., Tago, T., Satoh, T. and Satoh, A. K. (2019). ER membrane protein complex is required for the insertions of late-synthesized transmembrane helices of Rh1 in Drosophila photoreceptors. Mol Biol Cell: mbcE19080434. PubMed ID: 31553680

    Otsuka, Y., Satoh, T., Nakayama, N., Inaba, R., Yamashita, H. and Satoh, A. K. (2019). Parcas is the predominant Rab11GEF for rhodopsin transport in Drosophila photoreceptors. J Cell Sci. PubMed ID: 31296556

    Ogi, S., Matsuda, A., Otsuka, Y., Liu, Z., Satoh, T. and Satoh, A. K. (2019). Syndapin constricts microvillar necks to form a united rhabdomere in Drosophila photoreceptors. Development. PubMed ID: 31371377

    Satoh, T., Nakamura, Y. and Satoh, A. K. (2016). The roles of Syx5 in Golgi morphology and rhodopsin transport in Drosophila photoreceptors. Biol Open [Epub ahead of print]. PubMed ID: 27591190

    Satoh, T., Nakamura, Y. and Satoh, A. K. (2016). Rab6 functions in polarized transport in Drosophila photoreceptors. Fly (Austin): 0. PubMed ID: 27116570

    Iwanami, N., Nakamura, Y., Satoh, T., Liu, Z. and Satoh, A. K. (2016). Rab6 Is Required for Multiple Apical Transport Pathways but Not the Basolateral Transport Pathway in Drosophila Photoreceptors. PLoS Genet 12: e1005828. PubMed ID: 26890939

    Satoh, T., Ohba, A., Liu, Z., Inagaki, T. and Satoh, A. K. (2015). dPob/EMC is essential for biosynthesis of rhodopsin and other multi-pass membrane proteins in Drosophila photoreceptors. Elife 4. PubMed ID: 25715730

  • Harald Saumweber Institute of Biology, Humboldt-Universität zu Berlin
    Jammrath, J., Reim, I. and Saumweber, H. (2020). Cbl-Associated Protein CAP contributes to correct formation and robust function of the Drosophila heart tube. PLoS One 15(5): e0233719. PubMed ID: 32469960

    Zielke, T., Glotov, A. and Saumweber, H. (2015). High-resolution in situ hybridization analysis on the chromosomal interval 61C7-61C8 of Drosophila melanogaster reveals interbands as open chromatin domains. Chromosoma. PubMed ID: 26520107

    Zielke, T. and Saumweber, H. (2014). Dissection of open chromatin domain formation by site specific recombination in Drosophila. J Cell Sci [Epub ahead of print]. PubMed ID: 24639466

    Hofmann, A., Brunner, M., Schwendemann, A., Strodicke, M., Karberg, S., Klebes, A., Saumweber, H. and Korge, G. (2010). The winged-helix transcription factor JUMU regulates development, nucleolus morphology and function, and chromatin organization of Drosophila melanogaster. Chromosome Res 18: 307-324. PubMed ID: 20213139

    Schirling, C., Heseding, C., Heise, F., Kesper, D., Klebes, A., Klein-Hitpass, L., Vortkamp, A., Hoffmann, D., Saumweber, H. and Ehrenhofer-Murray, A. E. (2010). Widespread regulation of gene expression in the Drosophila genome by the histone acetyltransferase dTip60. Chromosoma 119: 99-113. PubMed ID: 19949809

  • Timothy Saunders Mechanobiology Institute, National University of Singapore
    Yadav, V., Tolwinski, N. and Saunders, T. E. (2021). Spatiotemporal sensitivity of mesoderm specification to FGFR signalling in the Drosophila embryo. Sci Rep 11(1): 14091. PubMed ID: 34238963

    Zhang, S., Teng, X., Toyama, Y. and Saunders, T. E. (2020). Periodic Oscillations of Myosin-II Mechanically Proofread Cell-Cell Connections to Ensure Robust Formation of the Cardiac Vessel. Curr Biol. PubMed ID: 32679105

    Zhang, S., Amourda, C., Garfield, D. and Saunders, T. E. (2018). Selective Filopodia Adhesion Ensures Robust Cell Matching in the Drosophila Heart. Dev Cell 46(2): 189-203 e184. PubMed ID: 30016621

    Chong, J., Amourda, C. and Saunders, T. E. (2018). Temporal development of Drosophila embryos is highly robust across a wide temperature range. J R Soc Interface 15(144). PubMed ID: 29997261

    Huang, A., Amourda, C., Zhang, S., Tolwinski, N. S. and Saunders, T. E. (2017). Decoding temporal interpretation of the morphogen Bicoid in the early Drosophila embryo. Elife 6. PubMed ID: 28691901

  • Kyoich Sawamura Faculty of Life and Environmental Sciences, University of Tsukuba
    Hirai, K., Wang, Z., Miura, K., Hayashi, T., Awasaki, T., Wada, M., Keira, Y., Ishikawa, H. O. and Sawamura, K. (2018). Genetic Analyses of Elys Mutations in Drosophila Show Maternal-Effect Lethality and Interactions with Nucleoporin Genes. G3 (Bethesda). PubMed ID: 29773558

    Shirata, M., Araye, Q., Maehara, K., Enya, S., Takano-Shimizu, T. and Sawamura, K. (2013). Allelic asymmetry of the Lethal hybrid rescue (Lhr) gene expression in the hybrid between Drosophila melanogaster and D. simulans: confirmation by using genetic variations of D. melanogaster. Genetica. PubMed ID: 24379167

    Araye, Q. and Sawamura, K. (2013). Genetic decay of balancer chromosomes in Drosophila melanogaster. Fly (Austin) 7: 184-186. PubMed ID: 23648996

    Maehara, K., Murata, T., Aoyama, N., Matsuno, K. and Sawamura, K. (2012). Genetic dissection of Nucleoporin 160 (Nup160), a gene involved in multiple phenotypes of reproductive isolation in Drosophila. Genes Genet Syst 87: 99-106. PubMed ID: 22820383

    Wen, S. Y., Yamada, H., Li, Y. F., Kimura, M. T., Oguma, Y., Sawamura, K. and Toda, M. J. (2011). Copulatory courtship behavior and sine song as a mate recognition cue in Drosophila lini and its sibling species. Zoolog Sci 28: 469-475. PubMed ID: 21728794

  • William Saxton Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz
    Lim, A., Rechtsteiner, A. and Saxton, W. M. (2017). Two kinesins drive anterograde neuropeptide transport. Mol Biol Cell [Epub ahead of print]. PubMed ID: 28904207

    Monteith, C. E., Brunner, M. E., Djagaeva, I., Bielecki, A. M., Deutsch, J. M. and Saxton, W. M. (2016). A mechanism for cytoplasmic streaming: Kinesin-driven alignment of microtubules and fast fluid flows. Biophys J 110: 2053-2065. PubMed ID: 27166813

    Djagaeva, I., Rose, D. J., Lim, A., Venter, C. E., Brendza, K. M., Moua, P. and Saxton, W. M. (2012). Three routes to suppression of the neurodegenerative phenotypes caused by kinesin heavy chain mutations. Genetics 192: 173-183. PubMed ID: 22714410

    Saxton, W. M. and Hollenbeck, P. J. (2012). The axonal transport of mitochondria. J Cell Sci 125: 2095-2104. PubMed ID: 22619228

    Casper-Lindley, C., Kimura, S., Saxton, D. S., Essaw, Y., Simpson, I., Tan, V. and Sullivan, W. (2011). Rapid fluorescence-based screening for Wolbachia endosymbionts in Drosophila germ line and somatic tissues. Appl Environ Microbiol 77: 4788-4794. PubMed ID: 21622788

  • Matthias Schaefer Deutsches Krebsforschungszentrum. Division of Epigenetics, Heidelberg, Germany
    Durdevic, Z., Mobin, M. B., Hanna, K., Lyko, F. and Schaefer, M. (2013). The RNA Methyltransferase Dnmt2 Is Required for Efficient Dicer-2-Dependent siRNA Pathway Activity in Drosophila. Cell Rep. PubMed ID: 24012760

    Durdevic, Z., Mobin, M. B., Hanna, K., Lyko, F. and Schaefer, M. (2013). The RNA Methyltransferase Dnmt2 Is Required for Efficient Dicer-2-Dependent siRNA Pathway Activity in Drosophila. Cell Rep. PubMed ID: 24012760

    Durdevic, Z., Hanna, K., Gold, B., Pollex, T., Cherry, S., Lyko, F. and Schaefer, M. (2013). Efficient RNA virus control in Drosophila requires the RNA methyltransferase Dnmt2. EMBO Rep 14: 269-275. PubMed ID: 23370384

  • Stephen Schaeffer Department of Biology, Penn State
    Fuller, Z. L., Haynes, G. D., Richards, S. and Schaeffer, S. W. (2017). Genomics of natural populations: Evolutionary forces that establish and maintain gene arrangements in Drosophila pseudoobscura. Mol Ecol. PubMed ID: 29055159 Fuller, Z. L., Haynes, G. D., Richards, S. and Schaeffer, S. W. (2016). Genomics of natural populations: How differentially expressed genes shape the evolution of chromosomal inversions in Drosophila pseudoobscura. Genetics [Epub ahead of print]. PubMed ID: 27401754

    Wallace, A. G., Detweiler, D., Schaeffer, S. W. (2013) Molecular Population Genetics of Inversion Breakpoint Regions in Drosophila pseudoobscura. G3 (Bethesda). PubMed ID: 23665879

    Wallace, A. G., Detweiler, D. and Schaeffer, S. W. (2011). Evolutionary history of the third chromosome gene arrangements of Drosophila pseudoobscura inferred from inversion breakpoints. Mol Biol Evol 28: 2219-2229. PubMed ID: 21339510

    Meisel, R. P., Hilldorfer, B. B., Koch, J. L., Lockton, S. and Schaeffer, S. W. (2010). Adaptive evolution of genes duplicated from the Drosophila pseudoobscura neo-X chromosome. Mol Biol Evol 27: 1963-1978. PubMed ID: 20351054

  • Paul Schedl Molecular Biology, Princeton
    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

    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

    Gilmutdinov, R. A., Kozlov, E. N., Yakovlev, K. V., Olenina, L. V., Kotov, A. A., Barr, J., Zhukova, M. V., Schedl, P. and Shidlovskii, Y. V. (2021). The 3'UTR of the orb2 gene encoding the Drosophila CPEB translation factor plays a critical role in spermatogenesis. Development. PubMed ID: 34473243

    Diegmiller, R., Zhang, L., Gameiro, M., Barr, J., Imran Alsous, J., Schedl, P., Shvartsman, S. Y. and Mischaikow, K. (2021). Mapping parameter spaces of biological switches. PLoS Comput Biol 17(2): e1008711. PubMed ID: 33556054

    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

    Kyrchanova, O., Maksimenko, O., Ibragimov, A., Sokolov, V., Postika, N., Lukyanova, M., Schedl, P. and Georgiev, P. (2020). The insulator functions of the Drosophila polydactyl C2H2 zinc finger protein CTCF: Necessity versus sufficiency. Sci Adv 6(13): eaaz3152. PubMed ID: 32232161

    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

    Kyrchanova, O., Wolle, D., Sabirov, M., Kurbidaeva, A., Aoki, T., Maksimenko, O., Kyrchanova, M., Georgiev, P. and Schedl, P. (2019). Distinct elements confer the blocking and bypass functions of the Bithorax Fab-8 boundary. Genetics. PubMed ID: 31551239

    Barr, J., Gilmutdinov, R., Wang, L., Shidlovskii, Y. and Schedl, P. (2019). The Drosophila CPEB Protein Orb Specifies Oocyte Fate by a 3'UTR Dependent Autoregulatory Loop. Genetics. PubMed ID: 31594794

  • Annette Schenck Radboud Center for Mitochondrial Medicine, Nijmegen, Netherlands
    Fenckova, M., Blok, L. E. R., Asztalos, L., Goodman, D. P., Cizek, P., Singgih, E. L., Glennon, J. C., IntHout, J., Zweier, C., Eichler, E. E., von Reyn, C. R., Bernier, R. A., Asztalos, Z. and Schenck, A. (2019). Habituation Learning Is a Widely Affected Mechanism in Drosophila Models of Intellectual Disability and Autism Spectrum Disorders. Biol Psychiatry 86(4): 294-305. PubMed ID: 31272685

    Riahi, H., Brekelmans, C., Foriel, S., Merkling, S. H., Lyons, T. A., Itskov, P. M., Kleefstra, T., Ribeiro, C., van Rij, R. P., Kramer, J. M. and Schenck, A. (2019). The histone methyltransferase G9a regulates tolerance to oxidative stress-induced energy consumption. PLoS Biol 17(3): e2006146. PubMed ID: 30860988

    Castells-Nobau, A., Nijhof, B., Eidhof, I., Wolf, L., Scheffer-de Gooyert, J. M., Monedero, I., Torroja, L., van der Laak, J. and Schenck, A. (2017). Two algorithms for high-throughput and multi-parametric quantification of Drosophila neuromuscular junction morphology. J Vis Exp(123). PubMed ID: 28518121

    Esmaeeli-Nieh, S., Fenckova, M., Porter, I. M., Motazacker, M. M., Nijhof, B., Castells-Nobau, A., Asztalos, Z., Weissmann, R., Behjati, F., Tzschach, A., Felbor, U., Scherthan, H., Sayfati, S. M., Ropers, H. H., Kahrizi, K., Najmabadi, H., Swedlow, J. R., Schenck, A. and Kuss, A. W. (2016). BOD1 Is required for cognitive function in humans and Drosophila. PLoS Genet 12: e1006022. PubMed ID: 27166630

    Lugtenberg, D., et al. (2016). De novo loss-of-function mutations in WAC cause a recognizable intellectual disability syndrome and learning deficits in Drosophila. Eur J Hum Genet. PubMed ID: 26757981

    van der Voet, M., Harich, B., Franke, B. and Schenck, A. (2015). ADHD-associated dopamine transporter, latrophilin and neurofibromin share a dopamine-related locomotor signature in Drosophila. Mol Psychiatry [Epub ahead of print]. PubMed ID: 25962619

    Vandewalle, J., Langen, M., Zschaetzsch, M., Nijhof, B., Kramer, J. M., Brems, H., Bauters, M., Lauwers, E., Srahna, M., Marynen, P., Verstreken, P., Schenck, A., Hassan, B. A. and Froyen, G. (2013). Ubiquitin Ligase HUWE1 Regulates Axon Branching through the Wnt/beta-Catenin Pathway in a Drosophila Model for Intellectual Disability. PLoS One 8: e81791. PubMed ID: 24303071

    Oortveld, M. A., Keerthikumar, S., Oti, M., Nijhof, B., Fernandes, A. C., Kochinke, K., Castells-Nobau, A., van Engelen, E., Ellenkamp, T., Eshuis, L., Galy, A., van Bokhoven, H., Habermann, B., Brunner, H. G., Zweier, C., Verstreken, P., Huynen, M. A. and Schenck, A. (2013). Human intellectual disability genes form conserved functional modules in Drosophila. PLoS Genet 9: e1003911. PubMed ID: 24204314

    Willemsen, M. H., et al. (2013). GATAD2B loss-of-function mutations cause a recognisable syndrome with intellectual disability and are associated with learning deficits and synaptic undergrowth in Drosophila. J Med Genet 50: 507-514. PubMed ID: 23644463

    Koper, A., Schenck, A. and Prokop, A. (2012). Analysis of adhesion molecules and basement membrane contributions to synaptic adhesion at the Drosophila embryonic NMJ. PLoS One 7: e36339. PubMed ID: 22558441

    Galy, A., Schenck, A., Sahin, H. B., Qurashi, A., Sahel, J. A., Diebold, C. and Giangrande, A. (2011). CYFIP dependent actin remodeling controls specific aspects of Drosophila eye morphogenesis. Dev Biol 359: 37-46. PubMed ID: 21884694

  • Helgi Schiöth
    Alsehli, A. M., Liao, S., Al-Sabri, M. H., Vasionis, L., Purohit, A., Behare, N., Clemensson, L. E., Williams, M. J. and Schioth, H. B. (2022). The Statin Target HMG-Coenzyme a Reductase (Hmgcr) Regulates Sleep Homeostasis in Drosophila. Pharmaceuticals (Basel) 15(1). PubMed ID: 35056136

    Liu, W., Cao, H., Kimari, M., Maronitis, G., Williams, M. J. and Schioth, H. B. (2021). Multidrug Resistance Like Protein 1 Activity in Malpighian Tubules Regulates Lipid Homeostasis in Drosophila. Membranes (Basel) 11(6). PubMed ID: 34201304

    Williams, M. J., Akram, M., Barkauskaite, D., Patil, S., Kotsidou, E., Kheder, S., Vitale, G., Filaferro, M., Blemings, S. W., Maestri, G., Hazim, N., Vergoni, A. V. and Schioth, H. B. (2020). CCAP regulates feeding behavior via the NPF pathway in Drosophila adults. Proc Natl Acad Sci U S A. PubMed ID: 32179671

    Williams, M. J., Perland, E., Eriksson, M. M., Carlsson, J., Erlandsson, D., Laan, L., Mahebali, T., Potter, E., Frediksson, R., Benedict, C. and Schioth, H. B. (2016). Recurrent Sleep Fragmentation Induces Insulin and Neuroprotective Mechanisms in Middle-Aged Flies. Front Aging Neurosci 8: 180. PubMed ID: 27531979

    Wiemerslage, L., Gohel, P. A., Maestri, G., Hilmarsson, T. G., Mickael, M., Fredriksson, R., Williams, M. J. and Schioth, H. B. (2016). The Drosophila ortholog of TMEM18 regulates insulin and glucagon-like signaling. J Endocrinol. PubMed ID: 27029472

    Cao, H., Wiemerslage, L., Marttila, P. S., Williams, M. J. and Schioth, H. B. (2016). Bis-(2-ethylhexyl) Phthalate (DEHP) Increases Insulin Expression and Lipid Levels in Drosophila melanogaster. Basic Clin Pharmacol Toxicol. PubMed ID: 27009472

    Williams, M. J., Eriksson, A., Shaik, M., Voisin, S., Yamskova, O., Paulsson, J., Thombare, K., Fredriksson, R. and Schioth, H. B. (2015). The Obesity-Linked Gene Nudt3 Drosophila Homolog Aps Is Associated With Insulin Signaling. Mol Endocrinol 29: 1303-1319. PubMed ID: 26168034

    Williams, M. J., Goergen, P., Rajendran, J., Zheleznyakova, G., Hagglund, M. G., Perland, E., Bagchi, S., Kalogeropoulou, A., Khan, Z., Fredriksson, R. and Schioth, H. B. (2014). Obesity-linked homologues TfAP-2 and Twz establish meal frequency in Drosophila melanogaster. PLoS Genet 10: e1004499. PubMed ID: 25187989

  • Todd Schlenke Entomology, University of Arizona
    Mortimer, N. T., Fischer, M. L., Waring, A. L., Kr, P., Kacsoh, B. Z., Brantley, S. E., Keebaugh, E. S., Hill, J., Lark, C., Martin, J., Bains, P., Lee, J., Vrailas-Mortimer, A. D. and Schlenke, T. A. (2021). Extracellular matrix protein N-glycosylation mediates immune self-tolerance in Drosophila melanogaster. Proc Natl Acad Sci U S A 118(39). PubMed ID: 34544850

    Lynch, Z. R., Schlenke, T. A. and de Roode, J. C. (2016). Evolution of behavioral and cellular defenses against parasitoid wasps in the Drosophila melanogaster subgroup. J Evol Biol. PubMed ID: 26859227

    Singh N.D., Criscoe, D.R., Skolfield, S., Kohl, K.P., Keebaugh, E.S. and Schlenke, T.A. (2015). Fruit flies diversify their offspring in response to parasite infection. Science 349: 747-750. Direct link

    Keebaugh, E. S. and Schlenke, T. A. (2013). Insights From Natural Host-Parasite Interactions: The Drosophila Model. Dev Comp Immunol. PubMed ID: 23764256

    Mortimer, N. T., Goecks, J., Kacsoh, B. Z., Mobley, J. A., Bowersock, G. J., Taylor, J., Schlenke, T. A. (2013) Parasitoid wasp venom SERCA regulates Drosophila calcium levels and inhibits cellular immunity. Proc Natl Acad Sci U S A. PubMed ID: 23690612

    Mortimer, N. T., Kacsoh, B. Z., Keebaugh, E. S. and Schlenke, T. A. (2012). Mgat1-dependent N-glycosylation of membrane components primes Drosophila melanogaster blood cells for the cellular encapsulation response. PLoS Pathog 8: e1002819. PubMed ID: 22829770

    Kacsoh, B. Z., Lynch, Z. R., Mortimer, N. T., Schlenke, T. A. (2013) Fruit flies medicate offspring after seeing parasites. Science 339: 947-950. PubMed ID: 23430653

    Keebaugh, E. S. and Schlenke, T. A. (2012). Adaptive evolution of a novel Drosophila lectin induced by parasitic wasp attack. Mol Biol Evol 29: 565-577. PubMed ID: 21873297

    Lefevre, T., de Roode, J. C., Kacsoh, B. Z. and Schlenke, T. A. (2012). Defence strategies against a parasitoid wasp in Drosophila: fight or flight? Biol Lett 8: 230-233. PubMed ID: 21865240

  • Michael Schleyer Leibniz Institute for Neurobiology, Magdeburg, Germany
    Thoener, J., Konig, C., Weiglein, A., Toshima, N., Mancini, N., Amin, F. and Schleyer, M. (2021). Associative learning in larval and adult Drosophila is impaired by the dopamine-synthesis inhibitor 3-Iodo-L-tyrosine. Biol Open. PubMed ID: 33978715

    Thane, M., Viswanathan, V., Meyer, T. C., Paisios, E. and Schleyer, M. (2019). Modulations of microbehaviour by associative memory strength in Drosophila larvae. PLoS One 14(10): e0224154. PubMed ID: 31634372

    Michels, B., Saumweber, T., Biernacki, R., Thum, J., Glasgow, R. D. V., Schleyer, M., Chen, Y. C., Eschbach, C., Stocker, R. F., Toshima, N., Tanimura, T., Louis, M., Arias-Gil, G., Marescotti, M., Benfenati, F. and Gerber, B. (2017). Pavlovian Conditioning of Larval Drosophila: An Illustrated, Multilingual, Hands-On Manual for Odor-Taste Associative Learning in Maggots. Front Behav Neurosci 11: 45. PubMed ID: 28469564

    Kudow, N., Miura, D., Schleyer, M., Toshima, N., Gerber, B. and Tanimura, T. (2017). Preference for and learning of amino acids in larval Drosophila. Biol Open 6(3): 365-369. PubMed ID: 28193602

    Croset, V., Schleyer, M., Arguello, J. R., Gerber, B. and Benton, R. (2016). A molecular and neuronal basis for amino acid sensing in the Drosophila larva. Sci Rep 6: 34871. PubMed ID: 27982028

  • Christian Schlotterer Institute of Population Genetics, University of Veterinary Medicine, Vienna
    Lai, W. Y. and Schlotterer, C. (2021). Evolution of phenotypic variance in response to a novel hot environment. Mol Ecol. PubMed ID: 34775658

    Otte, K. A., Nolte, V., Mallard, F. and Schlotterer, C. (2021). The genetic architecture of temperature adaptation is shaped by population ancestry and not by selection regime. Genome Biol 22(1): 211. PubMed ID: 34271951

    Mallard, F., Nolte, V. and Schlotterer, C. (2020). The evolution of phenotypic plasticity in response to temperature stress. Genome Biol Evol. PubMed ID: 33022043

    Langmuller, A. M., Nolte, V., Galagedara, R., Poupardin, R., Dolezal, M. and Schlotterer, C. (2020). Fitness effects for Ace insecticide resistance mutations are determined by ambient temperature. BMC Biol 18(1): 157. PubMed ID: 33121485

    Hsu, S. K., Belmouaden, C., Nolte, V. and Schlotterer, C. (2020). Parallel gene expression evolution in natural and laboratory evolved populations. Mol Ecol. PubMed ID: 32979867

    Jaksic, A. M., Karner, J., Nolte, V., Hsu, S. K., Barghi, N., Mallard, F., Otte, K. A., Svecnjak, L., Senti, K. A. and Schlotterer, C. (2020). Neuronal function and dopamine signaling evolve at high temperature in Drosophila. Mol Biol Evol. PubMed ID: 32402077

    Hsu, S. K., Jaksic, A. M., Nolte, V., Lirakis, M., Kofler, R., Barghi, N., Versace, E. and Schlotterer, C. (2020). Rapid sex-specific adaptation to high temperature in Drosophila. Elife 9. PubMed ID: 32083552
    Howie, J. M., Mazzucco, R., Taus, T., Nolte, V. and Schlotterer, C. (2019). DNA motifs are not general predictors of recombination in two Drosophila sister species. Genome Biol Evol. PubMed ID: 30980655

    Hsu, S. K., Jaksic, A. M., Nolte, V., Barghi, N., Mallard, F., Otte, K. A. and Schlotterer, C. (2019). A 24 h age difference causes twice as much gene expression divergence as 100 generations of adaptation to a novel environment. Genes (Basel) 10(2). PubMed ID: 30696109

    Barghi, N., Tobler, R., Nolte, V., Jaksic, A. M., Mallard, F., Otte, K. A., Dolezal, M., Taus, T., Kofler, R. and Schlotterer, C. (2019). Genetic redundancy fuels polygenic adaptation in Drosophila. PLoS Biol 17(2): e3000128. PubMed ID: 30716062

    Mallard, F., Nolte, V., Tobler, R., Kapun, M. and Schlotterer, C. (2018). A simple genetic basis of adaptation to a novel thermal environment results in complex metabolic rewiring in Drosophila. Genome Biol 19(1): 119. PubMed ID: 30122150

    Endler, L., Gibert, J. M., Nolte, V. and Schlotterer, C. (2018). Pleiotropic effects of regulatory variation in tan result in correlation of two pigmentation traits in Drosophila melanogaster. Mol Ecol. PubMed ID: 29957826

    Lirakis, M., Dolezal, M. and Schlotterer, C. (2018). Redefining reproductive dormancy in Drosophila as a general stress response to cold temperatures. J Insect Physiol 107: 175-185. PubMed ID: 29649483

    Tobler, R., Nolte, V. and Schlotterer, C. (2017). High rate of translocation-based gene birth on the Drosophila Y chromosome. Proc Natl Acad Sci U S A 114(44): 11721-11726. PubMed ID: 29078298

  • Paul Schmidt Department of Biology, University of Pennsylvania
    Rudman, S. M., Greenblum, S. I., Rajpurohit, S., Betancourt, N. J., Hanna, J., Tilk, S., Yokoyama, T., Petrov, D. A. and Schmidt, P. (2022). Direct observation of adaptive tracking on ecological time scales in Drosophila. Science 375(6586): eabj7484. PubMed ID: 35298245

    Betancourt, N. J., Rajpurohit, S., Durmaz, E., Fabian, D. K., Kapun, M., Flatt, T. and Schmidt, P. (2021). Allelic polymorphism at foxo contributes to local adaptation in Drosophila melanogaster. Mol Ecol. PubMed ID: 33914989

    Grainger, T. N., Rudman, S. M., Schmidt, P. and Levine, J. M. (2021). Competitive history shapes rapid evolution in a seasonal climate. Proc Natl Acad Sci U S A 118(6). PubMed ID: 33536336

    Rajpurohit, S., Vrkoslav, V., Hanus, R., Gibbs, A. G., Cvacka, J. and Schmidt, P. S. (2021). Post-eclosion temperature effects on insect cuticular hydrocarbon profiles. Ecol Evol 11(1): 352-364. PubMed ID: 33437434

    Rudman, S. M., Greenblum, S., Hughes, R. C., Rajpurohit, S., Kiratli, O., Lowder, D. B., Lemmon, S. G., Petrov, D. A., Chaston, J. M. and Schmidt, P. (2019). Microbiome composition shapes rapid genomic adaptation of Drosophila melanogaster. Proc Natl Acad Sci U S A 116(40): 20025-20032. PubMed ID: 31527278

    Rajpurohit, S., Gefen, E., Bergland, A. O., Petrov, D. A., Gibbs, A. G. and Schmidt, P. (2018). Spatiotemporal dynamics and genome-wide association analysis of desiccation tolerance in Drosophila melanogaster. Mol Ecol. PubMed ID: 30051644

    Rajpurohit, S., Richardson, R., Dean, J., Vazquez, R., Wong, G. and Schmidt, P. S. (2016). Pigmentation and fitness trade-offs through the lens of artificial selection. Biol Lett 12(10). PubMed ID: 28120808

    Mathur, V. and Schmidt, P.S. (2016). Adaptive patterns of phenotypic plasticity in laboratory and field environments in Drosophila melanogaster. Evolution [Epub ahead of print]. PubMed ID: 27925178

    Rajpurohit, S. and Schmidt, P. S. (2016). Measuring thermal behavior in smaller insects: a case study in Drosophila melanogaster demonstrates effects of sex, geographic origin, and rearing temperature on adult behavior. Fly (Austin): 0. PubMed ID: 27230726

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

  • Urs Schmidt-Ott Department of Organismal Biology and Anatomy, University of Chicago
    Yoon, Y., Klomp, J., Martin-Martin, I., Criscione, F., Calvo, E., Ribeiro, J. and Schmidt-Ott, U. (2019). Embryo polarity in moth flies and mosquitoes relies on distinct old genes with localized transcript isoforms. Elife 8. PubMed ID: 31591963

    Liu, Q., Onal, P., Datta, R. R., Rogers, J. M., Schmidt-Ott, U., Bulyk, M. L., Small, S. and Thornton, J. W. (2018). Ancient mechanisms for the evolution of the bicoid homeodomain's function in fly development. Elife 7. PubMed ID: 28005004

    Klomp, J., Athy, D., Kwan, C. W., Bloch, N. I., Sandmann, T., Lemke, S. and Schmidt-Ott, U. (2015). Embryo development. A cysteine-clamp gene drives embryo polarity in the midge Chironomus. Science 348(6238): 1040-1042. PubMed ID: 25953821

  • Dietmar Schmucker Vesalius Research Center, Leuven, Belgium
    Izadifar, A., Courchet, J., Virga, D. M., Verreet, T., Hamilton, S., Ayaz, D., Misbaer, A., Vandenbogaerde, S., Monteiro, L., Petrovic, M., Sachse, S., Yan, B., Erfurth, M. L., Dascenco, D., Kise, Y., Yan, J., Edwards-Faret, G., Lewis, T., Polleux, F. and Schmucker, D. (2021). Axon morphogenesis and maintenance require an evolutionary conserved safeguard function of Wnk kinases antagonizing Sarm and Axed. Neuron. PubMed ID: 34384519

    Urwyler, O., Izadifar, A., Vandenbogaerde, S., Sachse, S., Misbaer, A. and Schmucker, D. (2019). Branch-restricted localization of phosphatase Prl-1 specifies axonal synaptogenesis domains. Science 364(6439). PubMed ID: 31048465

    Koch, M., Nicolas, M., Zschaetzsch, M., de Geest, N., Claeys, A., Yan, J., Morgan, M. J., Erfurth, M. L., Holt, M., Schmucker, D. and Hassan, B. A. (2017). A Fat-Facets-Dscam1-JNK Pathway Enhances Axonal Growth in Development and after Injury. Front Cell Neurosci 11: 416. PubMed ID: 29472843

    Dascenco, D., Erfurth, M. L., Izadifar, A., Song, M., Sachse, S., Bortnick, R., Urwyler, O., Petrovic, M., Ayaz, D., He, H., Kise, Y., Thomas, F., Kidd, T. and Schmucker, D. (2015). Slit and Receptor tyrosine phosphatase 69D confer spatial specificity to axon branching via Dscam1. Cell 162: 1140-1154. PubMed ID: 26317474

    Urwyler, O., Izadifar, A., Dascenco, D., Petrovic, M., He, H., Ayaz, D., Kremer, A., Lippens, S., Baatsen, P., Guerin, C. J. and Schmucker, D. (2014). Investigating CNS synaptogenesis at single-synapse resolution by combining reverse genetics with correlative light and electron microscopy. Development. PubMed ID: 25503410

    Armitage, S. A., Sun, W., You, X., Kurtz, J., Schmucker, D. and Chen, W. (2014). Quantitative profiling of Drosophila melanogaster Dscam1 isoforms reveals no changes in splicing after bacterial exposure. PLoS One 9: e108660. PubMed ID: 25310676

    Kise, Y. and Schmucker, D. (2013). Role of self-avoidance in neuronal wiring. Curr Opin Neurobiol 23: 983-989. PubMed ID: 24161734

    He, H., Kise, Y., Izadifar, A., Urwyler, O., Ayaz, D., Parthasarthy, A., Yan, B., Erfurth, M. L., Dascenco, D. and Schmucker, D. (2014). Cell-Intrinsic Requirement of Dscam1 Isoform Diversity for Axon Collateral Formation. Science. PubMed ID: 24831526

    Sun, W., You, X., Gogol-Doring, A., He, H., Kise, Y., Sohn, M., Chen, T., Klebes, A., Schmucker, D. and Chen, W. (2013). Ultra-deep profiling of alternatively spliced Drosophila Dscam isoforms by circularization-assisted multi-segment sequencing. EMBO J 32: 2029-2038. PubMed ID: 23792425

    Langen, M., Koch, M., Yan, J., De Geest, N., Erfurth, M. L., Pfeiffer, B. D., Schmucker, D., Moreau, Y. and Hassan, B. A. (2013). Mutual inhibition among postmitotic neurons regulates robustness of brain wiring in Drosophila. Elife 2: e00337. PubMed ID: 23471010

  • David Schneider Stanford School of Medicine
    Chambers, M. C., Lightfield, K. L., Schneider, D. S. (2012) How the fly balances its ability to combat different pathogens. PLoS Pathog 8: e1002970. PubMed ID: 23271964

    Chambers, M. C., Song, K. H. and Schneider, D. S. (2012). Listeria monocytogenes Infection Causes Metabolic Shifts in Drosophila melanogaster. PLoS One 7: e50679. PubMed ID: 23272066

    Linderman, J. A., Chambers, M. C., Gupta, A. S. and Schneider, D. S. (2012). Infection-related declines in chill coma recovery and negative geotaxis in Drosophila melanogaster. PLoS One 7: e41907. PubMed ID: 23028430 Free Radic Biol Med [Epub ahead of print]. PubMed ID: 25841783

    Navarro, J. A., Hessner, S., Yenisetti, S. C., Bayersdorfer, F., Zhang, L., Voigt, A., Schneuwly, S. and Botella, J. A. (2014). Analysis of dopaminergic neuronal dysfunction in genetic and toxin-induced models of Parkinson's disease in Drosophila. J Neurochem. PubMed ID: 25040725

    Rowshanravan, B., Woodcock, S. A., Botella, J. A., Kiermayer, C., Schneuwly, S. and Hughes, D. A. (2014). RasGAP mediates neuronal survival in Drosophila through direct regulation of Rab5-dependent endocytosis. J Cell Sci. PubMed ID: 24816559

    Kosmidis, S., Missirlis, F., Botella, J. A., Schneuwly, S., Rouault, T. A. and Skoulakis, E. M. (2014). Behavioral decline and premature lethality upon pan-neuronal ferritin overexpression in Drosophila infected with a virulent form of Wolbachia. Front Pharmacol 5: 66. PubMed ID: 24772084

    Fischer, S., Bayersdorfer, F., Harant, E., Reng, R., Arndt, S., Bosserhoff, A. K. and Schneuwly, S. (2012). fussel (fuss)--a negative regulator of BMP signaling in Drosophila melanogaster. PLoS One 7: e42349. PubMed ID: 22879948

    Kosmidis, S., Botella, J. A., Mandilaras, K., Schneuwly, S., Skoulakis, E. M., Rouault, T. A. and Missirlis, F. (2011). Ferritin overexpression in Drosophila glia leads to iron deposition in the optic lobes and late-onset behavioral defects. Neurobiol Dis 43: 213-219. PubMed ID: 21440626

    Navarro, J. A., Ohmann, E., Sanchez, D., Botella, J. A., Liebisch, G., Molto, M. D., Ganfornina, M. D., Schmitz, G. and Schneuwly, S. (2010). Altered lipid metabolism in a Drosophila model of Friedreich's ataxia. Hum Mol Genet 19: 2828-2840. PubMed ID: 20460268

  • Frank Schnorrer Developmental Biology Institute of Marseille
    Avellaneda, J., Rodier, C., Daian, F., Brouilly, N., Rival, T., Luis, N. M. and Schnorrer, F. (2021). Myofibril and mitochondria morphogenesis are coordinated by a mechanical feedback mechanism in muscle. Nat Commun 12(1): 2091. PubMed ID: 33828099

    Kaya-Copur, A., Marchiano, F., Hein, M. Y., Alpern, D., Russeil, J., Luis, N. M., Mann, M., Deplancke, B., Habermann, B. H. and Schnorrer, F. (2021). The Hippo pathway controls myofibril assembly and muscle fiber growth by regulating sarcomeric gene expression. Elife 10. PubMed ID: 33404503

    Catinozzi, M., Mallik, M., Frickenhaus, M., Been, M., Sijlmans, C., Kulshrestha, D., Alexopoulos, I., Weitkunat, M., Schnorrer, F. and Storkebaum, E. (2020). The Drosophila FUS ortholog cabeza promotes adult founder myoblast selection by Xrp1-dependent regulation of FGF signaling. PLoS Genet 16(4): e1008731. PubMed ID: 32302304

    Weitkunat, M., Lindauer, M., Bausch, A. and Schnorrer, F. (2017). Mechanical tension and spontaneous muscle twitching precede the formation of cross-striated muscle in vivo. Development [Epub ahead of print]. PubMed ID: 28174246

    Ordan, E., Brankatschk, M., Dickson, B., Schnorrer, F. and Volk, T. (2015). Slit cleavage is essential for producing an active, stable, non-diffusible short-range signal that guides muscle migration. Development [Epub ahead of print]. PubMed ID: 25813540

    Spletter, M.L., Barz, C., Yeroslaviz, A., Schönbauer, C., Ferreira, I.R., Sarov, M., Gerlach, D., Stark, A., Habermann, B.H. and Schnorrer, F. (2014). The RNA-binding protein Arrest (Bruno) regulates alternative splicing to enable myofibril maturation in Drosophila flight muscle. EMBO Rep. [Epub ahead of print]. PubMed ID: 25532219

    Zhang, X., Ferreira, I. R. and Schnorrer, F. (2014). A simple TALEN-based protocol for efficient genome-editing in Drosophila. Methods. PubMed ID: 24680697

    Weitkunat, M., Kaya-Copur, A., Grill, S. W. and Schnorrer, F. (2014). Tension and Force-Resistant Attachment Are Essential for Myofibrillogenesis in Drosophila Flight Muscle. Curr Biol. PubMed ID: 24631244

    Klein, P., Muller-Rischart, A. K., Motori, E., Schonbauer, C., Schnorrer, F., Winklhofer, K. F. and Klein, R. (2014). Ret rescues mitochondrial morphology and muscle degeneration of Drosophila Pink1 mutants. EMBO J. PubMed ID: 24473149

    Spletter, M. L. and Schnorrer, F. (2013). Transcriptional regulation and alternative splicing cooperate in muscle fiber-type specification in flies and mammals. Exp Cell Res. PubMed ID: 24145055

  • Stephan Schneuwly University of Regensburg
    Rass, M., Gizler, L., Bayersdorfer, F., Irlbeck, C., Schramm, M. and Schneuwly, S. (2022). The Drosophila functional Smad suppressing element fuss, a homologue of the human Skor genes, retains pro-oncogenic properties of the Ski/Sno family. PLoS One 17(1): e0262360. PubMed ID: 35030229

    Rass, M., Oestreich, S., Guetter, S., Fischer, S. and Schneuwly, S. (2019). The Drosophila fussel gene is required for bitter gustatory neuron differentiation acting within an Rpd3 dependent chromatin modifying complex. PLoS Genet 15(2): e1007940. PubMed ID: 30730884

    Moschall, R., Rass, M., Rossbach, O., Lehmann, G., Kullmann, L., Eichner, N., Strauss, D., Meister, G., Schneuwly, S., Krahn, M. P. and Medenbach, J. (2018). Drosophila Sister-of-Sex-lethal reinforces a male-specific gene expression pattern by controlling Sex-lethal alternative splicing. Nucleic Acids Res. PubMed ID: 30590805

    Edenharter, O., Schneuwly, S. and Navarro, J. A. (2018). Mitofusin-Dependent ER Stress Triggers Glial Dysfunction and Nervous System Degeneration in a Drosophila Model of Friedreich's Ataxia. Front Mol Neurosci 11: 38. PubMed ID: 29563863

    Edenharter, O., Clement, J., Schneuwly, S. and Navarro, J. A. (2017). Overexpression of Drosophila frataxin triggers cell death in an iron-dependent manner. J Neurogenet 31(4): 189-202. PubMed ID: 28838288

  • Frieder Schöck Department of Biology, McGill University, Montreal
    Liao, K. A., Gonzalez-Morales, N. and Schock, F. (2020). Characterizing the actin-binding ability of Zasp52 and its contribution to myofibril assembly. PLoS One 15(7): e0232137. PubMed ID: 32614896

    Gonzalez-Morales, N., Xiao, Y. S., Schilling, M. A., Marescal, O., Liao, K. A. and Schock, F. (2019). Myofibril diameter is set by a finely tuned mechanism of protein oligomerization in Drosophila. Elife 8. PubMed ID: 31746737

    Gonzalez-Morales, N., Xiao, Y. S., Schilling, M. A., Marescal, O., Liao, K. A. and Schock, F. (2019). Myofibril diameter is set by a finely tuned mechanism of protein oligomerization in Drosophila. Elife 8. PubMed ID: 31746737

    Katzemich, A., Long, J. Y., Panneton, V., Fisher, L., Hipfner, D. and Schock, F. (2019). Slik phosphorylation of talin T152 is crucial for proper talin recruitment and maintenance of muscle attachment in Drosophila. Development. PubMed ID: 31511253

    Katzemich, A., Liao, K. A., Czerniecki, S., Schock, F. (2013) Alp/Enigma family proteins cooperate in z-disc formation and myofibril assembly. PLoS Genet 9: e1003342. PubMed ID: 23505387

    Bouaouina, M., Jani, K., Long, J. Y., Czerniecki, S., Morse, E. M., Ellis, S. J., Tanentzapf, G., Schock, F. and Calderwood, D. A. (2012). Zasp regulates integrin activation. J Cell Sci 125: 5647-5657. PubMed ID: 22992465

    Rohn, J. L., Sims, D., Liu, T., Fedorova, M., Schock, F., Dopie, J., Vartiainen, M. K., Kiger, A. A., Perrimon, N. and Baum, B. (2011). Comparative RNAi screening identifies a conserved core metazoan actinome by phenotype. J Cell Biol 194: 789-805

    Katzemich, A., Long, J. Y., Jani, K., Lee, B. R. and Schock, F. (2011). Muscle type-specific expression of Zasp52 isoforms in Drosophila. Gene Expr Patterns 11: 484-490. PubMed ID: 21867777

  • Jonathan Scholey University of California, Davis
    Brust-Mascher, I., Civelekoglu-Scholey, G. and Scholey, J. M. (2015). Mechanism for Anaphase B: Evaluation of "Slide-and-Cluster" versus "Slide-and-Flux-or-Elongate" Models. Biophys J 108: 2007-2018. PubMed ID: 25902440

    Wang, H., Brust-Mascher, I. and Scholey, J. M. (2015). The microtubule crosslinker Feo controls the midzone stability, motor composition and elongation of the anaphase B spindle in Drosophila embryos. Mol Biol Cell. PubMed ID: 25694445

    Brust-Mascher, I., Civelekoglu-Scholey, G. and Scholey, J. M. (2014). Analysis of mitotic protein dynamics and function in Drosophila embryos by live cell imaging and quantitative modeling. Methods Mol Biol 1136: 3-30. PubMed ID: 24633790

    Acar, S., Carlson, D. B., Budamagunta, M. S., Yarov-Yarovoy, V., Correia, J. J., Ninonuevo, M. R., Jia, W., Tao, L., Leary, J. A., Voss, J. C., Evans, J. E. and Scholey, J. M. (2013). The bipolar assembly domain of the mitotic motor kinesin-5. Nat Commun 4: 1343. PubMed ID: 23299893

    de Lartigue, J., Brust-Mascher, I. and Scholey, J. M. (2011). Anaphase B spindle dynamics in Drosophila S2 cells: Comparison with embryo spindles. Cell Div 6: 8. PubMed ID: 21477279

    Brust-Mascher, I. and Scholey, J. M. (2009). Microinjection techniques for studying mitosis in the Drosophila melanogaster syncytial embryo. J Vis Exp. PubMed ID: 19755959

  • Henrike Scholz Universität zu Köln
    He, J., Hommen, F., Lauer, N., Balmert, S. and Scholz, H. (2020). Serotonin transporter dependent modulation of food-seeking behavior. PLoS One 15(1): e0227554. PubMed ID: 31978073

    Classen, G. and Scholz, H. (2018). Octopamine shifts the behavioral response from indecision to approach or aversion in Drosophila melanogaster. Front Behav Neurosci 12: 131. PubMed ID: 30018540

    Giang, T., He, J., Belaidi, S. and Scholz, H. (2017). Key odorants regulate food attraction in Drosophila melanogaster. Front Behav Neurosci 11: 160. PubMed ID: 28928642

    Ruppert, M., Franz, M., Saratsis, A., Velo Escarcena, L., Hendrich, O., Gooi, L. M., Schwenkert, I., Klebes, A. and Scholz, H. (2017). Hangover Links Nuclear RNA Signaling to cAMP Regulation via the Phosphodiesterase 4d Ortholog dunce. Cell Rep 18(2): 533-544. PubMed ID: 28076795

    Xu, L., He, J., Kaiser, A., Graber, N., Schlager, L., Ritze, Y. and Scholz, H. (2016). A single pair of serotonergic neurons counteracts serotonergic inhibition of ethanol attraction in Drosophila. PLoS One 11(12): e0167518. PubMed ID: 27936023

    Scheiner, R., Steinbach, A., Classen, G., Strudthoff, N. and Scholz, H. (2014). Octopamine indirectly affects proboscis extension response habituation in Drosophila melanogaster by controlling sucrose responsiveness. J Insect Physiol. PubMed ID: 24819202

    Schneider, A., Ruppert, M., Hendrich, O., Giang, T., Ogueta, M., Hampel, S., Vollbach, M., Buschges, A. and Scholz, H. (2012). Neuronal basis of innate olfactory attraction to ethanol in Drosophila. PLoS One 7: e52007. PubMed ID: 23284851

    Wosnitza, A., Bockemuhl, T., Dubbert, M., Scholz, H. and Buschges, A. (2012). Inter-leg coordination in the control of walking speed in Drosophila. J Exp Biol. PubMed ID: 23038731

    Aso, Y., Herb, A., Ogueta, M., Siwanowicz, I., Templier, T., Friedrich, A. B., Ito, K., Scholz, H. and Tanimoto, H. (2012). Three dopamine pathways induce aversive odor memories with different stability. PLoS Genet 8: e1002768. PubMed ID: 22807684

  • Reinhard Schuh Max Planck Institute for Biophysical Chemistry, Dept. of Molecular Developmental Biology, Gõttingen
    Konigsmann, T., Parfentev, I., Urlaub, H., Riedel, D. and Schuh, R. (2020). The bicistronic gene wurmchen encodes two essential components for epithelial development in Drosophila. Dev Biol. PubMed ID: 32361005

    Hildebrandt, A., Pflanz, R., Behr, M., Tarp, T., Riedel, D. and Schuh, R. (2015). Bark beetle controls epithelial morphogenesis by septate junction maturation in Drosophila. Dev Biol [Epub ahead of print]. PubMed ID: 25704509

    Jaspers, M. H., Pflanz, R., Riedel, D., Kawelke, S., Feussner, I. and Schuh, R. (2013). The fatty acyl-CoA reductase Waterproof mediates airway clearance in Drosophila. Dev Biol. PubMed ID: 24183938

    Weiss, A., Charbonnier, E., Ellertsdottir, E., Tsirigos, A., Wolf, C., Schuh, R., Pyrowolakis, G. and Affolter, M. (2010). A conserved activation element in BMP signaling during Drosophila development. Nat Struct Mol Biol 17: 69-76. PubMed ID: 20010841

  • Oren Schuldiner Department of Molecular Cell Biology, Weizmann Institute of Science
    Bornstein, B., Meltzer, H., Adler, R., Alyagor, I., Berkun, V., Cummings, G., Reh, F., Keren-Shaul, H., David, E., Riemensperger, T. and Schuldiner, O. (2021). Transneuronal Dpr12/DIP-δ interactions facilitate compartmentalized dopaminergic innervation of Drosophila mushroom body axons. Embo J: e105763. PubMed ID: 33847376

    Yaniv, S. P., Meltzer, H., Alyagor, I. and Schuldiner, O. (2020). Developmental axon regrowth and primary neuron sprouting utilize distinct actin elongation factors. J Cell Biol 219(5). PubMed ID: 32191286

    Sudarsanam, S., Yaniv, S., Meltzer, H. and Schuldiner, O. (2020). Cofilin regulates axon growth and branching of Drosophila gamma neurons. J Cell Sci. PubMed ID: 32152181

    Marmor-Kollet, N., Gutman, I., Issman-Zecharya, N. and Schuldiner, O. (2019). Glial derived TGF-beta instructs axon midline stopping. Front Mol Neurosci 12: 232. PubMed ID: 31611773

    Alyagor, I., Berkun, V., Keren-Shaul, H., Marmor-Kollet, N., David, E., Mayseless, O., Issman-Zecharya, N., Amit, I. and Schuldiner, O. (2018). Combining Developmental and Perturbation-Seq Uncovers Transcriptional Modules Orchestrating Neuronal Remodeling. Dev Cell 47(1): 38-52.e36. PubMed ID: 30300589

    Mayseless, O., Berns, D. S., Yu, X. M., Riemensperger, T., Fiala, A. and Schuldiner, O. (2018). Developmental coordination during olfactory circuit remodeling in Drosophila. Neuron. PubMed ID: 30146303

    Rabinovich, D., Yaniv, S.P., Alyagor, I. and Schuldiner, O. (2016). Nitric oxide as a switching mechanism between axon degeneration and regrowth during developmental remodeling. Cell 164: 170-182. PubMed ID: 26771490

    Bornstein, B., Zahavi, E. E., Gelley, S., Zoosman, M., Yaniv, S. P., Fuchs, O., Porat, Z., Perlson, E. and Schuldiner, O. (2015). Developmental axon pruning requires destabilization of cell adhesion by JNK signaling. Neuron 88: 926-940. PubMed ID: 26586184

    Rabinovich, D., Mayseless, O. and Schuldiner, O. (2015). Long term ex vivo culturing of Drosophila brain as a method to live image pupal brains: insights into the cellular mechanisms of neuronal remodeling. Front Cell Neurosci 9: 327. PubMed ID: 26379498

    Marmor-Kollet, N. and Schuldiner, O. (2015). Contrasting developmental axon regrowth and neurite sprouting of Drosophila mushroom body neurons reveals shared and unique molecular mechanisms. Dev Neurobiol. PubMed ID: 26037037

  • Cordula Schultz Department of Cellular Biology, University of Georgia, Athens, Georgia
    Malpe, M. S., McSwain, L. F., Kudyba, K., Ng, C. L., Nicholson, J., Brady, M., Qian, Y., Choksi, V., Hudson, A. G., Parrott, B. B. and Schulz, C. (2020). G-protein signaling is required for increasing germline stem cell division frequency in response to mating in Drosophila males. Sci Rep 10(1): 3888. PubMed ID: 32127590

    Ng, C. L., Qian, Y. and Schulz, C. (2019). Notch and Delta are required for survival of the germline stem cell lineage in testes of Drosophila melanogaster. PLoS One 14(9): e0222471. PubMed ID: 31513679

    Rabinovich, D., Yaniv, S. P., Alyagor, I. and Schuldiner, O. (2016). Nitric oxide as a switching mechanism between axon degeneration and regrowth during developmental remodeling. Cell 164: 170-182. PubMed ID: 26771490

    Qian, Y., Ng, C. L. and Schulz, C. (2014). CSN maintains the germline cellular microenvironment and controls the level of stem cell genes via distinct CRLs in testes of Drosophila melanogaster. Dev Biol. PubMed ID: 25459658

    Qian, Y., Dominado, N., Zoller, R., Ng, C., Kudyba, K., Siddall, N. A., Hime, G. G. and Schulz, C. (2014). Ecdysone signaling opposes epidermal growth factor signaling in regulating cyst differentiation in the male gonad of Drosophila melanogaster. Dev Biol. PubMed ID: 25169192

    Hudson, A. G., Parrott, B. B., Qian, Y. and Schulz, C. (2013). A Temporal Signature of Epidermal Growth Factor Signaling Regulates the Differentiation of Germline Cells in Testes of Drosophila melanogaster. PLoS One 8: e70678. PubMed ID: 23940622

    Zoller, R. and Schulz, C. (2012). The Drosophila cyst stem cell lineage: Partners behind the scenes? Spermatogenesis 2: 145-157. PubMed ID: 23087834

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

  • Robert Schulz Department of Biological Sciences, Notre Dame University
    Tokusumi, Y., Tokusumi, T. and Schulz, R. A. (2018). Mechanical stress to Drosophila larvae stimulates a cellular immune response through the JAK/STAT signaling pathway. Biochem Biophys Res Commun. Pubmed ID: 29856996

    Tokusumi, Y., Tokusumi, T. and Schulz, R. A. (2017). The nociception genes painless and Piezo are required for the cellular immune response of Drosophila larvae to wasp parasitization. Biochem Biophys Res Commun. PubMed ID: 28342875

    Tokusumi, T., Tokusumi, Y., Brahier, M. S., Lam, V., Stoller-Conrad, J. R., Kroeger, P. T., Jr. and Schulz, R. A. (2016). Screening and analysis of Janelia FlyLight project enhancer-Gal4 strains identifies multiple gene enhancers active during hematopoiesis in normal and wasp-challenged Drosophila larvae. G3 (Bethesda). PubMed ID: 27913635

    Tokusumi, T., Tokusumi, Y., Hopkins, D. W. and Schulz, R. A. (2015). Bag of Marbles controls the size and organization of the Drosophila hematopoietic niche through interactions with the Insulin-like growth factor pathway and Retinoblastoma-family protein. Development. PubMed ID: 26041767

    Lam, V., Tokusumi, T., Tokusumi, Y. and Schulz, R. A. (2014). bantam miRNA is important for Drosophila blood cell homeostasis and a regulator of proliferation in the hematopoietic progenitor niche. Biochem Biophys Res Commun. PubMed ID: 25280996

    Kroeger, P. T., Jr., Shoue, D. A., Mezzacappa, F. M., Gerlach, G. F., Wingert, R. A. and Schulz, R. A. (2013). Knockdown of SCF(Skp2) Function Causes Double-Parked Accumulation in the Nucleus and DNA Re-Replication in Drosophila Plasmatocytes. PLoS One 8: e79019. PubMed ID: 24205363

    Tokusumi, Y., Tokusumi, T., Shoue, D. A. and Schulz, R. A. (2012). Gene regulatory networks controlling hematopoietic progenitor niche cell production and differentiation in the Drosophila lymph gland. PLoS One 7: e41604. PubMed ID: 22911822

    Xu, P., Johnson, T. L., Stoller-Conrad, J. R. and Schulz, R. A. (2012). Spire, an actin nucleation factor, regulates cell division during Drosophila heart development. PLoS One 7: e30565. PubMed ID: 22276214

    Kramm, A., Kisiela, M., Schulz, R. and Maser, E. (2012). Short-chain dehydrogenases/reductases in cyanobacteria. FEBS J. PubMed ID: 22251568

  • Trudy Schüpbach Molecular Biology, Princeton
    Wittes, J. and Schupbach, T. (2018). A gene expression screen in Drosophila melanogaster identifies novel JAK/STAT and EGFR targets during oogenesis. G3 (Bethesda). PubMed ID: 30385460

    Pritykin, Y., Brito, T., Schupbach, T., Singh, M. and Pane, A. (2017). Integrative analysis unveils new functions for the Drosophila Cutoff protein in non-coding RNA biogenesis and gene regulation. RNA [Epub ahead of print]. PubMed ID: 28420675

    Devergne, O., Sun, G. H. and Schupbach, T. (2017). Stratum, a homolog of the human GEF Mss4, partnered with Rab8, controls the basal restriction of basement membrane proteins in epithelial cells. Cell Rep 18(8): 1831-1839. PubMed ID: 28228250

    Johnson, H. E., Goyal, Y., Pannucci, N. L., Schupbach, T., Shvartsman, S. Y. and Toettcher, J. E. (2017). The spatiotemporal limits of developmental Erk signaling. Dev Cell 40(2): 185-192. PubMed ID: 28118601

    Goyal, Y., Jindal, G. A., Pelliccia, J. L., Yamaya, K., Yeung, E., Futran, A. S., Burdine, R. D., Schupbach, T. and Shvartsman, S. Y. (2017). Divergent effects of intrinsically active MEK variants on developmental Ras signaling. Nat Genet [Epub ahead of print]. PubMed ID: 28166211

    Jindal, G. A., Goyal, Y., Yamaya, K., Futran, A. S., Kountouridis, I., Balgobin, C. A., Schupbach, T., Burdine, R. D. and Shvartsman, S. Y. (2017). In vivo severity ranking of Ras pathway mutations associated with developmental disorders. Proc Natl Acad Sci U S A. PubMed ID: 28049852

    Lim, B., Dsilva, C.J., Levario, T.J., Lu, H., Schüpbach, T., Kevrekidis, I.G. and Shvartsman, S.Y. (2015). Dynamics of inductive ERK signaling in the Drosophila embryo.Curr Biol [Epub ahead of print]. PubMed ID: 26096970

    Osterfield, M., Schupbach, T., Wieschaus, E. and Shvartsman, S. Y. (2015). Diversity of epithelial morphogenesis during eggshell formation in drosophilids. Development. PubMed ID: 25953345

    Li, W., Klovstad, M. and Schupbach, T. (2014). Repression of Gurken translation by a meiotic checkpoint in Drosophila oogenesis is suppressed by a reduction in the dose of eIF1A. Development. PubMed ID: 25231760

    Devergne, O., Tsung, K., Barcelo, G. and Schupbach, T. (2014). Polarized deposition of basement membrane proteins depends on Phosphatidylinositol synthase and the levels of Phosphatidylinositol 4,5-bisphosphate. Proc Natl Acad Sci U S A. PubMed ID: 24828534

    Domanitskaya, E., Anllo, L. and Schupbach, T. (2013). Phantom, a cytochrome P450 enzyme essential for ecdysone biosynthesis, plays a critical role in the control of border cell migration in Drosophila. Dev Biol. PubMed ID: 24373956

    Fontenele, M., Lim, B., Oliveira, D., Buffolo, M., Perlman, D. H., Schupbach, T. and Araujo, H. (2013). Calpain A modulates Toll responses by limited Cactus/IkappaB proteolysis. Mol Biol Cell. PubMed ID: 23864715

  • Lawrence Schwartz Biology, University of Massachusetts, Amherst
    Sheel, A., Shao, R., Brown, C., Johnson, J., Hamilton, A., Sun, D., Oppenheimer, J., Smith, W., Visconti, P. E., Markstein, M., Bigelow, C. and Schwartz, L. M. (2020). Acheron/Larp6 Is a Survival Protein That Protects Skeletal Muscle From Programmed Cell Death During Development. Front Cell Dev Biol 8: 622. PubMed ID: 32850788

    Cao, X., He, Y., Hu, Y., Wang, Y., Chen, Y. R., Bryant, B., Clem, R. J., Schwartz, L. M., Blissard, G. and Jiang, H. (2015). The immune signaling pathways of Manduca sexta. Insect Biochem Mol Biol 62: 64-74. PubMed ID: 25858029

    Parelkar, S. S., Cadena, J. G., Kim, C., Wang, Z., Sugal, R., Bentley, B., Moral, L., Ardley, H. C. and Schwartz, L. M. (2012). The parkin-like human homolog of Drosophila ariadne-1 (HHARI) can induce aggresome formation in mammalian cells and is immunologically detectable in Lewy bodies. J Mol Neurosci 46(1): 109-121. PubMed ID: 21590270

  • Tom Schwartz Boston Children's Hospital, Kirby Neurobiology Center
    Zhao, G., Oztan, A., Ye, Y. and Schwarz, T. L. (2019). Kinetochore proteins have a post-mitotic function in neurodevelopment. Dev Cell. PubMed ID: 30827899

    Lee, G. and Schwarz, T.L. (2016). Filamin, a synaptic organizer in Drosophila, determines glutamate receptor composition and membrane growth. Elife [Epub ahead of print]. PubMed ID: 27914199

    Lee, G. and Schwarz, T. L. (2016). Filamin, a synaptic organizer in Drosophila, determines glutamate receptor composition and membrane growth. Elife 5. PubMed ID: 27914199

    Mosca, T. J. and Schwarz, T. L. (2010). Drosophila Importin-alpha2 is involved in synapse, axon and muscle development. PLoS One 5(12): e15223. PubMed ID: 21151903

    Murthy, M., Teodoro, R. O., Miller, T. P. and Schwarz, T. L. (2010). Sec5, a member of the exocyst complex, mediates Drosophila embryo cellularization. Development 137(16): 2773-2783. PubMed ID: 20630948

  • Yuri Schwartz Department of Molecular Biology, Umea University
    Dorafshan, E., Kahn, T. G., Glotov, A., Savitsky, M. and Schwartz, Y. B. (2019). Genetic dissection reveals the role of Ash1 domains in counteracting Polycomb repression. G3 (Bethesda). PubMed ID: 31540973

    Dorafshan, E., Kahn, T. G., Glotov, A., Savitsky, M., Walther, M., Reuter, G. and Schwartz, Y. B. (2019). Ash1 counteracts Polycomb repression independent of histone H3 lysine 36 methylation. EMBO Rep. PubMed ID: 30833342

    Dorafshan, E., Kahn, T. G. and Schwartz, Y. B. (2017). Hierarchical recruitment of Polycomb complexes revisited. Nucleus: 1-10. PubMed ID: 28910569

    Kahn, T. G., Dorafshan, E., Schultheis, D., Zare, A., Stenberg, P., Reim, I., Pirrotta, V. and Schwartz, Y. B. (2016). Interdependence of PRC1 and PRC2 for recruitment to Polycomb response elements. Nucleic Acids Res [Epub ahead of print]. PubMed ID: 27557709

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

    Savitsky, M., Kim, M., Kravchuk, O. and Schwartz, Y. B. (2015). Distinct Roles of Chromatin Insulator Proteins in Control of the Drosophila Bithorax Complex. Genetics. PubMed ID: 26715665

    Riddle, N. C., Jung, Y. L., Gu, T., Alekseyenko, A. A., Asker, D., Gui, H., Kharchenko, P. V., Minoda, A., Plachetka, A., Schwartz, Y. B., Tolstorukov, M. Y., Kuroda, M. I., Pirrotta, V., Karpen, G. H., Park, P. J. and Elgin, S. C. (2012). Enrichment of HP1a on Drosophila chromosome 4 genes creates an alternate chromatin structure critical for regulation in this heterochromatic domain. PLoS Genet 8: e1002954. PubMed ID: 23028361

    Schwartz, Y. B., et al. (2012). Nature and function of insulator protein binding sites in the Drosophila genome. Genome Res 22: 2188-2198. PubMed ID: 22767387

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

    Monck, H., Toppe, D., Michael, E., Sigrist, S., Richter, V., Hilpert, D., Raccuglia, D., Efetova, M. and Schwarzel, M. (2017). A new method to characterize function of the Drosophila heart by means of optical flow. J Exp Biol 220(Pt 24): 4644-4653. PubMed ID: 29237767

    Richlitzki, A., Latour, P. and Schwarzel, M. (2017). Null EPAC mutants reveal a sequential order of versatile cAMP effects during Drosophila aversive odor learning. Learn Mem 24(5): 210-215. PubMed ID: 28416632

    Scholz-Kornehl, S. and Schwarzel, M. (2016). Circuit Analysis of a Drosophila Dopamine Type 2 Receptor That Supports Anesthesia-Resistant Memory. J Neurosci 36: 7936-7945. PubMed ID: 27466338

    Andlauer, T. F., Scholz-Kornehl, S., Tian, R., Kirchner, M., Babikir, H. A., Depner, H., Loll, B., Quentin, C., Gupta, V. K., Holt, M. G., Dipt, S., Cressy, M., Wahl, M. C., Fiala, A., Selbach, M., Schwarzel, M. and Sigrist, S. J. (2014). Drep-2 is a novel synaptic protein important for learning and memory. Elife 3. PubMed ID: 25392983

    Scheunemann, L., Skroblin, P., Hundsrucker, C., Klussmann, E., Efetova, M. and Schwarzel, M. (2013). AKAPS Act in a Two-Step Mechanism of Memory Acquisition. J Neurosci 33: 17422-17428. PubMed ID: 24174675

    Gupta, V. K., Scheunemann, L., Eisenberg, T., Mertel, S., Bhukel, A., Koemans, T. S., Kramer, J. M., Liu, K. S., Schroeder, S., Stunnenberg, H. G., Sinner, F., Magnes, C., Pieber, T. R., Dipt, S., Fiala, A., Schenck, A., Schwaerzel, M., Madeo, F. and Sigrist, S. J. (2013). Restoring polyamines protects from age-induced memory impairment in an autophagy-dependent manner. Nat Neurosci. PubMed ID: 23995066

    Efetova, M., Petereit, L., Rosiewicz, K., Overend, G., Haussig, F., Hovemann, B. T., Cabrero, P., JA, T. D. and Schwarzel, M. (2012). Separate roles of PKA and EPAC in renal function unraveled by the optogenetic control of cAMP levels in vivo. J Cell Sci. PubMed ID: 23264735

    Scheunemann, L., Jost, E., Richlitzki, A., Day, J. P., Sebastian, S., Thum, A. S., Efetova, M., Davies, S. A. and Schwarzel, M. (2012). Consolidated and Labile Odor Memory Are Separately Encoded within the Drosophila Brain. J Neurosci 32: 17163-17171. PubMed ID: 23197709

    Volders, K., Scholz, S., Slabbaert, J. R., Nagel, A. C., Verstreken, P., Creemers, J. W., Callaerts, P. and Schwarzel, M. (2012). Drosophila rugose is a functional homolog of mammalian Neurobeachin and affects synaptic architecture, brain morphology, and associative learning. J Neurosci 32: 15193-15204. PubMed ID: 23100440

  • François Schweisguth Institut Pasteur, Paris
    Shard, C., Luna-Escalante, J. and Schweisguth, F. (2020). Tissue-wide coordination of epithelium-to-neural stem cell transition in the Drosophila optic lobe requires Neuralized. J Cell Biol 219(11). PubMed ID: 32946560

    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

    Trylinski, M. and Schweisguth, F. (2019). Activation of Arp2/3 by WASp is essential for the endocytosis of Delta only during cytokinesis in Drosophila. Cell Rep 28(1): 1-10.e13. PubMed ID: 31269431

    Couturier, L., Mazouni, K., Corson, F. and Schweisguth, F. (2019). Regulation of Notch output dynamics via specific E(spl)-HLH factors during bristle patterning in Drosophila. Nat Commun 10(1): 3486. PubMed ID: 31375669

    Gervais, L., van den Beek, M., Josserand, M., Salle, J., Stefanutti, M., Perdigoto, C. N., Skorski, P., Mazouni, K., Marshall, O. J., Brand, A. H., Schweisguth, F. and Bardin, A. J. (2019). Stem cell proliferation is kept in check by the chromatin regulators Kismet/CHD7/CHD8 and Trr/MLL3/4. Dev Cell 49(4): 556-573.e556. PubMed ID: 31112698

    Andriatsilavo, M., Stefanutti, M., Siudeja, K., Perdigoto, C. N., Boumard, B., Gervais, L., Gillet-Markowska, A., Al Zouabi, L., Schweisguth, F. and Bardin, A. J. (2018). Spen limits intestinal stem cell self-renewal. PLoS Genet 14(11): e1007773. PubMed ID: 30452449

    Perez-Mockus, G., Mazouni, K., Roca, V., Corradi, G., Conte, V. and Schweisguth, F. (2017). Spatial regulation of contractility by Neuralized and Bearded during furrow invagination in Drosophila. Nat Commun 8(1): 1594. PubMed ID: 29150614

    Couturier, L., Mazouni, K., Bernard, F., Besson, C., Reynaud, E. and Schweisguth, F. (2017). Regulation of cortical stability by RhoGEF3 in mitotic sensory organ precursor cells in Drosophila. Biol Open. PubMed ID: 29101098

    Trylinski, M., Mazouni, K. and Schweisguth, F. (2017). Intra-lineage fate decisions involve activation of Notch receptors basal to the midbody in Drosophila sensory organ precursor cells. Curr Biol 27(15): 2239-2247.e2233. PubMed ID: 28736165

    Perez-Mockus, G., Roca, V., Mazouni, K. and Schweisguth, F. (2017). Neuralized regulates Crumbs endocytosis and epithelium morphogenesis via specific Stardust isoforms. J Cell Biol 216(5):1405-1420. PubMed ID: 28400441

    Corson, F., Couturier, L., Rouault, H., Mazouni, K. and Schweisguth, F. (2017). Self-organized Notch dynamics generate stereotyped sensory organ patterns in Drosophila. Science. PubMed ID: 28386027

  • Christof Schwiening Neuroscience, University of Cambridge
    Caldwell, L., Harries, P., Sydlik, S. and Schwiening, C. J. (2013). Presynaptic pH and vesicle fusion in Drosophila larvae neurones. Synapse. PubMed ID: 23649934

    Huang, J., Liu, C. H., Hughes, S. A., Postma, M., Schwiening, C. J. and Hardie, R. C. (2010). Activation of TRP channels by protons and phosphoinositide depletion in Drosophila photoreceptors. Curr Biol 20: 189-197. PubMed ID: 20116246

  • Jeffrey Scott Department of Entomology, Cornell University
    Silva, J. J. and Scott, J. G. (2019). Conservation of the voltage-sensitive sodium channel protein within the Insecta. Insect Mol Biol. PubMed ID: 31206812

    Rinkevich, F. D. and Scott, J. G. (2013). Limitations of RNAi of alpha6 nicotinic acetylcholine receptor subunits for assessing the in vivo sensitivity to spinosad. Insect Sci 20: 101-108. PubMed ID: 23955830

    Lin, G. G. and Scott, J. G. (2011). Investigations of the constitutive overexpression of CYP6D1 in the permethrin resistantLPR strain of house fly (Musca domestica). Pestic Biochem Physiol 100: 130-134. PubMed ID: 21765560

    Lin, G. G., Kozaki, T. and Scott, J. G. (2011). Hormone receptor-like in 96 and Broad-Complex modulate phenobarbital induced transcription of cytochrome P450 CYP6D1 in Drosophila S2 cells. Insect Mol Biol 20: 87-95. PubMed ID: 21029232

  • Kirsten Scott Molecular and Cell Biology, University of California Berkeley
    Yao, Z. and Scott, K. (2022). Serotonergic neurons translate taste detection into internal nutrient regulation. Neuron. PubMed ID: 35051377

    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

    Leinwand, S. G. and Scott, K. (2021). Juvenile hormone drives the maturation of spontaneous mushroom body neural activity and learned behavior. Neuron. PubMed ID: 33915110

    Bidaye, S. S., Laturney, M., Chang, A. K., Liu, Y., Bockemühl, T., Bischges, A. and Scott, K. (2020). Two Brain Pathways Initiate Distinct Forward Walking Programs in Drosophila. Neuron. PubMed ID: 32822613

    Chia, J. and Scott, K. (2020). Activation of specific mushroom body output neurons inhibits proboscis extension and sucrose consumption. PLoS One 15(1): e0223034. PubMed ID: 31990947

    Reisenman, C. E. and Scott, K. (2019). Food-derived volatiles enhance consumption in Drosophila melanogaster. J Exp Biol. PubMed ID: 31085598

    Youn, H., Kirkhart, C., Chia, J. and Scott, K. (2018). A subset of octopaminergic neurons that promotes feeding initiation in Drosophila melanogaster. PLoS One 13(6): e0198362. PubMed ID: 29949586

    Cheung, S. K. and Scott, K. (2017). GABAA receptor-expressing neurons promote consumption in Drosophila melanogaster. PLoS One 12(3): e0175177. PubMed ID: 28362856

    Kim, H., Kirkhart, C. and Scott, K. (2017). Long-range projection neurons in the taste circuit of Drosophila. Elife 6. PubMed ID: 28164781

    Jourjine, N., Mullaney, B.C., Mann, K. and Scott, K. (2016). Coupled sensing of hunger and thirst signals balances sugar and water consumption. Cell [Epub ahead of print]. PubMed ID: 27477513

    Kallman, B. R., Kim, H. and Scott, K. (2015). Excitation and inhibition onto central courtship neurons biases mate choice. Elife 4 [Epub ahead of print]. PubMed ID: 26568316

    Harris, D.T., Kallman, B.R., Mullaney, B.C. and Scott, K. (2015). Representations of taste modality in the Drosophila brain. Neuron [Epub ahead of print]. PubMed ID: 26051423

    Kirkhart, C. and Scott, K. (2015). Gustatory learning and processing in the Drosophila mushroom bodies. J Neurosci 35: 5950-5958. PubMed ID: 25878268

    Pool, A. H. and Scott, K. (2014). Feeding regulation in Drosophila. Curr Opin Neurobiol 29C: 57-63. PubMed ID: 24937262

  • Matthew Scott Stanford University School of Medicine
    Webster, S. H., Vella, M. R. and Scott, M. J. (2020). Development and testing of a novel killer-rescue self-limiting gene drive system in Drosophila melanogaster. Proc Biol Sci 287(1925): 20192994. PubMed ID: 32292114

    Li, F. and Scott, M. J. (2015). CRISPR/Cas9-mediated mutagenesis of the white and Sex lethal loci in the invasive pest, Drosophila suzukii. Biochem Biophys Res Commun. PubMed ID: 26721433

    Hartl, T. A. and Scott, M. P. (2014). Wing Tips: The wing disc as a platform for studying Hedgehog signaling. Methods. PubMed ID: 24556557

    Cao, J., Ni, J., Ma, W., Shiu, V., Milla, L. A., Park, S., Spletter, M. L., Tang, S., Zhang, J., Wei, X., Kim, S. K. and Scott, M. P. (2014). Insight into Insulin Secretion from Transcriptome and Genetic Analysis of Insulin-Producing Cells of Drosophila. Genetics. PubMed ID: 24558258

    Hartl, T. A., Ni, J., Cao, J., Suyama, K. L., Patchett, S., Bussiere, C., Gui, D. Y., Tang, S., Kaplan, D. D., Fish, M., Johnson, A. W., Scott, M. P. (2013) Regulation of Ribosome Biogenesis by Nucleostemin 3 Promotes Local and Systemic Growth in Drosophila. Genetics. PubMed ID: 23436180

    Jeon, M., Scott, M. P. and Zinn, K. (2012). Interactions between Type III receptor tyrosine phosphatases and growth factor receptor tyrosine kinases regulate tracheal tube formation in Drosophila. Biol Open 1: 548-558. PubMed ID: 23213447

    Wang, C., Ma, Z., Scott, M. P. and Huang, X. (2011). The cholesterol trafficking protein NPC1 is required for Drosophila spermatogenesis. Dev Biol 351: 146-155. PubMed ID: 21215267

    Yavari, A., Nagaraj, R., Owusu-Ansah, E., Folick, A., Ngo, K., Hillman, T., Call, G., Rohatgi, R., Scott, M. P. and Banerjee, U. (2010). Role of lipid metabolism in smoothened derepression in hedgehog signaling. Dev Cell 19: 54-65. PubMed ID: 20643350

  • Max Scott Genetics, North Carolina State University, Raleigh
    Fitzsimons, H. L., Schwartz, S., Given, F. M. and Scott, M. J. (2013). The Histone Deacetylase HDAC4 Regulates Long-Term Memory in Drosophila. PLoS One 8: e83903. PubMed ID: 24349558

    Fitzsimons, H. L. and Scott, M. J. (2011). Genetic modulation of Rpd3 expression impairs long-term courtship memory in Drosophila. PLoS One 6: e29171. PubMed ID: 22195015

    Laverty, C., Li, F., Belikoff, E. J. and Scott, M. J. (2011). Abnormal dosage compensation of reporter genes driven by the Drosophila glass multiple reporter (GMR) enhancer-promoter. PLoS One 6: e20455. PubMed ID: 21655213

  • Julie Secombe Albert Einstein College of Medicine, Bronx
    Hatch, H. A. M., Belalcazar, H. M., Marshall, O. J. and Secombe, J. (2021). A KDM5-Prospero transcriptional axis functions during early neurodevelopment to regulate mushroom body formation. Elife 10. PubMed ID: 33729157

    Belalcazar, H. M., Hendricks, E. L., Zamurrad, S., Liebl, F. L. W. and Secombe, J. (2021). The histone demethylase KDM5 is required for synaptic structure and function at the Drosophila neuromuscular junction. Cell Rep 34(7): 108753. PubMed ID: 33596422

    Drelon, C., Belalcazar, H. M. and Secombe, J. (2018). The histone demethylase KDM5 is essential for larval frowth in Drosophila. Genetics. PubMed ID: 29764901

    Zamurrad, S., Hatch, H. A. M., Drelon, C., Belalcazar, H. M. and Secombe, J. (2018). A Drosophila Model of Intellectual Disability Caused by Mutations in the Histone Demethylase KDM5. Cell Rep 22(9): 2359-2369. PubMed ID: 29490272

    Navarro-Costa, P., McCarthy, A., Prudencio, P., Greer, C., Guilgur, L. G., Becker, J. D., Secombe, J., Rangan, P. and Martinho, R. G. (2016). Early programming of the oocyte epigenome temporally controls late prophase I transcription and chromatin remodelling. Nat Commun 7: 12331. PubMed ID: 27507044

    Liu, X., Greer, C. and Secombe, J. (2014). KDM5 interacts with Foxo to modulate cellular levels of oxidative stress. PLoS Genet 10: e1004676. PubMed ID: 25329053

    Liu, X. and Secombe, J. (2015). The Histone demethylase KDM5 activates gene expression by recognizing chromatin context through its PHD reader motif. Cell Rep 13: 2219-2231. PubMed ID: 26673323
    Li, L., Anderson, S., Secombe, J. and Eisenman, R. N. (2013). The Drosophila ubiquitin-specific protease Puffyeye regulates dMyc-mediated growth. Development. PubMed ID: 24173801

    Greer, C., Lee, M., Westerhof, M., Milholland, B., Spokony, R., Vijg, J. and Secombe, J. (2013). Myc-dependent genome instability and lifespan in Drosophila. PLoS One 8: e74641. PubMed ID: 24040302

    Quinn, L. M., Secombe, J. and Hime, G. R. (2013). Myc in stem cell behaviour: insights from Drosophila. Adv Exp Med Biol 786: 269-285. PubMed ID: 23696362

  • Danny Segal Molecular Biology and Biotechnology, Tel Aviv University
    Losev, Y., Frenkel-Pinter, M., Abu-Hussien, M., Viswanathan, G. K., Elyashiv-Revivo, D., Geries, R., Khalaila, I., Gazit, E. and Segal, D. (2020). Differential effects of putative N-glycosylation sites in human Tau on Alzheimer's disease-related neurodegeneration. Cell Mol Life Sci. PubMed ID: 32926180

    Viswanathan, G. K., Shwartz, D., Losev, Y., Arad, E., Shemesh, C., Pichinuk, E., Engel, H., Raveh, A., Jelinek, R., Cooper, I., Gosselet, F., Gazit, E. and Segal, D. (2019). Purpurin modulates Tau-derived VQIVYK fibrillization and ameliorates Alzheimer's disease-like symptoms in animal model. Cell Mol Life Sci. PubMed ID: 31562564

    Shmueli, M. D., Levy-Kanfo, L., Haj, E., Schoenfeld, A. R., Gazit, E. and Segal, D. (2018). Arginine refolds, stabilizes, and restores function of mutant pVHL proteins in animal model of the VHL cancer syndrome. Oncogene. PubMed ID: 30194449

    Frenkel-Pinter, M., Stempler, S., Tal-Mazaki, S., Losev, Y., Singh-Anand, A., Escobar-Alvarez, D., Lezmy, J., Gazit, E., Ruppin, E. and Segal, D. (2017). Altered protein glycosylation predicts Alzheimer's disease and modulates its pathology in disease model Drosophila. Neurobiol Aging. PubMed ID: 28552182

    Dorot, O., Steller, H., Segal, D. and Horowitz, M. (2017). Past1 modulates Drosophila eye development. PLoS One 12(1): e0169639. PubMed ID: 28060904

    Maor, G., Cabasso, O., Krivoruk, O., Rodriguez, J., Steller, H., Segal, D. and Horowitz, M. (2016). The contribution of mutant GBA to the development of parkinson disease in Drosophila. Hum Mol Genet. PubMed ID: 27162249

    Frenkel-Pinter, M., Tal, S., Scherzer-Attali, R., Abu-Hussien, M., Alyagor, I., Eisenbaum, T., Gazit, E. and Segal, D. (2016). Naphthoquinone-Tryptophan Hybrid Inhibits Aggregation of the Tau-Derived Peptide PHF6 and Reduces Neurotoxicity. J Alzheimers Dis. PubMed ID: 26836184

    Shmueli, M. D., Schnaider, L., Herzog, G., Gazit, E. and Segal, D. (2014). Computational and Experimental characterization of dVHL establish a Drosophila model of VHL syndrome. PLoS One 9: e109864. PubMed ID: 25310726

    Saad, Y., Segal, D. and Ayali, A. (2014). Enhanced Neurite Outgrowth and Branching Precede Increased Amyloid-beta-Induced Neuronal Apoptosis in a Novel Alzheimer's Disease Model. J Alzheimers Dis. PubMed ID: 25125474

    Gakamsky, A., Oron, E., Valente, D., Mitra, P. P., Segal, D., Benjamini, Y. and Golani, I. (2013). The angular interval between the direction of progression and body orientation in normal, alcohol- and cocaine treated fruit flies. PLoS One 8: e76257. PubMed ID: 24146845

    Herzog, G., Joerger, A. C., Shmueli, M. D., Fersht, A. R., Gazit, E. and Segal, D. (2012). Evaluating Drosophila p53 as a Model System for Studying Cancer Mutations. J Biol Chem 287: 44330-44337. PubMed ID: 23135266

    Shaltiel-Karyo, R., Davidi, D., Menuchin, Y., Frenkel-Pinter, M., Marcus-Kalish, M., Ringo, J., Gazit, E. and Segal, D. (2012). A novel, sensitive assay for behavioral defects in Parkinson's disease model Drosophila. Parkinsons Dis 2012: 697564. PubMed ID: 22888468

  • Carmen Segarra Departament de Genètica, University of Barcelona
    Calvo-Martin, J. M., Papaceit, M. and Segarra, C. (2017). Evidence of neofunctionalization after the duplication of the highly conserved Polycomb group gene Caf1-55 in the obscura group of Drosophila. Sci Rep 7: 40536. PubMed ID: 28094282

    Calvo-Martin, J. M., Librado, P., Aguade, M., Papaceit, M. and Segarra, C. (2015). Adaptive selection and coevolution at the proteins of the Polycomb repressive complexes in Drosophila. Heredity (Edinb). PubMed ID: 26486609

    Pratdesaba, R., Segarra, C. and Aguade, M. (2015). Inferring the demographic history of Drosophila subobscura from nucleotide variation at regions not affected by chromosomal inversions. Mol Ecol 24: 1729-1741. PubMed ID: 25776124

    Papaceit, M., Segarra, C. and Aguade, M. (2013). Structure and population genetics of the breakpoints of a polymorphic inversion in Drosophila subobscura. Evolution 67: 66-79. PubMed ID: 23289562

    Khadem, M., Munte, A., Camacho, R., Aguade, M. and Segarra, C. (2012). Multilocus analysis of nucleotide variation in Drosophila madeirensis, an endemic species of the Laurisilva forest in Madeira. J Evol Biol 25: 726-739. PubMed ID: 22320146

  • Amita Sehgal Howard Hughes Medical Institute/Department of Neuroscience, University of Pennsylvania, Philadelphia
    Schwarz, J. E., King, A. N., Hsu, C. T., Barber, A. F. and Sehgal, A. (2021). Hugin (+) neurons provide a link between sleep homeostat and circadian clock neurons. Proc Natl Acad Sci U S A 118(47). PubMed ID: 34782479

    Barber, A. F., Fong, S. Y., Kolesnik, A., Fetchko, M. and Sehgal, A. (2021). Drosophila clock cells use multiple mechanisms to transmit time-of-day signals in the brain. Proc Natl Acad Sci U S A 118(10). PubMed ID: 33658368

    Hsu, C. T., Choi, J. T. Y. and Sehgal, A. (2020). Manipulations of the olfactory circuit highlight the role of sensory stimulation in regulating sleep amount. Sleep. PubMed ID: 33313876

    Chouhan, N. S., Griffith, L. C., Haynes, P. and Sehgal, A. (2020). Availability of food determines the need for sleep in memory consolidation. Nature. PubMed ID: 33268891

    Barber, A. F. and Sehgal, A. (2021). Monitoring Electrical Activity in Drosophila Circadian Output Neurons. Methods Mol Biol 2130: 221-232. PubMed ID: 33284448

    Davla, S., Artiushin, G., Li, Y., Chitsaz, D., Li, S., Sehgal, A. and van Meyel, D. J. (2020). AANAT1 functions in astrocytes to regulate sleep homeostasis. Elife 9. PubMed ID: 32955431

    Shakhmantsir, I., Dooley, S. J., Kishore, S., Chen, D., Pierce, E., Bennett, J. and Sehgal, A. (2019). RNA splicing factor mutations that cause retinitis pigmentosa result in circadian dysregulation. J Biol Rhythms: 748730419887876. PubMed ID: 31726916

    Hsieh, A. L., Zheng, X., Yue, Z., Stine, Z. E., Mancuso, A., Rhoades, S. D., Brooks, R., Weljie, A. M., Eisenman, R. N., Sehgal, A. and Dang, C. V. (2019). Misregulation of Drosophila Myc disrupts circadian behavior and metabolism. Cell Rep 29(7): 1778-1788.e1774. PubMed ID: 31722196

    Stone, R. A., McGlinn, A. M., Chakraborty, R., Lee, D. C., Yang, V., Elmasri, A., Landis, E., Shaffer, J., Iuvone, P. M., Zheng, X., Sehgal, A. and Pardue, M. T. (2019). Altered ocular parameters from circadian clock gene disruptions. PLoS One 14(6): e0217111. PubMed ID: 31211778

  • Jeff Sekelsky Department of Biology, University of North Carolina at Chapel Hill
    Deger, N., Cao, X., Selby, C. P., Gulec, S., Kawara, H., Dewey, E. B., Wang, L., Yang, Y., Archibald, S., Selcuk, B., Adebali, O., Sekelsky, J., Sancar, A. and Liu, Z. (2022). CSB-independent, XPC-dependent transcription-coupled repair in Drosophila. Proc Natl Acad Sci U S A 119(9). PubMed ID: 35217627

    Ruchert, J. M., Brady, M. M., McMahan, S., Lacey, K. J., Latta, L. C., Sekelsky, J. and Stoffregen, E. P. (2021). Blm helicase facilitates rapid replication of repetitive DNA sequences in early Drosophila development. Genetics 220(1). PubMed ID: 34849849

    Hatkevich, T., Miller, D. E., Turcotte, C. A., Miller, M. C. and Sekelsky, J. (2021). A pathway for error-free non-homologous end joining of resected meiotic double-strand breaks. Nucleic Acids Res. PubMed ID: 33406239

    Carvajal-Garcia, J., Gales, E. R., Ramsden, D. A. and Sekelsky, J. (2020). The Drosophila melanogaster ortholog of RFWD3 functions independently of RAD51 during DNA repair. G3 (Bethesda). PubMed ID: 31900333

    Hatkevich, T., Boudreau, V., Rubin, T., Maddox, P. S., Huynh, J. R. and Sekelsky, J. (2019). Centromeric SMC1 promotes centromer clustering and stabilizes meiotic homolog pairing. PLoS Genet 15(10): e1008412. PubMed ID: 31609962

    Hartmann, M., Umbanhowar, J. and Sekelsky, J. (2019). Centromere-proximal meiotic crossovers in Drosophila melanogaster are suppressed by both highly-repetitive heterochromatin and proximity to the centromere. Genetics. PubMed ID: 31345993

    Hartmann, M., Kohl, K. P., Sekelsky, J. and Hatkevich, T. (2019). Meiotic MCM proteins promote and inhibit crossovers during meiotic recombination. Genetics. PubMed ID: 31028111

    Bellendir, S. P., Rognstad, D. J., Morris, L. P., Zapotoczny, G., Walton, W. G., Redinbo, M. R., Ramsden, D. A., Sekelsky, J. and Erie, D. A. (2017). Substrate preference of Gen endonucleases highlights the importance of branched structures as DNA damage repair intermediates. Nucleic Acids Res. PubMed ID: 28369583

    Korda Holsclaw, J. and Sekelsky, J. (2017). Annealing of complementary DNA sequences during double-strand break repair in Drosophila is mediated by the ortholog of SMARCAL1. Genetics [Epub ahead of print]. PubMed ID: 28258182

    Hatkevich, T., Kohl, K. P., McMahan, S., Hartmann, M. A., Williams, A. M. and Sekelsky, J. (2016). Bloom syndrome helicase promotes meiotic crossover patterning and homolog disjunction. Curr Biol . PubMed ID: 27989672

    Romero, N. E., Matson, S. W. and Sekelsky, J. (2016). Biochemical activities and genetic functions of the Drosophila melanogaster Fancm nelicase in DNA repair. Genetics [Epub ahead of print]. PubMed ID: 27466228

    Kuo, H. K., McMahan, S., Rota, C. M., Kohl, K. P. and Sekelsky, J. (2014). Drosophila FANCM Helicase Prevents Spontaneous Mitotic Crossovers Generated by the MUS81 and SLX1 Nucleases. Genetics. PubMed ID: 25205745

  • Scott Selleck Biochemistry and Molecular Biology, Penn State University
    Reynolds-Peterson, C. E., Zhao, N., Xu, J., Serman, T. M., Xu, J. and Selleck, S. B. (2017). Heparan sulfate proteoglycans regulate autophagy in Drosophila. Autophagy: 0. PubMed ID: 28402693

    Lee, H. G., Zhao, N., Campion, B. K., Nguyen, M. M., Selleck, S. B. (2013) Akt regulates glutamate receptor trafficking and postsynaptic membrane elaboration at the Drosophila neuromuscular junction. Dev Neurobiol. PubMed ID: 23592328

    Dimitroff, B., Howe, K., Watson, A., Campion, B., Lee, H. G., Zhao, N., O'Connor, M. B., Neufeld, T. P. and Selleck, S. B. (2012). Diet and energy-sensing inputs affect TorC1-mediated axon misrouting but not TorC2-directed synapse growth in a Drosophila model of tuberous sclerosis. PLoS One 7: e30722. PubMed ID: 22319582

    Ren, Y., Kirkpatrick, C. A., Rawson, J. M., Sun, M. and Selleck, S. B. (2009). Cell type-specific requirements for heparan sulfate biosynthesis at the Drosophila neuromuscular junction: effects on synapse function, membrane trafficking, and mitochondrial localization. J Neurosci 29: 8539-8550. PubMed ID: 19571145

  • James Sellers National Heart, Lung and Blood Institute, NIH, Bethesda, Md
    Liu, R., Billington, N., Yang, Y., Bond, C., Hong, A., Siththanandan, V., Takagi, Y. and Sellers, J. R. (2021). A binding protein regulates myosin-7a dimerization and actin bundle assembly. Nat Commun 12(1): 563. PubMed ID: 33495456

    Nishimura, Y., Shi, S., Zhang, F., Liu, R., Takagi, Y., Bershadsky, A. D., Viasnoff, V. and Sellers, J. R. (2021). The Formin Inhibitor, SMIFH2, Inhibits Members of the Myosin Superfamily. J Cell Sci. PubMed ID: 33589498

    Liu, R., Billington, N., Yang, Y., Bond, C., Hong, A., Siththanandan, V., Takagi, Y. and Sellers, J. R. (2021). A binding protein regulates myosin-7a dimerization and actin bundle assembly. Nat Commun 12(1): 563. PubMed ID: 33495456

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

  • Mihaela Serpe NICHD, NIH, Bethesda
    Vicidomini, R., Nguyen, T. H., Choudhury, S. D., Brody, T. and Serpe, M. (2021). Assembly and Exploration of a Single Cell Atlas of the Drosophila Larval Ventral Cord. Identification of Rare Cell Types. Curr Protoc 1(2): e37. PubMed ID: 33600085

    Nguyen, T. H., Vicidomini, R., Choudhury, S. D., Coon, S. L., Iben, J., Brody, T. and Serpe, M. (2021). Single-Cell RNA Sequencing Analysis of the Drosophila Larval Ventral Cord. Curr Protoc 1(2): e38. PubMed ID: 33620770

    Nguyen, T. H., Han, T. H., Newfeld, S. J. and Serpe, M. (2020). Selective Disruption of Synaptic BMP Signaling by a Smad Mutation Adjacent to the Highly Conserved H2 Helix. Genetics. PubMed ID: 32737119

    Han, T. H., Vicidomini, R., Ramos, C. I., Wang, Q., Nguyen, P., Jarnik, M., Lee, C. H., Stawarski, M., Hernandez, R. X., Macleod, G. T. and Serpe, M. (2020). Neto-alpha Controls Synapse Organization and Homeostasis at the Drosophila Neuromuscular Junction. Cell Rep 32(1): 107866. PubMed ID: 32640231

    Wang, Q., Han, T. H., Nguyen, P., Jarnik, M. and Serpe, M. (2018). Tenectin recruits integrin to stabilize bouton architecture and regulate vesicle release at the Drosophila neuromuscular junction. Elife 7. PubMed ID: 29901439

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

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

    Han, T.H., Dharkar, P., Mayer, M.L. and Serpe, M. (2015). Functional reconstitution of Drosophila melanogaster NMJ glutamate receptors. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 25918369

    Ramos, C. I., Igiesuorobo, O., Wang, Q. and Serpe, M. (2015). Neto-mediated intracellular interactions shape postsynaptic composition at the Drosophila neuromuscular junction. PLoS Genet 11: e1005191. PubMed ID: 25905467

    Kim, Y.J., Igiesuorobo, O., Ramos, C.I., Bao, H., Zhang, B. and Serpe, M. (2015). Prodomain removal enables Neto to stabilize glutamate receptors at the Drosophila neuromuscular junction. PLoS Genet 11: e1004988. PubMed ID: 25723514

    Sulkowski, M., Kim, Y. J. and Serpe, M. (2013). Postsynaptic glutamate receptors regulate local BMP signaling at the Drosophila neuromuscular junction. Development. PubMed ID: 24353060

  • Florenci Serras Departament de Genètica, University of Barcelona
    Esteban-Collado, J., Corominas, M. and Serras, F. (2021). Nutrition and PI3K/Akt signaling are required for p38-dependent regeneration. Development 148(8). PubMed ID: 33913483

    Santabarbara-Ruiz, P., Esteban-Collado, J., Perez, L., Viola, G., Abril, J. F., Milan, M., Corominas, M. and Serras, F. (2019). Ask1 and Akt act synergistically to promote ROS-dependent regeneration in Drosophila. PLoS Genet 15(1): e1007926. PubMed ID: 30677014

    Santabarbara-Ruiz, P., Lopez-Santillan, M., Martinez-Rodriguez, I., Binagui-Casas, A., Perez, L., Milan, M., Corominas, M. and Serras, F. (2015). ROS-induced JNK and p38 signaling is required for Unpaired cytokine activation during Drosophila regeneration. PLoS Genet 11: e1005595. PubMed ID: 26496642

    Ruiz-Romero, M., Blanco, E., Paricio, N., Serras, F. and Corominas, M. (2015). Cabut/dTIEG associates with the transcription factor Yorkie for growth control. EMBO Rep. PubMed ID: 25572844

    Ruiz, P. S. and Serras, F. (2014). Mind the gap: Cells respond to tissue damage by changing orientation of cell divisions. Fly (Austin) 8. PubMed ID: 24406337

    Hombria, J. C. and Serras, F. (2013). Why should we care about fly tumors?: The case of JAK-STAT and EGFR cooperation in oncogenesis. JAKSTAT 2: e23203. PubMed ID: 24058803

    Repiso, A., Bergantinos, C. and Serras, F. (2013). Cell fate respecification and cell division orientation drive intercalary regeneration in Drosophila wing discs. Development. PubMed ID: 23903186

    Mora, N., Santa Barbara, P., Ferreira, N. and Serras, F. (2013). Ras signal triggers beta-Amyloid Precursor Protein (APP) expression. Small GTPases 4. PubMed ID: 23648941

    Mora, N., Almudi, I., Alsina, B., Corominas, M. and Serras, F. (2012). The ss-amyloid protein precursor like (Appl) is a Ras1/MAPK regulated gene required for axonal targeting in Drosophila photoreceptor neurons. J Cell Sci. PubMed ID: 23178937

    Repiso, A., Bergantinos, C., Corominas, M. and Serras, F. (2011). Tissue repair and regeneration in Drosophila imaginal discs. Dev Growth Differ 53: 177-185. PubMed ID: 21338344

  • Carla Sgro Biological Sciences, Monash University, Victoria, Australia
    van Heerwaarden, B. and Sgro, C. M. (2021). Male fertility thermal limits predict vulnerability to climate warming. Nat Commun 12(1): 2214. PubMed ID: 33850157

    Kellermann, V., van Heerwaarden, B. and Sgro, C. M. (2017). How important is thermal history? Evidence for lasting effects of developmental temperature on upper thermal limits in Drosophila melanogaster. Proc Biol Sci 284(1855). PubMed ID: 28539515

    Telonis-Scott, M., Clemson, A. S., Johnson, T. K. and Sgro, C. M. (2014). Spatial analysis of gene regulation reveals new insights into the molecular basis of upper thermal limits. Mol Ecol. PubMed ID: 25401770

    Telonis-Scott, M., van Heerwaarden, B., Johnson, T. K., Hoffmann, A. A. and Sgro, C. M. (2013). New Levels of Transcriptome Complexity at Upper Thermal Limits in Wild Drosophila Revealed by Exon Expression Analysis. Genetics. PubMed ID: 24002645

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

    van Heerwaarden, B. and Sgro, C. M. (2013). Multivariate analysis of adaptive capacity for upper thermal limits in Drosophila simulans. J Evol Biol 26: 800-809. PubMed ID: 23517493

    Bechsgaard, J. S., Hoffmann, A. A., Sgro, C., Loeschcke, V., Bilde, T. and Kristensen, T. N. (2013). A comparison of inbreeding depression in tropical and widespread Drosophila species. PLoS One 8: e51176. PubMed ID: 23460779

  • Olie Shafer Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor
    Fernandez, M. P., Pettibone, H. L., Bogart, J. T., Roell, C. J., Davey, C. E., Pranevicius, A., Huynh, K. V., Lennox, S. M., Kostadinov, B. S. and Shafer, O. T. (2020). Sites of Circadian Clock Neuron Plasticity Mediate Sensory Integration and Entrainment. Curr Biol. PubMed ID: 32386535

    Hsu, I. U., Linsley, J. W., Varineau, J. E., Shafer, O. T. and Kuwada, J. Y. (2018). Dstac is required for normal circadian activity rhythms in Drosophila. Chronobiol Int: 1-11. PubMed ID: 29621409

    Yadlapalli, S., Jiang, C., Bahle, A., Reddy, P., Meyhofer, E. and Shafer, O. T. (2018). Circadian clock neurons constantly monitor environmental temperature to set sleep timing. Nature 555(7694): 98-102. PubMed ID: 29466329

    Abruzzi, K. C., Zadina, A., Luo, W., Wiyanto, E., Rahman, R., Guo, F., Shafer, O. and Rosbash, M. (2017). RNA-seq analysis of Drosophila clock and non-clock neurons reveals neuron-specific cycling and novel candidate neuropeptides. PLoS Genet 13(2): e1006613. PubMed ID: 28182648

    Yao, Z., Bennett, A.J., Clem, J.L. and Shafer, O.T. (2016). The Drosophila clock neuron network features diverse coupling modes and requires network-wide coherence for robust circadian rhythms. Cell Rep 17: 2873-2881. PubMed ID: 27974202

    Schlichting, M., Menegazzi, P., Lelito, K. R., Yao, Z., Buhl, E., Dalla Benetta, E., Bahle, A., Denike, J., Hodge, J. J., Helfrich-Forster, C. and Shafer, O. T. (2016). A neural network underlying circadian entrainment and photoperiodic adjustment of sleep and activity in Drosophila. J Neurosci 36: 9084-9096. PubMed ID: 27581451

    Oh, Y., Yoon, S. E., Zhang, Q., Chae, H. S., Daubnerova, I., Shafer, O. T., Choe, J. and Kim, Y. J. (2014). A homeostatic sleep-stabilizing pathway in Drosophila composed of the sex Peptide receptor and its ligand, the myoinhibitory Peptide. PLoS Biol 12: e1001974. PubMed ID: 25333796

    Yao, Z. and Shafer, O. T. (2014). The Drosophila circadian clock is a variably coupled network of multiple peptidergic units. Science 343: 1516-1520. PubMed ID: 24675961

    Talsma, A. D., Christov, C. P., Terriente-Felix, A., Linneweber, G. A., Perea, D., Wayland, M., Shafer, O. T. and Miguel-Aliaga, I. (2012). Remote control of renal physiology by the intestinal neuropeptide pigment-dispersing factor in Drosophila. Proc Natl Acad Sci U S A 109: 12177-12182. PubMed ID: 22778427

    Yao, Z., Macara, A. M., Lelito, K. R., Minosyan, T. Y. and Shafer, O. T. (2012). Analysis of functional neuronal connectivity in the Drosophila brain. J Neurophysiol 108: 684-696. PubMed ID: 22539819

  • Parvin Shahrestani Department of Biological Science California State University, Fullerton
    Phillips, M. A., Rutledge, G. A., Kezos, J. N., Greenspan, Z. S., Talbott, A., Matty, S., Arain, H., Mueller, L. D., Rose, M. R. and Shahrestani, P. (2018). Effects of evolutionary history on genome wide and phenotypic convergence in Drosophila populations. BMC Genomics 19(1): 743. PubMed ID: 30305018

    Shahrestani, P., Burke, M. K., Birse, R., Kezos, J. N., Ocorr, K., Mueller, L. D., Rose, M. R. and Bodmer, R. (2017). Experimental Evolution and Heart Function in Drosophila. Physiol Biochem Zool 90(2): 281-293. PubMed ID: 28277957

    Mueller, L. D., Shahrestani, P., Rauser, C. L. and Rose, M. R. (2016). The death spiral: predicting death in Drosophila cohorts. Biogerontology 17(5-6): 805-816. PubMed ID: 26914589

  • Anurag Sharma Indian Institute of Toxicology Research Lucknow, Uttar Pradesh, India
    D'Souza, L. C., Dwivedi, S., Raihan, F., Yathisha, U. G., Raghu, S. V., Mamatha, B. S. and Sharma, A. (2022). Hsp70 overexpression in Drosophila hemocytes attenuates benzene-induced immune and developmental toxicity via regulating ROS/JNK signaling pathway. Environ Toxicol. PubMed ID: 35301792

    Vimal, D., D'Souza, L. C., Rai, V., Lal, S., Sharma, A. and Gupta, S. C. (2022). Efficacy of cannabis and its constituents in disease management: Insights from clinical studies. Curr Med Chem. PubMed ID: 35619266

    D'Souza, L. C., Dwivedi, S., Raihan, F., Yathisha, U. G., Raghu, S. V., Mamatha, B. S. and Sharma, A. (2022). Hsp70 overexpression in Drosophila hemocytes attenuates benzene-induced immune and developmental toxicity via regulating ROS/JNK signaling pathway. Environ Toxicol 37(7): 1723-1739. PubMed ID: 35301792

    Paithankar, J. G., Kushalan, S., S, N., Hegde, S., Kini, S. and Sharma, A. (2022). Systematic toxicity assessment of CdTe quantum dots in Drosophila melanogaster. Chemosphere 295: 133836. PubMed ID: 35120950


  • Vijay Kumar Sharma Chronobiology Laboratory, Evolutionary & Organismal Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Indis
    Lone, S. R., Potdar, S., Venkataraman, A., Sharma, N., Kulkarni, R., Rao, S., Mishra, S., Sheeba, V. and Sharma, V. K. (2021). Mechanosensory Stimulation via Nanchung Expressing Neurons Can Induce Daytime Sleep in Drosophila. J Neurosci. PubMed ID: 34635540

    Abhilash, L. and Sharma, V. K. (2020). Mechanisms of photic entrainment of activity/rest rhythms in populations of Drosophila selected for divergent timing of eclosion. Chronobiol Int: 1-16. PubMed ID: 32079418

    Varma, V., Krishna, S., Srivastava, M., Sharma, V. K. and Sheeba, V. (2019). Accuracy of fruit-fly eclosion rhythms evolves by strengthening circadian gating rather than developmental fine-tuning. Biol Open 8(8). PubMed ID: 31455663

    Srivastava, M., Varma, V., Abhilash, L., Sharma, V. K. and Sheeba, V. (2019). Circadian clock properties and their relationships as a function of free-running period in Drosophila melanogaster. J Biol Rhythms: 748730419837767. PubMed ID: 30939971

    Shindey, R., Varma, V., Nikhil, K. L. and Sharma, V. K. (2017). Evolution of circadian rhythms in Drosophila melanogaster populations reared in constant light and dark regimes for over 330 generations. Chronobiol Int: 1-14. PubMed ID: 28156168

    Shindey, R., Varma, V., Nikhil, K. L. and Sharma, V. K. (2016). Evolution of robust circadian clocks in Drosophila melanogaster populations reared in constant dark for over 330 generations. Naturwissenschaften 103: 74. PubMed ID: 27585442

    Nikhil, K. L., Ratna, K. and Sharma, V. K. (2016). Life-history traits of Drosophila melanogaster populations exhibiting early and late eclosion chronotypes. BMC Evol Biol 16: 46. PubMed ID: 26922082

    Lone, S.R., Potdar, S., Srivastava, M. and Sharma, V.K. (2016). Social experience is sufficient to modulate sleep need of Drosophila without increasing wakefulness. PLoS One 11: e0150596. PubMed ID: 26938057

    Nikhil, K. L., Abhilash, L. and Sharma, V. K. (2016). Molecular correlates of circadian clocks in fruit fly Drosophila melanogaster populations exhibiting early and late emergence chronotypes. J Biol Rhythms [Epub ahead of print]. PubMed ID: 26833082

    Nikhil, K. L., Vaze, K. M. and Sharma, V. K. (2015). Late emergence chronotypes of fruit flies Drosophila melanogaster exhibit higher accuracy of entrainment. Chronobiol Int 32: 1477-1485. PubMed ID: 26595175

    Nikhil, K.L., Vaze, K.M., Ratna, K. and Sharma, V.K. (2015). Circadian clock properties of fruit flies Drosophila melanogaster exhibiting early and late emergence chronotypes. Chronobiol Int [Epub ahead of print]. PubMed ID: 26654995

    Lone, S.R., Venkataraman, A., Srivastava, M., Potdar, S. and Sharma, V.K. (2015). Or47b-neurons promote male-mating success in Drosophila. Biol Lett 11(5) [Epub ahead of print]. PubMed ID: 26018835

  • L. S. Shashidhara Indian Institute of Science Education and Research, Pune
    Nagarkar, S., Wasnik, R., Govada, P., Cohen, S. and Shashidhara, L. S. (2020). Promoter Proximal Pausing Limits Tumorous Growth Induced by the Yki Transcription Factor in Drosophila. Genetics. PubMed ID: 32737120

    Shukla, J. P., Deshpande, G. and Shashidhara, L. S. (2017). Ataxin 2-binding protein 1 is a context-specific positive regulator of Notch signaling during neurogenesis in Drosophila melanogaster. Development 144(5): 905-915. PubMed ID: 28174239

    Prasad, N. and Shashidhara, L. S. (2015). Gliolectin positively regulates Notch signalling during wing-vein specification in Drosophila. Int J Dev Biol 59(4-6): 187-194. PubMed ID: 26505251

    Singh, S., Sanchez-Herrero, E. and Shashidhara, L. S. (2015). Critical role for Fat/Hippo and IIS/Akt pathways downstream of Ultrabithorax during haltere specification in Drosophila. Mech Dev 138 Pt 2: 198-209. PubMed ID: 26299254

  • Paul Shaw Division of Biology and Biomedical Sciences, Washington University, St. Louis
    Tainton-Heap, L. A. L., Kirszenblat, L. C., Notaras, E. T., Grabowska, M. J., Jeans, R., Feng, K., Shaw, P. J. and van Swinderen, B. (2020). A Paradoxical Kind of Sleep in Drosophila melanogaster. Curr Biol. PubMed ID: 33238155

    Melnattur, K., Morgan, E., Duong, V., Kalra, A. and Shaw, P. J. (2020). The Sleep Nullifying Apparatus: A Highly Efficient Method of Sleep Depriving Drosophila. J Vis Exp(166). PubMed ID: 33369606

    Melnattur, K., Kirszenblat, L., Morgan, E., Militchin, V., Sakran, B., English, D., Patel, R., Chan, D., van Swinderen, B. and Shaw, P. J. (2020). A conserved role for sleep in supporting Spatial Learning in Drosophila. Sleep. PubMed ID: 32959053

    Melnattur, K., Zhang, B. and Shaw, P. J. (2020). Disrupting flight increases sleep and identifies a novel sleep-promoting pathway in Drosophila. Sci Adv 6(19): eaaz2166. PubMed ID: 32494708

    Kirszenblat, L., Ertekin, D., Goodsell, J., Zhou, Y., Shaw, P. J. and van Swinderen, B. (2018). Sleep regulates visual selective attention in Drosophila. J Exp Biol. PubMed ID: 30355611

    Baggett, V., Mishra, A., Kehrer, A. L., Robinson, A. O., Shaw, P. and Zars, T. (2018). Place learning overrides innate behaviors in Drosophila. Learn Mem 25(3): 122-128. PubMed ID: 29449456 Yap, M. H. W., Grabowska, M. J., Rohrscheib, C., Jeans, R., Troup, M., Paulk, A. C., van Alphen, B., Shaw, P. J. and van Swinderen, B. (2017). Oscillatory brain activity in spontaneous and induced sleep stages in flies. Nat Commun 8(1): 1815. PubMed ID: 29180766

    Seugnet, L., Dissel, S., Thimgan, M., Cao, L. and Shaw, P. J. (2017). Identification of genes that maintain behavioral and structural plasticity during sleep loss. Front Neural Circuits 11: 79. PubMed ID: 29109678

    Dissel, S., Klose, M., Donlea, J., Cao, L., English, D., Winsky-Sommerer, R., van Swinderen, B. and Shaw, P. J. (2017). Enhanced sleep reverses memory deficits and underlying pathology in Drosophila models of Alzheimer's disease. Neurobiol Sleep Circadian Rhythms 2: 15-26. PubMed ID: 29094110

    Hautbergue, G. M., Castelli, L. M., Ferraiuolo, L., Sanchez-Martinez, A., Cooper-Knock, J., Higginbottom, A., Lin, Y. H., Bauer, C. S., Dodd, J. E., Myszczynska, M. A., Alam, S. M., Garneret, P., Chandran, J. S., Karyka, E., Stopford, M. J., Smith, E. F., Kirby, J., Meyer, K., Kaspar, B. K., Isaacs, A. M., El-Khamisy, S. F., De Vos, K. J., Ning, K., Azzouz, M., Whitworth, A. J. and Shaw, P. J. (2017). SRSF1-dependent nuclear export inhibition of C9ORF72 repeat transcripts prevents neurodegeneration and associated motor deficits. Nat Commun 8: 16063. PubMed ID: 28677678

  • Halyna Shcherbata Max-Planck-Institute Fur Biophysikalische Chemie, Gottingen
    Karki, P., Carney, T. D., Maracci, C., Yatsenko, A. S., Shcherbata, H. R. and Rodnina, M. V. (2021). Tissue-specific regulation of translational readthrough tunes functions of the traffic jam transcription factor. Nucleic Acids Res. PubMed ID: 34897510

    Melentev, P. A., Ryabova, E. V., Surina, N. V., Zhmujdina, D. R., Komissarov, A. E., Ivanova, E. A., Boltneva, N. P., Makhaeva, G. F., Sliusarenko, M. I., Yatsenko, A. S., Mohylyak, II, Matiytsiv, N. P., Shcherbata, H. R. and Sarantseva, S. V. (2021). Loss of swiss cheese in Neurons Contributes to Neurodegeneration with Mitochondria Abnormalities, Reactive Oxygen Species Acceleration and Accumulation of Lipid Droplets in Drosophila Brain. Int J Mol Sci 22(15). PubMed ID: 34361042

    Ryabova, E. V., Melentev, P. A., Komissarov, A. E., Surina, N. V., Ivanova, E. A., Matiytsiv, N., Shcherbata, H. R. and Sarantseva, S. V. (2021). Morpho-Functional Consequences of Swiss Cheese Knockdown in Glia of Drosophila melanogaster. Cells 10(3). PubMed ID: 33801404

    Yatsenko, A. S. and Shcherbata, H. R. (2021). Distant activation of Notch signaling induces stem cell niche assembly. PLoS Genet 17(3): e1009489. PubMed ID: 33780456

    Yatsenko, A. S., Kucherenko, M. M., Xie, Y., Urlaub, H. and Shcherbata, H. R. (2021). Exocyst-mediated membrane trafficking of the lissencephaly-associated ECM receptor dystroglycan is required for proper brain compartmentalization. Elife 10. PubMed ID: 33620318

    Yatsenko, A. S., Kucherenko, M. M., Xie, Y., Aweida, D., Urlaub, H., Scheibe, R. J., Cohen, S. and Shcherbata, H. R. (2020). Profiling of the muscle-specific dystroglycan interactome reveals the role of Hippo signaling in muscular dystrophy and age-dependent muscle atrophy. BMC Med 18(1): 8. PubMed ID: 31959160

    Yatsenko, A. S. and Shcherbata, H. R. (2018). Stereotypical architecture of the stem cell niche is spatiotemporally established by miR-125-dependent coordination of Notch and steroid signaling. Development. PubMed ID: 29361571

    Kucherenko, M. M., Ilangovan, V., Herzig, B., Shcherbata, H. R. and Bringmann, H. (2016). TfAP-2 is required for night sleep in Drosophila. BMC Neurosci 17: 72. PubMed ID: 27829368

    Çiçek, I.Ö., Karaca, S., Brankatschk, M., Eaton, S., Urlaub, H. and Shcherbata, H.R. (2016). Hedgehog signaling strength is orchestrated by the mir-310 cluster of microRNAs in response to diet. Genetics [Epub ahead of print]. PubMed ID: 26801178

    König, A. and Shcherbata, H.R. (2015). Soma influences GSC progeny differentiation via the cell adhesion-mediated steroid-let-7-Wingless signaling cascade that regulates chromatin dynamics. Biol Open [Epub ahead of print]. PubMed ID: 25661868

    Yatsenko, A. S., Marrone, A. K. and Shcherbata, H. R. (2014). miRNA-based buffering of the cobblestone-lissencephaly-associated extracellular matrix receptor dystroglycan via its alternative 3'-UTR. Nat Commun 5: 4906. PubMed ID: 25232965

  • Vasu Sheeba Jawaharlal Nehru Centre for Advanced Scientific Research, Evolutionary and Organismal Biology Unit, Jakkur
    Iyengar, A. S., Kulkarni, R. and Sheeba, V. (2022). Under warm ambient conditions, Drosophila melanogaster suppresses nighttime activity via the neuropeptide pigment dispersing factor. Genes Brain Behav 21(4): e12802. PubMed ID: 35285135

    Iyengar, A. S., Kulkarni, R. and Sheeba, V. (2022). Under warm ambient conditions, Drosophila melanogaster suppresses nighttime activity via the neuropeptide pigment dispersing factor. Genes Brain Behav 21(4): e12802. PubMed ID: 35285135

    Ramakrishnan, A. and Sheeba, V. (2021). Gap junction protein Innexin2 modulates the period of free-running rhythms in Drosophila melanogaster. iScience 24(9): 103011. PubMed ID: 34522854

    Ghosh, A., Sharma, P., Dansana, S. and Sheeba, V. (2021). Evidence for Co-Evolution of Masking With Circadian Phase in Drosophila Melanogaster. J Biol Rhythms 36(3): 254-270. PubMed ID: 33752486

    Abhilash, L., Kalliyil, A. and Sheeba, V. (2020). Responses of activity rhythms to temperature cues evolve in Drosophila populations selected for divergent timing of eclosion. J Exp Biol. PubMed ID: 32291322

    Abhilash, L., Ghosh, A. and Sheeba, V. (2019). Selection for timing of eclosion results in co-evolution of temperature responsiveness in Drosophila melanogaster. J Biol Rhythms: 748730419877315. PubMed ID: 31608742

    Varma, V., Krishna, S., Srivastava, M., Sharma, V. K. and Sheeba, V. (2019). Accuracy of fruit-fly eclosion rhythms evolves by strengthening circadian gating rather than developmental fine-tuning. Biol Open 8(8). PubMed ID: 31455663

    Srivastava, M., James, A., Varma, V., Sharma, V. K. and Sheeba, V. (2018). Environmental cycles regulate development time via circadian clock mediated gating of adult emergence. BMC Dev Biol 18(1): 21. PubMed ID: 30577765

    Potdar, S. and Sheeba, V. (2018). Wakefulness is promoted during day time by PDFR signalling to dopaminergic neurons in Drosophila melanogaster. eNeuro 5(4). PubMed ID: 30131970

    Potdar, S., Daniel, D. K., Thomas, F. A., Lall, S. and Sheeba, V. (2018). Sleep deprivation negatively impacts reproductive output in Drosophila melanogaster. J Exp Biol. PubMed ID: 29361608

    Prakash, P., Nambiar, A. and Sheeba, V. (2017). Oscillating PDF in termini of circadian pacemaker neurons and synchronous molecular clocks in downstream neurons are not sufficient for sustenance of activity rhythms in constant darkness. PLoS One 12(5): e0175073. PubMed ID: 28558035

  • Jingshi Shen MCDB Department, University of Colorado, Boulder
    Fan, Z., Zhang, J., Wang, D. and Shen, J. (2021). T-box transcription factors Dorsocross and optomotor-blind control Drosophila leg patterning in a functionally redundant manner. Insect Biochem Mol Biol 129: 103516. PubMed ID: 33412239

    Ai, X., Wang, D., Zhang, J. and Shen, J. (2020). Hippo signaling promotes Ets21c-dependent apical cell extrusion in the Drosophila wing disc. Development 147(22). PubMed ID: 33028612

    Sun, J., Zhang, J., Wang, D. and Shen, J. (2020). The transcription factor spalt and human homologue SALL4 induce cell invasion via the dMyc-JNK pathway in Drosophila. Biol Open. PubMed ID: 32098783

    Fan, W. M., Luo, D., Zhang, J. Z., Wang, D. and Shen, J. (2020). Vestigial suppresses apoptosis and cell migration in a manner dependent on the level of JNK/Caspase signaling in the Drosophila wing disc. Insect Sci. PubMed ID: 32037698

    Kang, J., Shin, S., Perrimon, N. and Shen, J. (2017). An evolutionarily conserved role of presenilin in neuronal protection in the aging Drosophila brain. Genetics [Epub ahead of print]. PubMed ID: 28495961

    Wang, D., Li, J., Liu, S., Zhou, H., Zhang, L., Shi, W. and Shen, J. (2017). spalt is functionally conserved in Locusta and Drosophila to promote wing growth. Sci Rep 7: 44393. PubMed ID: 28300136

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

    Tang, W., Wang, D. and Shen, J. (2016). Asymmetric distribution of Spalt in Drosophila wing squamous and columnar epithelia ensures correct cell morphogenesis. Sci Rep 6: 30236. PubMed ID: 27452716

    Wang, D., Li, L., Lu, J., Liu, S. and Shen, J. (2016). Complementary expression of optomotor-blind and the Iroquois complex promotes fold formation to separate wing notum and hinge territories. Dev Biol. PubMed ID: 27212024

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

  • Ping Shen Cellular Biology, University of Georgia

    Pu, Y., Zhang, Y., Zhang, Y. and Shen, P. (2018). Two Drosophila Neuropeptide Y-like neurons define a reward module for transforming appetitive odor representations to motivation. Sci Rep 8(1): 11658. PubMed ID: 30076343

    Pu, Y., Palombo, M. M. M. and Shen, P. (2018). Contribution of DA signaling to appetitive odor perception in a Drosophila model. Sci Rep 8(1): 5978. PubMed ID: 29654277

    Zhang, T., Branch, A. and Shen, P. (2013). Octopamine-mediated circuit mechanism underlying controlled appetite for palatable food in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 24003139

    Wang, Y., Pu, Y., Shen, P. (2013) Neuropeptide-Gated Perception of Appetitive Olfactory Inputs in Drosophila Larvae. Cell Rep. PubMed ID: 23453968

    Chen, J., Wang, Y., Zhang, Y. and Shen, P. (2013). Mutations in Bacchus reveal a tyramine-dependent nuclear regulator for acute ethanol sensitivity in Drosophila. Neuropharmacology 67: 25-31. PubMed ID: 23142736

    Xu, J., Li, M. and Shen, P. (2010). A G-protein-coupled neuropeptide Y-like receptor suppresses behavioral and sensory response to multiple stressful stimuli in Drosophila. J Neurosci 30: 2504-2512. PubMed ID: 20164335

    Chen, J., Zhang, Y. and Shen, P. (2010). Protein kinase C deficiency-induced alcohol insensitivity and underlying cellular targets in Drosophila. Neuroscience 166: 34-39. PubMed ID: 20006676


  • Nina Sherwood Department of Biology, Duke University
    Baxter, S. L., Allard, D. E., Crowl, C. and Sherwood, N. T. (2014). Cold temperature improves mobility and survival in drosophila models of Autosomal-Dominant Hereditary Spastic Paraplegia (AD-HSP). Dis Model Mech. PubMed ID: 24906373

    Stewart, A., Tsubouchi, A., Rolls, M. M., Tracey, W. D. and Sherwood, N. T. (2012). Katanin p60-like1 promotes microtubule growth and terminal dendrite stability in the larval class IV sensory neurons of Drosophila. J Neurosci 32: 11631-11642. PubMed ID: 22915107

    Stone, M. C., Rao, K., Gheres, K. W., Kim, S., Tao, J., La Rochelle, C., Folker, C. T., Sherwood, N. T. and Rolls, M. M. (2012). Normal spastin gene dosage is specifically required for axon regeneration. Cell Rep 2: 1340-1350. PubMed ID: 23122959

    Ozdowski, E. F., Gayle, S., Bao, H., Zhang, B. and Sherwood, N. T. (2011). Loss of Drosophila melanogaster p21-activated kinase 3 suppresses defects in synapse structure and function caused by spastin mutations. Genetics 189: 123-135. PubMed ID: 21705760

  • Yuri Shevelyov Yasenovich Institute of Molecular Genetics of National Research Centre Kurchatov Institute, Moscow
    Ilyin, A. A., Kononkova, A. D., Golova, A. V., Shloma, V. V., Olenkina, O. M., Nenasheva, V. V., Abramov, Y. A., Kotov, A. A., Maksimov, D. A., Laktionov, P. P., Pindyurin, A. V., Galitsyna, A. A., Ulianov, S. V., Khrameeva, E. E., Gelfand, M. S., Belyakin, S. N., Razin, S. V. and Shevelyov, Y. Y. (2022). Comparison of genome architecture at two stages of male germline cell differentiation in Drosophila. Nucleic Acids Res 50(6): 3203-3225. PubMed ID: 35166842

    >Ulianov, S. V., Zakharova, V. V., Galitsyna, A. A., Kos, P. I., Polovnikov, K. E., Flyamer, I. M., Mikhaleva, E. A., Khrameeva, E. E., Germini, D., Logacheva, M. D., Gavrilov, A. A., Gorsky, A. S., Nechaev, S. K., Gelfand, M. S., Vassetzky, Y. S., Chertovich, A. V., Shevelyov, Y. Y. and Razin, S. V. (2021). Order and stochasticity in the folding of individual Drosophila genomes. Nat Commun 12(1): 41. PubMed ID: 33397980

    Ilyin, A. A., Stolyarenko, A. D., Klenov, M. S. and Shevelyov, Y. Y. (2020). Various modes of HP1a interactions with the euchromatic chromosome arms in Drosophila ovarian somatic cells. Chromosoma 129(3-4): 201-214. PubMed ID: 32500264

    Sokolova, O. A., Ilyin, A. A., Poltavets, A. S., Nenasheva, V. V., Mikhaleva, E. A., Shevelyov, Y. Y. and Klenov, M. S. (2019). Yb body assembly on the flamenco piRNA precursor transcripts reduces genic piRNA production. Mol Biol Cell 30(12): 1544-1554. PubMed ID: 30943101

  • Ali Shilatifard Department of Biochemistry and Molecular Genetics, Northwestern University
    Rickels, R., Wang, L., Iwanaszko, M., Ozark, P. A., Morgan, M. A., Piunti, A., Khalatyan, N., Soliman, S. H. A., Rendleman, E. J., Savas, J. N., Smith, E. R. and Shilatifard, A. (2020). A small UTX stabilization domain of Trr is conserved within mammalian MLL3-4/COMPASS and is sufficient to rescue loss of viability in null animals. Genes Dev 34(21-22): 1493-1502. PubMed ID: 33033055

    Rickels, R., Herz, H. M., Sze, C. C., Cao, K., Morgan, M. A., Collings, C. K., Gause, M., Takahashi, Y. H., Wang, L., Rendleman, E. J., Marshall, S. A., Krueger, A., Bartom, E. T., Piunti, A., Smith, E. R., Abshiru, N. A., Kelleher, N. L., Dorsett, D. and Shilatifard, A. (2017). Histone H3K4 monomethylation catalyzed by Trr and mammalian COMPASS-like proteins at enhancers is dispensable for development and viability. Nat Genet 49(11): 1647-1653. PubMed ID: 28967912

    Morgan, M. A. J., Rickels, R. A., Collings, C. K., He, X., Cao, K., Herz, H. M., Cozzolino, K. A., Abshiru, N. A., Marshall, S. A., Rendleman, E. J., Sze, C. C., Piunti, A., Kelleher, N. L., Savas, J. N. and Shilatifard, A. (2017). A cryptic Tudor domain links BRWD2/PHIP to COMPASS-mediated histone H3K4 methylation. Genes Dev 31(19): 2003-2014. PubMed ID: 29089422

    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

    Rickels, R., Hu, D., Collings, C. K., Woodfin, A. R., Piunti, A., Mohan, M., Herz, H. M., Kvon, E. and Shilatifard, A. (2016). An evolutionary conserved epigenetic mark of Polycomb response elements implemented by Trx/MLL/COMPASS. Mol Cell 63: 318-328. PubMed ID: 27447986

    Piunti, A. and Shilatifard, A. (2016). Epigenetic balance of gene expression by Polycomb and COMPASS families. Science 352: aad9780. PubMed ID: 27257261

    Herz, H. M., Morgan, M., Gao, X., Jackson, J., Rickels, R., Swanson, S. K., Florens, L., Washburn, M. P., Eissenberg, J. C. and Shilatifard, A. (2014). Histone H3 lysine-to-methionine mutants as a paradigm to study chromatin signaling. Science 345: 1065-1070. PubMed ID: 25170156
    Smith, E. and Shilatifard, A. (2014). Enhancer biology and enhanceropathies. Nat Struct Mol Biol 21: 210-219. PubMed ID: 24599251

    Herz, H. M., Hu, D. and Shilatifard, A. (2014). Enhancer Malfunction in Cancer. Mol Cell 53: 859-866. PubMed ID: 24656127

    Hu, D., Smith, E. R., Garruss, A. S., Mohaghegh, N., Varberg, J. M., Lin, C., Jackson, J., Gao, X., Saraf, A., Florens, L., Washburn, M. P., Eissenberg, J. C. and Shilatifard, A. (2013). The Little Elongation Complex Functions at Initiation and Elongation Phases of snRNA Gene Transcription. Mol Cell. PubMed ID: 23932780

    Morgan, M. A., Shilatifard, A. (2013) Drosophila SETs its Sights on Cancer: Trr/MLL3/4 COMPASS-like complexes in Development and Disease. Mol Cell Biol. Pubmed ID: 23459940

  • Benny Shilo Weitzman Institute
    Carmon, S., Jonas, F., Barkai, N., Schejter, E. D. and Shilo, B. Z. (2021). Generation and timing of graded responses to morphogen gradients. Development 148(24). PubMed ID: 34918740

    Kamalesh, K., Scher, N., Biton, T., Schejter, E. D., Shilo, B. Z. and Avinoam, O. (2021). Exocytosis by vesicle crumpling maintains apical membrane homeostasis during exocrine secretion. Dev Cell 56(11): 1603-1616.e1606. PubMed ID: 34102104

    Dhanyasi, N., VijayRaghavan, K., Shilo, B. Z. and Schejter, E. D. (2020). Microtubules provide guidance cues for myofibril and sarcomere assembly and growth. Dev Dyn. PubMed ID: 32725855

    Rahimi, N., Carmon, S., Averbukh, I., Khajouei, F., Sinha, S., Schejter, E. D., Barkai, N. and Shilo, B. Z. (2020). Global shape of Toll activation is determined by wntD enhancer properties. Proc Natl Acad Sci U S A. PubMed ID: 31900360

    Rahimi, N., Averbukh, I., Carmon, S., Schejter, E. D., Barkai, N. and Shilo, B. Z. (2019). Dynamics of Spaetzle morphogen shuttling in the Drosophila embryo shapes gastrulation patterning. Development 146(21). PubMed ID: 31719046

    Segal, D., Zaritsky, A., Schejter, E. D. and Shilo, B. Z. (2018). Feedback inhibition of actin on Rho mediates content release from large secretory vesicles. J Cell Biol. PubMed ID: 29496739

    Shwartz, A., Dhanyasi, N., Schejter, E.D. and Shilo, B.Z. (2016). The Drosophila formin Fhos is a primary mediator of sarcomeric thin-filament array assembly. Elife 5. PubMed ID: 27731794

    Segal, D., Dhanyasi, N., Schejter, E. D. and Shilo, B. Z. (2016). Adhesion and Fusion of Muscle Cells Are Promoted by Filopodia. Dev Cell 38: 291-304. PubMed ID: 27505416

    Rahimi, N., Averbukh, I., Haskel-Ittah, M., Degani, N., Schejter, E. D., Barkai, N. and Shilo, B. Z. (2016). A WntD-dependent integral feedback loop attenuates variability in Drosophila toll signaling. Dev Cell 36: 401-414. PubMed ID: 26906736

    Gavish, A., Shwartz, A., Weizman, A., Schejter, E., Shilo, B. Z. and Barkai, N. (2016). Periodic patterning of the Drosophila eye is stabilized by the diffusible activator Scabrous. Nat Commun 7: 10461. PubMed ID: 26876750

    Rousso, T., Schejter, E.D. and Shilo, B.Z. (2015). Orchestrated content release from Drosophila glue-protein vesicles by a contractile actomyosin network. Nat Cell Biol [Epub ahead of print]. PubMed ID: 26641716

    Dhanyasi, N., Segal, D., Shimoni, E., Shinder, V., Shilo, B.Z., VijayRaghavan, K. and Schejter, E.D. (2015). Surface apposition and multiple cell contacts promote myoblast fusion in Drosophila flight muscles. J Cell Biol 211: 191-203. PubMed ID: 26459604

  • Osamu Shimmi Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
    Toddie-Moore, D. J., Montanari, M. P., Tran, N. V., Brik, E. M., Antson, H., Salazar-Ciudad, I. and Shimmi, O. (2021). Mechano-chemical feedback mediated competition for BMP signalling leads to pattern formation. Dev Biol 481: 43-51. PubMed ID: 34555363

    Montanari, M. P., Tran, N. V. and Shimmi, O. (2021). Regulation of spatial distribution of BMP ligands for pattern formation. Dev Dyn. PubMed ID: 34241935

    Gui, J., Huang, Y., Montanari, M., Toddie-Moore, D., Kikushima, K., Nix, S., Ishimoto, Y. and Shimmi, O. (2019). Coupling between dynamic 3D tissue architecture and BMP morphogen signaling during Drosophila wing morphogenesis. Proc Natl Acad Sci U S A. PubMed ID: 30760594

    Gui, J., Huang, Y. and Shimmi, O. (2016). Scribbled optimizes BMP signaling through its receptor internalization to the Rab5 endosome and promote robust epithelial morphogenesis. PLoS Genet 12: e1006424. PubMed ID: 27814354

    Tauscher, P. M., Gui, J. and Shimmi, O. (2016). Adaptive protein divergence of BMP ligands takes place under developmental and evolutionary constraints. Development. PubMed ID: 27578781

    Zeng, Z., de Gorter, D. J., Kowalski, M., Ten Dijke, P. and Shimmi, O. (2014). Ter94/VCP is a novel component involved in BMP signaling. PLoS One 9: e114475. PubMed ID: 25469707

    Kunnapuu, J., Tauscher, P., Tiusanen, N., Nguyen, M., Loytynoja, A., Arora, K. and Shimmi, O. (2014). Cleavage of the Drosophila Screw prodomain is critical for a dynamic BMP morphogen gradient in embryogenesis. Dev Biol. PubMed ID: 24560644

    Matsuda, S., Blanco, J., Shimmi, O. (2013) A Feed-Forward Loop Coupling Extracellular BMP Transport and Morphogenesis in Drosophila Wing. PLoS Genet 9: e1003403. PubMed ID: 23555308

    Matsuda, S. and Shimmi, O. (2012). Directional transport and active retention of Dpp/BMP create wing vein patterns in Drosophila. Dev Biol 366: 153-162. PubMed ID: 22542596

    Umulis, D. M., Shimmi, O., O'Connor, M. B. and Othmer, H. G. (2010). Organism-scale modeling of early Drosophila patterning via bone morphogenetic proteins. Dev Cell 18: 260-274. PubMed ID: 20159596

  • Alex Shingleton Michigan State University, East Lansing
    Kapali, G. P., Callier, V., Gascoigne, S. J. L., Harrison, J. F. and Shingleton, A. W. (2022). The steroid hormone ecdysone regulates growth rate in response to oxygen availability. Sci Rep 12(1): 4730. PubMed ID: 35304878

    McDonald, J. M. C., Nabili, P., Thorsen, L., Jeon, S. and Shingleton, A. W. (2021). Sex-specific plasticity and the nutritional geometry of insulin-signaling gene expression in Drosophila melanogaster. Evodevo 12(1): 6. PubMed ID: 33990225

    Dreyer, A. P. and Shingleton, A. W. (2019). Insulin-insensitivity of male genitalia maintains reproductive success in Drosophila. Biol Lett 15(5): 20190057. PubMed ID: 31088279

    Gokhale, R. H., Hayashi, T., Mirque, C. D. and Shingleton, A. W. (2016). Intra-organ growth coordination in Drosophila is mediated by systemic ecdysone signaling. Dev Biol [Epub ahead of print]. PubMed ID: 27452628

    Koyama, T., Rodrigues, M. A., Athanasiadis, A., Shingleton, A. W. and Mirth, C. K. (2014). Nutritional control of body size through FoxO-Ultraspiracle mediated ecdysone biosynthesis. Elife 3. PubMed ID: 25421296

    Oliveira, M. M., Shingleton, A. W. and Mirth, C. K. (2014). Coordination of Wing and Whole-Body Development at Developmental Milestones Ensures Robustness against Environmental and Physiological Perturbations. PLoS Genet 10: e1004408. PubMed ID: 24945255

    Mirth, C. K., Tang, H. Y., Makohon-Moore, S. C., Salhadar, S., Gokhale, R. H., Warner, R. D., Koyama, T., Riddiford, L. M. and Shingleton, A. W. (2014). Juvenile hormone regulates body size and perturbs insulin signaling in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 24778227

    Ghosh, S. M., Testa, N. D., Shingleton, A. W. (2013) Temperature-size rule is mediated by thermal plasticity of critical size in Drosophila melanogaster. Proc Biol Sci 280: 20130174. PubMed ID: 23595269

    Testa, N. D., Ghosh, S. M., Shingleton, A. W. (2013) Sex-Specific Weight Loss Mediates Sexual Size Dimorphism in Drosophila melanogaster. PLoS One 8: e58936. PubMed ID: 23555608

    Shingleton, A. W. and Tang, H. Y. (2012). Plastic flies: The regulation and evolution of trait variability in Drosophila. Fly (Austin) 6. PubMed ID: 22705976

    Mirth, C. K. and Shingleton, A. W. (2012). Integrating body and organ size in Drosophila: recent advances and outstanding problems. Front Endocrinol (Lausanne) 3: 49. PubMed ID: 22654869

  • Mimi Shirasu-Hiza Department of Genetics and Development, Columbia University
    Ulgherait, M., Midoun, A. M., Park, S. J., Gatto, J. A., Tener, S. J., Siewert, J., Klickstein, N., Canman, J. C., Ja, W. W. and Shirasu-Hiza, M. (2021). Circadian autophagy drives iTRF-mediated longevity. Nature 598(7880): 353-358. PubMed ID: 34588695

    Delventhal, R., O'Connor, R. M., Pantalia, M. M., Ulgherait, M., Kim, H. X., Basturk, M. K., Canman, J. C. and Shirasu-Hiza, M. (2019). Dissection of central clock function in Drosophila through cell-specific CRISPR-mediated clock gene disruption. Elife 8. PubMed ID: 31613218

    Hill, V. M., O'Connor, R. M., Sissoko, G. B., Irobunda, I. S., Leong, S., Canman, J. C., Stavropoulos, N. and Shirasu-Hiza, M. (2018). A bidirectional relationship between sleep and oxidative stress in Drosophila. PLoS Biol 16(7): e2005206. PubMed ID: 30001323

    Qiao, B., Li, C., Allen, V. W., Shirasu-Hiza, M. and Syed, S. (2018). Automated analysis of long-term grooming behavior in Drosophila using a k-nearest neighbors classifier. Elife 7. PubMed ID: 29485401

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

  • Akiko Shiratsuchi Graduate School Medical Science, Kanazawa University
    Nonaka, S., Ando, Y., Kanetani, T., Hoshi, C., Nakai, Y., Nainu, F., Nagaosa, K., Shiratsuchi, A. and Nakanishi, Y. (2017). Signaling Pathway for Phagocyte Priming upon Encounter with Apoptotic Cells. J Biol Chem. PubMed ID: 28325838

    Pukklay, P., Nakanishi, Y., Nitta, M., Yamamoto, K., Ishihama, A. and Shiratsuchi, A. (2013). Involvement of EnvZ-OmpR two-component system in virulence control of Escherichia coli in Drosophila melanogaster. Biochem Biophys Res Commun. PubMed ID: 23886953

    Nonaka, S., Nagaosa, K., Mori, T., Shiratsuchi, A. and Nakanishi, Y. (2013). Integrin alphaPS3/betanu-mediated phagocytosis of apoptotic cells and bacteria in Drosophila. J Biol Chem 288: 10374-10380. PubMed ID: 23426364

    Fujita, Y., Nagaosa, K., Shiratsuchi, A. and Nakanishi, Y. (2012). Role of NPxY motif in Draper-mediated apoptotic cell clearance in Drosophila. Drug Discov Ther 6: 291-297. PubMed ID: 23337816

    Shiratsuchi, A., Mori, T., Sakurai, K., Nagaosa, K., Sekimizu, K., Lee, B. L. and Nakanishi, Y. (2012). Independent recognition of Staphylococcus aureus by two receptors for phagocytosis in Drosophila. J Biol Chem 287: 21663-21672. PubMed ID: 22547074

  • Margret Shirinian American University of Beirut
    Frey, F., Sandakly, J., Ghannam, M., Doueiry, C., Hugosson, F., Berlandi, J., Ismail, J. N., Gayden, T., Hasselblatt, M., Jabado, N. and Shirinian, M. (2022). Drosophila Tet is required for maintaining glial homeostasis in developing and adult fly brains. eNeuro. PubMed ID: 35396259
    Chaouch, A., Berlandi, J., Chen, C. C. L., Frey, F., Badini, S., Harutyunyan, A. S., Chen, X., Krug, B., Hebert, S., Jeibmann, A., Lu, C., Kleinman, C. L., Hasselblatt, M., Lasko, P., Shirinian, M. and Jabado, N. (2021). Histone H3.3 K27M and K36M mutations de-repress transposable elements through perturbation of antagonistic chromatin marks. Mol Cell 81(23): 4876-4890 e4877. PubMed ID: 34739871

    Madi, J. R., Outa, A. A., Ghannam, M., Hussein, H. M., Shehab, M., Hasan, Z., Fayad, A. A., Shirinian, M. and Rahal, E. A. (2021). Drosophila melanogaster as a Model System to Assess the Effect of Epstein-Barr Virus DNA on Inflammatory Gut Diseases. Front Immunol 12: 586930. PubMed ID: 33828545

    Ismail, J. N., Ghannam, M., Al Outa, A., Frey, F. and Shirinian, M. (2020). Ten-eleven translocation proteins and their role beyond DNA demethylation - what we can learn from the fly. Epigenetics 15(11): 1139-1150. PubMed ID: 32419604

  • Galit Shohat-Ophir Faculty of Life Sciences, Bar-Ilan University, Institute of Nanotechnology and Advanced Materials, Ramat-Gan, Israel
    Bentzur, A., Ben-Shaanan, S., Benichou, J. I. C., Costi, E., Levi, M., Ilany, A. and Shohat-Ophir, G. (2020). Early Life Experience Shapes Male Behavior and Social Networks in Drosophila. Curr Biol. PubMed ID: 33186552

    Deng, P., Khan, A., Jacobson, D., Sambrani, N., McGurk, L., Li, X., Jayasree, A., Hejatko, J., Shohat-Ophir, G., O'Connell, M. A., Li, J. B. and Keegan, L. P. (2020). Adar RNA editing-dependent and -independent effects are required for brain and innate immune functions in Drosophila. Nat Commun 11(1): 1580. PubMed ID: 32221286

    Sapiro, A. L., Shmueli, A., Henry, G. L., Li, Q., Shalit, T., Yaron, O., Paas, Y., Billy Li, J. and Shohat-Ophir, G. (2019). Illuminating spatial A-to-I RNA editing signatures within the Drosophila brain. Proc Natl Acad Sci U S A 116(6): 2318-2327. PubMed ID: 30659150

  • Joshua Shulman Laboratory for Integrative Functional Genomics, Baylor University, Houston
    Zhao, P., Mangleburg, C. G., Al-Ramahi, I., Botas, J. and Shulman, J. M. (2021). Systems genetic dissection of Alzheimer's disease brain gene expression networks. Alzheimers Dement 17 Suppl 2: e058716. PubMed ID: 34971159

    Abreha, M. H., Ojelade, S., Dammer, E. B., McEachin, Z. T., Duong, D. M., Gearing, M., Bassell, G. J., Lah, J. J., Levey, A. I., Shulman, J. M. and Seyfried, N. T. (2021). TBK1 interacts with tau and enhances neurodegeneration in tauopathy. J Biol Chem: 100760. PubMed ID: 33965374

    Mangleburg, C. G., Wu, T., Yalamanchili, H. K., Guo, C., Hsieh, Y. C., Duong, D. M., Dammer, E. B., De Jager, P. L., Seyfried, N. T., Liu, Z. and Shulman, J. M. (2020). Integrated analysis of the aging brain transcriptome and proteome in tauopathy. Mol Neurodegener 15(1): 56. PubMed ID: 32993812

    Ye, H., Ojelade, S. A., Li-Kroeger, D., Zuo, Z., Wang, L., Li, Y., Gu, J. Y., Tepass, U., Rodal, A. A., Bellen, H. J. and Shulman, J. M. (2020). Retromer subunit, VPS29, regulates synaptic transmission and is required for endolysosomal function in the aging brain. Elife 9. PubMed ID: 32286230

    Hsieh, Y. C., Guo, C., Yalamanchili, H. K., Abreha, M., Al-Ouran, R., Li, Y., Dammer, E. B., Lah, J. J., Levey, A. I., Bennett, D. A., De Jager, P. L., Seyfried, N. T., Liu, Z. and Shulman, J. M. (2019). Tau-mediated disruption of the spliceosome triggers cryptic RNA splicing and neurodegeneration in Alzheimer's disease. Cell Rep 29(2): 301-316.e310. PubMed ID: 31597093

    Guo, C., Jeong, H. H., Hsieh, Y. C., Klein, H. U., Bennett, D. A., De Jager, P. L., Liu, Z. and Shulman, J. M. (2018). Tau activates transposable elements in Alzheimer's disease. Cell Rep 23(10): 2874-2880. PubMed ID: 29874575

    Chouhan, A. K., Guo, C., Hsieh, Y. C., Ye, H., Senturk, M., Zuo, Z., Li, Y., Chatterjee, S., Botas, J., Jackson, G. R., Bellen, H. J. and Shulman, J. M. (2016). Uncoupling neuronal death and dysfunction in Drosophila models of neurodegenerative disease. Acta Neuropathol Commun 4: 62. PubMed ID: 27338814

    Shulman, J. M. (2015). Drosophila and experimental neurology in the post-genomic era. Exp Neurol 274: 4-13. PubMed ID: 25814441

    Shulman, J. M., Imboywa, S., Giagtzoglou, N., Powers, M. P., Hu, Y., Devenport, D., Chipendo, P., Chibnik, L. B., Diamond, A., Perrimon, N., Brown, N. H., De Jager, P. L. and Feany, M. B. (2014). Functional screening in Drosophila identifies Alzheimer's disease susceptibility genes and implicates Tau-mediated mechanisms. Hum Mol Genet 23: 870-877. PubMed ID: 24067533

  • Stanislav Shvartsman Lewis-Sigler Institute for Integrative Genomics, Princeton University
    Ho, E. K., Oatman, H. R., McFann, S. E., Yang, L., Johnson, H. E., Shvartsman, S. Y. and Toettcher, J. E. (2023). Dynamics of an incoherent feedforward loop drive ERK-dependent pattern formation in the early Drosophila embryo. bioRxiv. PubMed ID: 36945584

    Stern, T., Shvartsman, S. Y. and Wieschaus, E. F. (2022). Deconstructing gastrulation at single-cell resolution. Curr Biol. PubMed ID: 35290798

    Alsous, J. I., Rozman, J., Marmion, R. A., Kosmrlj, A. and Shvartsman, S. Y. (2021). Clonal dominance in excitable cell networks. Nat Phys 17(12): 1391-1395. PubMed ID: 35242199

    Keenan, S. E., Avdeeva, M., Yang, L., Alber, D. S., Wieschaus, E. F. and Shvartsman, S. Y. (2022). Dynamics of Drosophila endoderm specification. Proc Natl Acad Sci U S A 119(15): e2112892119. PubMed ID: 35412853

    Shao, B., Diegmiller, R. and Shvartsman, S. Y. (2021). Collective oscillations of coupled cell cycles. Biophys J. PubMed ID: 34197797

    Patel, A. L., Zhang, L., Keenan, S. E., Rushlow, C. A., Fradin, C. and Shvartsman, S. Y. (2021). Capicua is a fast-acting transcriptional brake. Curr Biol. PubMed ID: 34166605

    Doherty, C. A., Diegmiller, R., Kapasiawala, M., Gavis, E. R. and Shvartsman, S. Y. (2021). Coupled oscillators coordinate collective germline growth. Dev Cell. PubMed ID: 33689691

    Diegmiller, R., Zhang, L., Gameiro, M., Barr, J., Imran Alsous, J., Schedl, P., Shvartsman, S. Y. and Mischaikow, K. (2021). Mapping parameter spaces of biological switches. PLoS Comput Biol 17(2): e1008711. PubMed ID: 33556054

    Johnson, H. E., Djabrayan, N. J. V., Shvartsman, S. Y. and Toettcher, J. E. (2020). Optogenetic Rescue of a Patterning Mutant. Curr Biol. PubMed ID: 32707057

    Dutta, S., Djabrayan, N. J., Smits, C. M., Rowley, C. W. and Shvartsman, S. Y. (2020). Excess dNTPs Trigger Oscillatory Surface Flow in the Early Drosophila Embryo. Biophys J. PubMed ID: 32247330

    Keenan, S. E., Blythe, S. A., Marmion, R. A., Djabrayan, N. J., Wieschaus, E. F. and Shvartsman, S. Y. (2020). Rapid dynamics of signal-dependent transcriptional repression by Capicua. Dev Cell. PubMed ID: 32142631

  • Kausik Si Stowers Institute for Medical Research, University of Kansas School of Medicine
    Singh, N. P., De Kumar, B., Paulson, A., Parrish, M. E., Zhang, Y., Florens, L., Conaway, J. W., Si, K. and Krumlauf, R. (2020). A six-amino-acid motif is a major determinant in functional evolution of HOX1 proteins. Genes Dev. PubMed ID: 33184220

    Hervas, R., Rau, M. J., Park, Y., Zhang, W., Murzin, A. G., Fitzpatrick, J. A. J., Scheres, S. H. W. and Si, K. (2020). Cryo-EM structure of a neuronal functional amyloid implicated in memory persistence in Drosophila. Science 367(6483): 1230-1234. PubMed ID: 32165583

    Nil, Z., Millan, R. H., Gerbich, T., Leal, P., Yu, Z., Saraf, A., Sardiu, M., Lange, J. J., Yi, K., Unruh, J., Slaughter, B. and Si, K. (2019). Amyloid-like assembly activates a phosphatase in the developing Drosophila embryo. Cell 178(6): 1403-1420.e1421. PubMed ID: 31491385

    Barajas-Azpeleta, R., Wu, J., Gill, J., Welte, R., Seidel, C., McKinney, S., Dissel, S. and Si, K. (2018). Antimicrobial peptides modulate long-term memory. PLoS Genet 14(10): e1007440. PubMed ID: 30312294

    Gill, J., Park, Y., McGinnis, J. P., Perez-Sanchez, C., Blanchette, M. and Si, K. (2017). Regulated intron removal integrates motivational state and experience. Cell 169(5): 836-848.e815. PubMed ID: 28525754

    McGinnis, J. P., Jiang, H., Agha, M. A., Perez Sanchez, C., Lange, J. J., Yu, Z., Marion-Poll, F. and Si, K. (2016). Immediate perception of a reward is distinct from the reward's long-term salience. Elife 5. PubMed ID: 28005005

    Li, L., Sanchez, C. P., Slaughter, B. D., Zhao, Y., Khan, M. R., Unruh, J. R., Rubinstein, B. and Si, K. (2016). A Putative Biochemical Engram of Long-Term Memory. Curr Biol 26(23): 3143-3156. PubMed ID: 27818176

    Hervas, R., Li, L., Majumdar, A., Fernandez-Ramirez Mdel, C., Unruh, J. R., Slaughter, B. D., Galera-Prat, A., Santana, E., Suzuki, M., Nagai, Y., Bruix, M., Casas-Tinto, S., Menendez, M., Laurents, D. V., Si, K. and Carrion-Vazquez, M. (2016). Molecular basis of Orb2 amyloidogenesis and blockade of memory Consolidation. PLoS Biol 14: e1002361. PubMed ID: 26812143

    Khan, M.R., Li, L., Pérez-Sánchez, C., Saraf, A., Florens, L., Slaughter, B.D., Unruh, J.R. and Si, K. (2015). Amyloidogenic oligomerization transforms Drosophila Orb2 from a translation repressor to an activator. Cell 163: 1468-1483. PubMed ID: 26638074

    White-Grindley, E., Li, L., Mohammad Khan, R., Ren, F., Saraf, A., Florens, L. and Si, K. (2014). Contribution of Orb2A Stability in Regulated Amyloid-Like Oligomerization of Drosophila Orb2. PLoS Biol 12: e1001786. PubMed ID: 24523662

  • Ody Sibon Cell Biology, University of Groningen, The Netherlands
    Faber, A. I. E., van der Zwaag, M., Schepers, H., Eggens-Meijer, E., Kanon, B., C, I. J., Kuipers, J., Giepmans, B. N. G., Freire, R., Grzeschik, N. A., Rabouille, C. and Sibon, O. C. M. (2020). Vps13 is required for timely removal of nurse cell corpses. Development. PubMed ID: 32994170
    <>Vonk, J. J., Yeshaw, W. M., Pinto, F., Faber, A. I., Lahaye, L. L., Kanon, B., van der Zwaag, M., Velayos-Baeza, A., Freire, R., van, I. S. C., Grzeschik, N. A. and Sibon, O. C. (2017). Drosophila Vps13 Is Required for Protein Homeostasis in the Brain. PLoS One 12(1): e0170106. PubMed ID: 28107480

    Iuso, A., Sibon, O. C., Gorza, M., Heim, K., Organisti, C., Meitinger, T. and Prokisch, H. (2014). Impairment of Drosophila orthologs of the human orphan protein C19orf12 induces bang sensitivity and neurodegeneration. PLoS One 9(2): e89439. PubMed ID: 24586779

    Siudeja, K., Srinivasan, B., Xu, L., Rana, A., de Jong, J., Nollen, E. A., Jackowski, S., Sanford, L., Hayflick, S. and Sibon, O. C. (2011). Impaired Coenzyme A metabolism affects histone and tubulin acetylation in Drosophila and human cell models of pantothenate kinase associated neurodegeneration. EMBO Mol Med 3(12): 755-766. PubMed ID: 21998097

  • Yasir Hasan Siddique Aligarh Muslim University
    Rahul, Naz, F., Jyoti, S. and Siddique, Y. H. (2020). Effect of kaempferol on the transgenic Drosophila model of Parkinson's disease. Sci Rep 10(1): 13793. PubMed ID: 32796885

    Siddique, Y. H., Ara, G., Afzal, M. (2012) Effect of the Steroid K-canrenoate on hsp70 Expression and Tissue Damage in Transgenic Drosophila melanogaster (hsp70-lacZ) Bg (9). J Insect Sci 12: 92. PubMed ID: 23427921

    Khan, S., Jyoti, S., Naz, F., Shakya, B., Rahul, Afzal, M. and Siddique, Y. H. (2012). Effect of L-ascorbic Acid on the climbing ability and protein levels in the brain of Drosophila model of Parkinson's disease. Int J Neurosci 122: 704-709. PubMed ID: 22776006

    Siddique, Y. H., Ara, G., Jyoti, S. and Afzal, M. (2012). The dietary supplementation of nordihydroguaiaretic acid (NDGA) delayed the loss of climbing ability in Drosophila model of Parkinson's disease. J Diet Suppl 9: 1-8. PubMed ID: 22432798

  • Sarah Siegrist Department of Biology, University of Virginia, Charlottesville
    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

    Yuan, X., Sipe, C. W., Suzawa, M., Bland, M. L. and Siegrist, S. E. (2020). Dilp-2-mediated PI3-kinase activation coordinates reactivation of quiescent neuroblasts with growth of their glial stem cell niche. PLoS Biol 18(5): e3000721. PubMed ID: 32463838

    Pahl, M. C., Doyle, S. E. and Siegrist, S. E. (2019). E93 integrates neuroblast intrinsic state with developmental time to terminate MB neurogenesis via autophagy. Curr Biol 29(5): 750-762. PubMed ID: 30773368

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

    Doyle, S. E., Pahl, M. C., Siller, K. H., Ardiff, L. and Siegrist, S. E. (2017). Neuroblast niche position is controlled by PI3-kinase dependent DE-Cadherin adhesion. Development [Epub ahead of print]. PubMed ID: 28126840

    Siegrist, S. E., Haque, N. S., Chen, C. H., Hay, B. A. and Hariharan, I. K. (2010). Inactivation of both Foxo and reaper promotes long-term adult neurogenesis in Drosophila. Curr Biol 20(7): 643-648. PubMed ID: 20346676

  • Daria Siekhaus Institute of Science and Technology, Klosterneuburg, Austria
    Emtenani, S., Martin, E. T., Gyoergy, A., Bicher, J., Genger, J. W., Köcher, T., Akhmanova, M., Guarda, M., Roblek, M., Bergthaler, A., Hurd, T. R., Rangan, P. and Siekhaus, D. E. (2022). Macrophage mitochondrial bioenergetics and tissue invasion are boosted by an Atossa-Porthos axis in Drosophila. Embo j: e109049. PubMed ID: 35319107

    Belyaeva, V., Wachner, S., Gyoergy, A., Emtenani, S., Gridchyn, I., Akhmanova, M., Linder, M., Roblek, M., Sibilia, M. and Siekhaus, D. (2022). Fos regulates macrophage infiltration against surrounding tissue resistance by a cortical actin-based mechanism in Drosophila. PLoS Biol 20(1): e3001494. PubMed ID: 34990456

    Ratheesh, A., Biebl, J., Vesela, J., Smutny, M., Papusheva, E., Krens, S. F. G., Kaufmann, W., Gyoergy, A., Casano, A. M. and Siekhaus, D. E. (2018). Drosophila TNF Modulates Tissue Tension in the Embryo to Facilitate Macrophage Invasive Migration. Dev Cell 45(3): 331-346 e337. PubMed ID: 29738712

    Gyoergy, A., Roblek, M., Ratheesh, A., Valoskova, K., Belyaeva, V., Wachner, S., Matsubayashi, Y., Sanchez-Sanchez, B. J., Stramer, B. and Siekhaus, D. E. (2018). Tools Allowing Independent Visualization and Genetic Manipulation of Drosophila melanogaster Macrophages and Surrounding Tissues. G3 (Bethesda) 8(3): 845-857. PubMed ID: 29321168

    Ratheesh, A., Belyaeva, V. and Siekhaus, D. E. (2015). Drosophila immune cell migration and adhesion during embryonic development and larval immune responses. Curr Opin Cell Biol 36: 71-79. PubMed ID: 26210104

  • Sarah Signor Department of Biological Sciences, North Dakota State University, Fargo
    Wierzbicki, F., Kofler, R. and Signor, S. (2021). Evolutionary dynamics of piRNA clusters in Drosophila. Mol Ecol. PubMed ID: 34878692

    Tiedeman, Z. and Signor, S. (2021). The Transposable Elements of the Drosophila serrata Reference Panel. Genome Biol Evol 13(9). PubMed ID: 33950180

    Signor, S. A. (2020). Evolution of Plasticity in Response to Ethanol between Sister Species with Different Ecological Histories (Drosophila melanogaster and D. simulans). Am Nat 196(5): 620-633. PubMed ID: 33064591

    Signor, S. (2020). Transposable elements in individual genotypes of Drosophila simulans. Ecol Evol 10(7): 3402-3412. PubMed ID: 32273997

  • Stephan Sigrist Freie Universität Berlin, Institute of Biology, Genetics
    Hofer, S. J., Liang, Y., Zimmermann, A., Schroeder, S., Dengjel, J., Kroemer, G., Eisenberg, T., Sigrist, S. J. and Madeo, F. (2021). Spermidine-induced hypusination preserves mitochondrial and cognitive function during aging. Autophagy: 1-3. PubMed ID: 34105442

    Pooryasin, A., Maglione, M., Schubert, M., Matkovic-Rachid, T., Hasheminasab, S. M., Pech, U., Fiala, A., Mielke, T. and Sigrist, S. J. (2021). Unc13A and Unc13B contribute to the decoding of distinct sensory information in Drosophila. Nat Commun 12(1): 1932. PubMed ID: 33771998

    Liang, Y., Piao, C., Beuschel, C. B., Toppe, D., Kollipara, L., Bogdanow, B., Maglione, M., Lützkendorf, J., See, J. C. K., Huang, S., Conrad, T. O. F., Kintscher, U., Madeo, F., Liu, F., Sickmann, A. and Sigrist, S. J. (2021). eIF5A hypusination, boosted by dietary spermidine, protects from premature brain aging and mitochondrial dysfunction. Cell Rep 35(2): 108941. PubMed ID: 33852845

    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

    Zhao, K., Hong, H., Zhao, L., Huang, S., Gao, Y., Metwally, E., Jiang, Y., Sigrist, S. J. and Zhang, Y. Q. (2020). Postsynaptic cAMP signalling regulates the antagonistic balance of Drosophila glutamate receptor subtypes. Development 147(24). PubMed ID: 33234716

    Petzoldt, A. G., Gotz, T. W. B., Driller, J. H., Lutzkendorf, J., Reddy-Alla, S., Matkovic-Rachid, T., Liu, S., Knoche, E., Mertel, S., Ugorets, V., Lehmann, M., Ramesh, N., Beuschel, C. B., Kuropka, B., Freund, C., Stelzl, U., Loll, B., Liu, F., Wahl, M. C. and Sigrist, S. J. (2020). RIM-binding protein couples synaptic vesicle recruitment to release sites. J Cell Biol 219(7). PubMed ID: 32369542

    Huang, S., Piao, C., Beuschel, C. B., Gotz, T. and Sigrist, S. J. (2020). Presynaptic Active Zone Plasticity Encodes Sleep Need in Drosophila. Curr Biol. PubMed ID: 32142702

    Woitkuhn, J., Ender, A., Beuschel, C. B., Maglione, M., Matkovic-Rachid, T., Huang, S., Lehmann, M., Geiger, J. R. P. and Sigrist, S. J. (2020). The Unc13A isoform is important for phasic release and olfactory memory formation at mushroom body synapses. J Neurogenet: 1-9. PubMed ID: 31980003

    Raccuglia, D., Huang, S., Ender, A., Heim, M. M., Laber, D., Suarez-Grimalt, R., Liotta, A., Sigrist, S. J., Geiger, J. R. P. and Owald, D. (2019). Network-specific synchronization of electrical slow-wave oscillations regulates sleep drive in Drosophila. Curr Biol 29(21): 3611-3621. PubMed ID: 31630955

    Tenedini, F. M., Saez Gonzalez, M., Hu, C., Pedersen, L. H., Petruzzi, M. M., Spitzweck, B., Wang, D., Richter, M., Petersen, M., Szpotowicz, E., Schweizer, M., Sigrist, S. J., Calderon de Anda, F. and Soba, P. (2019). Maintenance of cell type-specific connectivity and circuit function requires Tao kinase. Nat Commun 10(1): 3506. PubMed ID: 31383864

  • Marion Silies European Neuroscience Institute, Göttingen, Germany
    Ketkar, M. D., Gur, B., Molina-Obando, S., Ioannidou, M., Martelli, C. and Silies, M. (2022). First-order visual interneurons distribute distinct contrast and luminance information across ON and OFF pathways to achieve stable behavior. Elife 11. PubMed ID: 35263247

    Henning, M., Ramos-Traslosheros, G., Gur, B. and Silies, M. (2022). Populations of local direction-selective cells encode global motion patterns generated by self-motion. Sci Adv 8(3): eabi7112. PubMed ID: 35044821

    Ramos-Traslosheros, G. and Silies, M. (2021). The physiological basis for contrast opponency in motion computation in Drosophila. Nat Commun 12(1): 4987. PubMed ID: 34404776

    Ketkar, M. D., Sporar, K., Gur, B., Ramos-Traslosheros, G., Seifert, M. and Silies, M. (2020). Luminance Information Is Required for the Accurate Estimation of Contrast in Rapidly Changing Visual Contexts. Curr Biol. PubMed ID: 32008904

    Gur, B., Sporar, K., Lopez-Behling, A. and Silies, M. (2019). Distinct expression of potassium channels regulates visual response properties of lamina neurons in Drosophila melanogaster. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. PubMed ID: 31823004

    Molina-Obando, S., Vargas-Fique, J. F., Henning, M., Gur, B., Schladt, T. M., Akhtar, J., Berger, T. K. and Silies, M. (2019). ON selectivity in Drosophila vision is a multisynaptic process involving both glutamatergic and GABAergic inhibition. Elife 8. PubMed ID: 31535971

    Liou, N. F., Lin, S. H., Chen, Y. J., Tsai, K. T., Yang, C. J., Lin, T. Y., Wu, T. H., Lin, H. J., Chen, Y. T., Gohl, D. M., Silies, M. and Chou, Y. H. (2018). Diverse populations of local interneurons integrate into the Drosophila adult olfactory circuit. Nat Commun 9(1): 2232. PubMed ID: 29884811

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

    Peng, J., Santiago, I. J., Ahn, C., Gur, B., Tsui, C. K., Su, Z., Xu, C., Karakhanyan, A., Silies, M. and Pecot, M. Y. (2018). Drosophila Fezf coordinates laminar-specific connectivity through cell-intrinsic and cell-extrinsic mechanisms. Elife 7. PubMed ID: 29513217

  • Neal Silverman Infectious Diseases And Immunology, University of Massachusetts Medical School, Worcester
    West, C., Rus, F., Chen, Y., Kleino, A., Gangloff, M., Gammon, D. B. and Silverman, N. (2019). IIV-6 inhibits NF-kappaB responses in Drosophila. Viruses 11(5). PubMed ID: 31052481

    Nandy, A., Lin, L., Velentzas, P. D., Wu, L. P., Baehrecke, E. H. and Silverman, N. (2018). The NF-kappaB factor Relish regulates Atg1 expression and controls autophagy. Cell Rep 25(8): 2110-2120.e2113. PubMed ID: 30463009

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

    West, C. and Silverman, N. (2018). p38b and JAK-STAT signaling protect against invertebrate iridescent virus 6 infection in Drosophila. PLoS Pathog 14(5): e1007020. Pubmed ID: 29746571

    Kleino, A., Ramia, N. F., Bozkurt, G., Shen, Y., Nailwal, H., Huang, J., Napetschnig, J., Gangloff, M., Chan, F. K., Wu, H., Li, J. and Silverman, N. (2017). Peptidoglycan-sensing receptors trigger the formation of functional amyloids of the adaptor protein Imd to initiate Drosophila NF-kappaB signaling. Immunity 47(4): 635-647.e636. PubMed ID: 29045898

    Paik, D., Monahan, A., Caffrey, D. R., Elling, R., Goldman, W. E. and Silverman, N. (2017). SLC46 family transporters facilitate cytosolic innate immune recognition of monomeric peptidoglycans. J Immunol 199(1):263-270. PubMed ID: 28539433

    Chen, L., Paquette, N., Mamoor, S., Rus, F., Nandy, A., Leszyk, J., Shaffer, S. A. and Silverman, N. (2017). Innate immune signaling in Drosophila is regulated by TGFbeta-activated kinase (Tak1)-triggered ubiquitin editing. J Biol Chem [Epub ahead of print]. PubMed ID: 28377500

    Okuda, K., Tong, M., Dempsey, B., Moore, K. J., Gazzinelli, R. T. and Silverman, N. (2016). Leishmania amazonensis engages CD36 to drive parasitophorous vacuole maturation. PLoS Pathog 12: e1005669. PubMed ID: 27280707

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

    Kim, C. H., Paik, D., Rus, F. and Silverman, N. (2014). The caspase-8 homolog Dredd cleaves Imd and Relish but is not inhibited by p35. J Biol Chem. PubMed ID: 24891502

    Kleino, A. and Silverman, N. (2013). The Drosophila IMD pathway in the activation of the humoral immune response. Dev Comp Immunol. PubMed ID: 23721820

    Rus, F., Flatt, T., Tong, M., Aggarwal, K., Okuda, K., Kleino, A., Yates, E., Tatar, M. and Silverman, N. (2013). Ecdysone triggered PGRP-LC expression controls Drosophila innate immunity. EMBO J. PubMed ID: 23652443

    Warmbold, C., Uliczka, K., Rus, F., Suck, R., Petersen, A., Silverman, N., Ulmer, A. J., Heine, H. and Roeder, T. (2013). Dermatophagoides pteronyssinus major allergen 1 activates the innate immune response of the fruit fly Drosophila melanogaster. J Immunol 190: 366-371. PubMed ID: 23203927

  • Amanda Simcox Department of Molecular Genetics, Ohio State University, Columbus
    Palsule, G., Gopalan, V. and Simcox, A. (2019). Biogenesis of RNase P RNA from an intron requires co-assembly with cognate protein subunits. Nucleic Acids Res. PubMed ID: 31287870

    Fagegaltier, D., Falciatori, I., Czech, B., Castel, S., Perrimon, N., Simcox, A. and Hannon, G. J. (2016). Oncogenic transformation of Drosophila somatic cells induces a functional piRNA pathway. Genes Dev 30: 1623-1635. PubMed ID: 27474441

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

    Lim, M. Y., Ng, A. W., Chou, Y., Lim, T. P., Simcox, A., Tucker-Kellogg, G. and Okamura, K. (2016). The Drosophila Dicer-1 partner Loquacious enhances miRNA processing from hairpins with unstable structures at the dicing site. Cell Rep 15: 1795-1808. PubMed ID: 27184838

    Manivannan, S. N., Jacobsen, T. L., Lyon, P., Selvaraj, B., Halpin, P. and Simcox, A. (2015). Targeted integration of single-copy transgenes in Drosophila melanogaster tissue-culture cells using recombination mediated cassette exchange. Genetics [Epub ahead of print]. PubMed ID: 26500255

    Dequéant, M.L., Fagegaltier, D., Hu, Y., Spirohn, K., Simcox, A., Hannon, G.J. and Perrimon, N. (2015). Discovery of progenitor cell signatures by time-series synexpression analysis during Drosophila embryonic cell immortalization. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26438832

    Manivannan, S. N., Lai, L. B., Gopalan, V. and Simcox, A. (2015). Transcriptional control of an essential ribozyme in Drosophila reveals an ancient evolutionary divide in animals. PLoS Genet 11: e1004893. PubMed ID: 25569672

    Paul, L., Wang, S. H., Manivannan, S. N., Bonanno, L., Lewis, S., Austin, C. L., Simcox, A. (2013) Dpp-induced Egfr signaling triggers postembryonic wing development in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 23479629

    Butchar, J. P., Cain, D., Manivannan, S. N., McCue, A. D., Bonanno, L., Halula, S., Truesdell, S., Austin, C. L., Jacobsen, T. L. and Simcox, A. (2012). New negative feedback regulators of Egfr signaling in Drosophila. Genetics 191: 1213-1226. PubMed ID: 22595244

    Justiniano, S. E., Mathew, A., Mitra, S., Manivannan, S. N. and Simcox, A. (2012). Loss of the tumor suppressor Pten promotes proliferation of Drosophila melanogaster cells in vitro and gives rise to continuous cell lines. PLoS One 7: e31417. PubMed ID: 22363644

  • Anne Simon Department of Biology, Western University, London, Ontario, Canada
    Yost, R. T., Liang, E., Stewart, M. P., Chui, S., Greco, A. F., Long, S. Q., McDonald, I. S., McDowell, T., McNeil, J. N. and Simon, A. F. (2021). Drosophila melanogaster Stress Odorant (dSO) Displays the Characteristics of an Interspecific Alarm Cue. J Chem Ecol. PubMed ID: 34402994

    Yost, R. T., Robinson, J. W., Baxter, C. M., Scott, A. M., Brown, L. P., Aletta, M. S., Hakimjavadi, R., Lone, A., Cumming, R. C., Dukas, R., Mozer, B. and Simon, A. F. (2020). Abnormal Social Interactions in a Drosophila Mutant of an Autism Candidate Gene: Neuroligin 3. Int J Mol Sci 21(13). PubMed ID: 32610435

    Castells-Nobau, A., Eidhof, I., Fenckova, M., Brenman-Suttner, D. B., Scheffer-de Gooyert, J. M., Christine, S., Schellevis, R. L., van der Laan, K., Quentin, C., van Ninhuijs, L., Hofmann, F., Ejsmont, R., Fisher, S. E., Kramer, J. M., Sigrist, S. J., Simon, A. F. and Schenck, A. (2019). Conserved regulation of neurodevelopmental processes and behavior by FoxP in Drosophila. PLoS One 14(2): e0211652. PubMed ID: 30753188

    Brenman-Suttner, D. B., Long, S. Q., Kamesan, V., de Belle, J. N., Yost, R. T., Kanippayoor, R. L. and Simon, A. F. (2018). Progeny of old parents have increased social space in Drosophila melanogaster. Sci Rep 8(1): 3673. PubMed ID: 29487349

    Fernandez, R. W., Akinleye, A. A., Nurilov, M., Feliciano, O., Lollar, M., Aijuri, R. R., O'Donnell, J. M. and Simon, A. F. (2017). Modulation of social space by dopamine in Drosophila melanogaster, but no effect on the avoidance of the Drosophila stress odorant. Biol Lett 13(8). PubMed ID: 28794277

    Croft, J. R., Liu, T., Camiletti, A. L., Simon, A. F. and Thompson, G. J. (2017). Sexual response of male Drosophila to honey bee queen mandibular pheromone: implications for genetic studies of social insects. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 203(2): 143-149. PubMed ID: 28150219

    McNeil, A. R., Jolley, S. N., Akinleye, A. A., Nurilov, M., Rouzyi, Z., Milunovich, A. J., Chambers, M. C. and Simon, A. F. (2015). Conditions Affecting Social Space in Drosophila melanogaster. J Vis Exp(105): e53242. PubMed ID: 26575105

    Wise, A., Tenezaca, L., Fernandez, R. W., Schatoff, E., Flores, J., Ueda, A., Zhong, X., Wu, C. F., Simon, A. F. and Venkatesh, T. (2015). Drosophila mutants of the autism candidate gene neurobeachin (rugose) exhibit neuro-developmental disorders, aberrant synaptic properties, altered locomotion, and impaired adult social behavior and activity patterns. J Neurogenet 29(2-3): 135-143. PubMed ID: 26100104

  • Andrew Simmonds Department of Cell Biology, University of Alberta
    Ueda, K., Anderson-Baron, M. N., Haskins, J., Hughes, S. C. and Simmonds, A. J. (2022). Recruitment of Peroxin 14 to lipid droplets affects lipid storage in Drosophila. J Cell Sci 135(7). PubMed ID: 35274690

    Pridie, C. and Simmonds, A. (2020). The role of Peroxin 7 during Drosophila embryonic development. Genome. PubMed ID: 32191843

    Di Cara, F., Rachubinski, R. A. and Simmonds, A. J. (2018). Distinct roles for peroxisomal targeting signal receptors Pex5 and Pex7 in Drosophila. Genetics. PubMed ID: 30389805

    Pimmett, V.L., Deng, H., Haskins, J.A., Mercier, R.J., LaPointe, P. and Simmonds, A.J. (2017). The activity of the Drosophila Vestigial protein is modified by Scalloped-dependent phosphorylation. Dev Biol [Epub ahead of print]. PubMed ID: 28322734

    Baron, M. N., Klinger, C. M., Rachubinski, R. A. and Simmonds, A. J. (2016). A Systematic Cell-Based Analysis of Localization of Predicted Drosophila Peroxisomal Proteins. Traffic. PubMed ID: 26865094

    Germain, D. R., Li, L., Hildebrandt, M. R., Simmonds, A. J., Hughes, S. C. and Godbout, R. (2015). Loss of the Drosophila melanogaster DEAD box protein Ddx1 leads to reduced size and aberrant gametogenesis. Dev Biol 407: 232-245. PubMed ID: 26433063

    Li, J., Hobman, T. C. and Simmonds, A. J. (2013). Gawky (GW) is the Drosophila melanogaster GW182 homologue. Adv Exp Med Biol 768: 127-145. PubMed ID: 23224968

    Mast, F. D., Li, J., Virk, M. K., Hughes, S. C., Simmonds, A. J. and Rachubinski, R. A. (2011). A Drosophila model for the Zellweger spectrum of peroxisome biogenesis disorders. Dis Model Mech 4: 659-672. PubMed ID: 21669930

  • Michael Simon Department of Biological Sciences, Stanford
    Zhao, X., Yang, C. H., Simon, M. A. (2013) The Drosophila Cadherin Fat Regulates Tissue Size and Planar Cell Polarity through Different Domains. PLoS One 8: e62998. PubMed ID: 23667559

    Bischoff, K., Ballew, A. C., Simon, M. A. and O'Reilly, A. M. (2009). Wing defects in Drosophila xenicid mutant clones are caused by C-terminal deletion of additional sex combs (Asx). PLoS One 4: e8106. PubMed ID: 19956620

    O'Reilly, A. M., Lee, H. H. and Simon, M. A. (2008). Integrins control the positioning and proliferation of follicle stem cells in the Drosophila ovary. J Cell Biol 182: 801-815. PubMed ID: 18725542

  • Martine Simonelig Labex EPIGENMED Montpellier, France
    Ribot, C., Soler, C., Chartier, A., Al Hayek, S., Naït-Saïdi, R., Barbezier, N., Coux, O. and Simonelig, M. (2022). Activation of the ubiquitin-proteasome system contributes to oculopharyngeal muscular dystrophy through muscle atrophy. PLoS Genet 18(1): e1010015. PubMed ID: 35025870

    Ramat, A., Garcia-Silva, M. R., Jahan, C., Nait-Saidi, R., Dufourt, J., Garret, C., Chartier, A., Cremaschi, J., Patel, V., Decourcelle, M., Bastide, A., Juge, F. and Simonelig, M. (2020). The PIWI protein Aubergine recruits eIF3 to activate translation in the germ plasm. Cell Res. PubMed ID: 32132673

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

    Rojas-Rios, P., Chartier, A., Pierson, S. and Simonelig, M. (2017). Aubergine and piRNAs promote germline stem cell self-renewal by repressing the proto-oncogene Cbl. Embo j 36(21): 3194-3211. PubMed ID: 29030484

    Dufourt, J., Bontonou, G., Chartier, A., Jahan, C., Meunier, A. C., Pierson, S., Harrison, P. F., Papin, C., Beilharz, T. H. and Simonelig, M. (2017). piRNAs and Aubergine cooperate with Wispy poly(A) polymerase to stabilize mRNAs in the germ plasm. Nat Commun 8(1): 1305. PubMed ID: 29101389

    Gotze, M., Dufourt, J., Ihling, C., Rammelt, C., Pierson, S., Sambrani, N., Temme, C., Sinz, A., Simonelig, M. and Wahle, E. (2017). Translational repression of the Drosophila nanos mRNA involves the RNA helicase Belle and RNA coating by Me31B and Trailer hitch. Rna. PubMed ID: 28701521

    Barckmann, B., Pierson, S., Dufourt, J., Papin, C., Armenise, C., Port, F., Grentzinger, T., Chambeyron, S., Baronian, G., Desvignes, J. P., Curk, T. and Simonelig, M. (2015). Aubergine iCLIP reveals piRNA-dependent decay of mRNAs involved in germ cell development in the early embryo. Cell Rep 12: 1205-1216. PubMed ID: 26257181

    Joly, W., Chartier, A., Rojas-Rios, P., Busseau, I. and Simonelig, M. (2013). The CCR4 Deadenylase Acts with Nanos and Pumilio in the Fine-Tuning of Mei-P26 Expression to Promote Germline Stem Cell Self-Renewal. Stem Cell Reports 1: 411-424. PubMed ID: 24286029

    Barckmann, B. and Simonelig, M. (2013). Control of maternal mRNA stability in germ cells and early embryos. Biochim Biophys Acta 1829: 714-724. PubMed ID: 23298642

    Simonelig, M. (2011). Developmental functions of piRNAs and transposable elements: a Drosophila point-of-view. RNA Biol 8: 754-759. PubMed ID: 21712652

  • Matias Simons University of Freiburg
    Mukherjee, C., Kling, T., Russo, B., Miebach, K., Kess, E., Schifferer, M., Pedro, L. D., Weikert, U., Fard, M. K., Kannaiyan, N., Rossner, M., Aicher, M. L., Goebbels, S., Nave, K. A., Kramer-Albers, E. M., Schneider, A. and Simons, M. (2020). Oligodendrocytes Provide Antioxidant Defense Function for Neurons by Secreting Ferritin Heavy Chain. Cell Metab. PubMed ID: 32531201

    Marchesin, V., Perez-Marti, A., Le Meur, G., Pichler, R., Grand, K., Klootwijk, E. D., Kesselheim, A., Kleta, R., Lienkamp, S. and Simons, M. (2019). Molecular basis for autosomal-dominant renal Fanconi Syndrome caused by HNF4A. Cell Rep 29(13): 4407-4421.e4405. PubMed ID: 31875549

    Guida, M. C., Hermle, T., Graham, L. A., Hauser, V., Ryan, M., Stevens, T. H. and Simons, M. (2018). ATP6AP2 functions as a V-ATPase assembly factor in the endoplasmic reticulum. Mol Biol Cell: mbcE18040234. PubMed ID: 29995586

    Gleixner, E. M., Canaud, G., Hermle, T., Guida, M. C., Kretz, O., Helmstadter, M., Huber, T. B., Eimer, S., Terzi, F. and Simons, M. (2014). V-ATPase/mTOR Signaling Regulates Megalin-Mediated Apical Endocytosis. Cell Rep. PubMed ID: 24953654

    Ghosh, A., Kling, T., Snaidero, N., Sampaio, J. L., Shevchenko, A., Gras, H., Geurten, B., Gopfert, M. C., Schulz, J. B., Voigt, A. and Simons, M. (2013). A Global In Vivo Drosophila RNAi Screen Identifies a Key Role of Ceramide Phosphoethanolamine for Glial Ensheathment of Axons. PLoS Genet 9: e1003980. PubMed ID: 24348263

    Hermle, T., Guida, M. C., Beck, S., Helmstadter, S. and Simons, M. (2013). Drosophila ATP6AP2/VhaPRR functions both as a novel planar cell polarity core protein and a regulator of endosomal trafficking. EMBO J. PubMed ID: 23292348

    Vo, S. H., Butzlaff, M., Pru, S. K., Ni Charthaigh, R. A., Karsten, P., Lankes, A., Hamm, S., Simons, M., Adryan, B., Schulz, J. B. and Voigt, A. (2012). Large-scale screen for modifiers of ataxin-3-derived polyglutamine-induced toxicity in Drosophila. PLoS One 7: e47452. PubMed ID: 23139745

    Helmstadter, M., Luthy, K., Godel, M., Simons, M., Ashish, Nihalani, D., Rensing, S. A., Fischbach, K. F. and Huber, T. B. (2012). Functional study of mammalian Neph proteins in Drosophila melanogaster. PLoS One 7: e40300. PubMed ID: 22792268

  • Anne Simonsen Institute of Basic Medical Sciences, University of Oslo
    Knaevelsrud, H., Soreng, K., Raiborg, C., Haberg, K., Rasmuson, F., Brech, A., Liestol, K., Rusten, T. E., Stenmark, H., Neufeld, T. P., Carlsson, S. R. and Simonsen, A. (2013). Membrane remodeling by the PX-BAR protein SNX18 promotes autophagosome formation. J Cell Biol 202: 331-349. PubMed ID: 23878278

    Cumming, R. C., Simonsen, A. and Finley, K. D. (2008). Quantitative analysis of autophagic activity in Drosophila neural tissues by measuring the turnover rates of pathway substrates. Methods Enzymol 451: 639-651. PubMed ID: 19185743

    Nezis, I. P., Simonsen, A., Sagona, A. P., Finley, K., Gaumer, S., Contamine, D., Rusten, T. E., Stenmark, H. and Brech, A. (2008). Ref(2)P, the Drosophila melanogaster homologue of mammalian p62, is required for the formation of protein aggregates in adult brain. J Cell Biol 180: 1065-1071. PubMed ID: 18347073

  • Matias Simons UNI Freiburg
    Petzoldt, A. G., Gleixner, E. M., Fumagalli, A., Vaccari, T., Simons, M. (2013) Activation of the proton pump, V-ATPase, triggers JNK-dependent cell invasion and overgrowth in a Drosophila epithelium. Dis Model Mech. PubMed ID: 23335205

    Hermle, T., Guida, M. C., Beck, S., Helmstadter, S. and Simons, M. (2013). Drosophila ATP6AP2/VhaPRR functions both as a novel planar cell polarity core protein and a regulator of endosomal trafficking. EMBO J 32: 245-259. PubMed ID: 23292348

    Hermle, T., Saltukoglu, D., Grunewald, J., Walz, G. and Simons, M. (2010). Regulation of Frizzled-dependent planar polarity signaling by a V-ATPase subunit. Curr Biol 20: 1269-1276. PubMed ID: 20579879

  • Julie Simpson
    Molecular, Cellular, and Developmental Biology - University of California, Santa Barbara
    Ravbar, P., Zhang, N. and Simpson, J. H. (2021). Behavioral evidence for nested central pattern generator control of Drosophila grooming. Elife 10. PubMed ID: 34936550

    Mueller, J. M., Zhang, N., Carlson, J. M. and Simpson, J. H. (2021). Variation and Variability in Drosophila Grooming Behavior. Front Behav Neurosci 15: 769372. PubMed ID: 35087385

    Guo, L., Zhang, N. and Simpson, J. H. (2022). Descending neurons coordinate anterior grooming behavior in Drosophila. Curr Biol 32(4): 823-833. PubMed ID: 35120659

    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

    Zhang, N., Guo, L. and Simpson, J. H. (2020). Spatial comparisons of mechanosensory information govern the grooming sequence in Drosophila. Curr Biol. PubMed ID: 32142695

    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

    Hampel, S., McKellar, C. E., Simpson, J. H. and Seeds, A. M. (2017). Simultaneous activation of parallel sensory pathways promotes a grooming sequence in Drosophila. Elife 6. PubMed ID: 28887878

    Robie, A. A., Hirokawa, J., Edwards, A. W., Umayam, L. A., Lee, A., Phillips, M. L., Card, G. M., Korff, W., Rubin, G. M., Simpson, J. H., Reiser, M. B. and Branson, K. (2017). Mapping the Neural Substrates of Behavior. Cell 170(2): 393-406.e328. PubMed ID: 28709004

    Hampel, S., Franconville, R., Simpson, J.H. and Seeds, A.M. (2015). A neural command circuit for grooming movement control. Elife [Epub ahead of print]. PubMed ID: 26344548

    Albin, S.D., Kaun, K.R., Knapp, J.M., Chung, P., Heberlein, U. and Simpson, J.H. (2015). A subset of serotonergic neurons evokes hunger in adult Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 26344091

    Ohyama, T., Schneider-Mizell, C. M., Fetter, R. D., Aleman, J. V., Franconville, R., Rivera-Alba, M., Mensh, B. D., Branson, K. M., Simpson, J. H., Truman, J. W., Cardona, A. and Zlatic, M. (2015). A multilevel multimodal circuit enhances action selection in Drosophila. Nature 520: 633-639. PubMed ID: 25896325

    Knapp, J. M., Chung, P. and Simpson, J. H. (2015). Generating Customized Transgene Landing Sites and Multi-Transgene Arrays in Drosophila Using phiC31 Integrase. Genetics. PubMed ID: 25680812

  • Patricia Simpson Department of Zoology, University of Cambridge
    Negre, B. and Simpson, P. (2015). The achaete-scute complex in Diptera: patterns of non-coding sequence evolution. J Evol Biol [Epub ahead of print]. PubMed ID: 26134680

    Costa, M., Calleja, M., Alonso, C. R. and Simpson, P. (2013). The bristle patterning genes hairy and extramacrochaetae regulate the development of structures required for flight in Diptera. Dev Biol. PubMed ID: 24384389

    Usui, K., Goldstone, C., Gibert, J. M. and Simpson, P. (2008). Redundant mechanisms mediate bristle patterning on the Drosophila thorax. Proc Natl Acad Sci U S A 105: 20112-20117. PubMed ID: 19104061

    Simpson, P. and Ayyar, S. (2008). Evolution of cis-regulatory sequences in Drosophila. Adv Genet 61: 67-106. PubMed ID: 18282503

  • Brent Sinclair Department of Biology The University of Western Ontario, London, Ontario
    Salehipour-Shirazi, G., Ferguson, L. V. and Sinclair, B. J. (2016). Does cold activate the Drosophila melanogaster immune system? J Insect Physiol [Epub ahead of print]. PubMed ID: 27765624

    MacMillan, H. A., Knee, J. M., Dennis, A. B., Udaka, H., Marshall, K. E., Merritt, T. J. and Sinclair, B. J. (2016). Cold acclimation wholly reorganizes the Drosophila melanogaster transcriptome and metabolome. Sci Rep 6: 28999. PubMed ID: 27357258

    Toxopeus, J., Jakobs, R., Ferguson, L. V., Gariepy, T. D. and Sinclair, B. J. (2016). Reproductive arrest and stress resistance in winter-acclimated Drosophila suzukii. J Insect Physiol 89: 37-51. PubMed ID: 27039032

    Jakobs, R., Gariepy, T. D. and Sinclair, B. J. (2015). Adult plasticity of cold tolerance in a continental-temperate population of Drosophila suzukii. J Insect Physiol 79: 1-9. PubMed ID: 25982520

  • Amit Singh Department of Biology, Tissue Regeneration and Engineering at Dayton, University of Dayton
    Deshpande, P., Chen, C. Y., Yeates, C., Chen, C. H., Kango-Singh, M. and Singh, A. (2021). miR-277 targets hid to ameliorate Aβ42-mediated neurodegeneration in Drosophila eye model of Alzheimer's Disease. Alzheimers Dement 17 Suppl 2: e058678. PubMed ID: 34971161

    Raj, A., Chimata, A. V. and Singh, A. (2020). Motif 1 Binding Protein suppresses wingless to promote eye fate in Drosophila. Sci Rep 10(1): 17221. PubMed ID: 33057115

    Tare, M., Chimata, A. V., Gogia, N., Narwal, S., Deshpande, P. and Singh, A. (2020). An E3 ubiquitin ligase, cullin-4 regulates retinal differentiation in Drosophila eye. Genesis: e23395. PubMed ID: 32990387

    Irwin, M., Tare, M., Singh, A., Puli, O. R., Gogia, N., Riccetti, M., Deshpande, P., Kango-Singh, M. and Singh, A. (2020). A Positive Feedback Loop of Hippo- and c-Jun-Amino-Terminal Kinase Signaling Pathways Regulates Amyloid-Beta-Mediated Neurodegeneration. Front Cell Dev Biol 8: 117. PubMed ID: 32232042

    Gogia, N., Sarkar, A., Mehta, A. S., Ramesh, N., Deshpande, P., Kango-Singh, M., Pandey, U. B. and Singh, A. (2020). Inactivation of Hippo and cJun-N-terminal Kinase (JNK) signaling mitigate FUS mediated neurodegeneration in vivo. Neurobiol Dis 140: 104837. PubMed ID: 32199908

    Sarkar, A., Gogia, N., Glenn, N., Singh, A., Jones, G., Powers, N., Srivastava, A., Kango-Singh, M. and Singh, A. (2018). A soy protein Lunasin can ameliorate amyloid-beta 42 mediated neurodegeneration in Drosophila eye. Sci Rep 8(1): 13545. PubMed ID: 30202077

    Sarkar, A., Gogia, N., Farley, K., Payton, L. and Singh, A. (2018). Characterization of a morphogenetic furrow specific Gal4 driver in the developing Drosophila eye. PLoS One 13(4): e0196365. PubMed ID: 29702674

    Tare, M., Sarkar, A., Bedi, S., Kango-Singh, M. and Singh, A. (2016). Cullin-4 regulates Wingless and JNK signaling-mediated cell death in the Drosophila eye. Cell Death Dis 7(12): e2566. PubMed ID: 28032862

    Wittkorn, E., Sarkar, A., Garcia, K., Kango-Singh, M. and Singh, A. (2015). The Hippo pathway effector Yki downregulates Wg signaling to promote retinal differentiation in the Drosophila eye. Development 142: 2002-2013. PubMed ID: 25977365

    Kwon, H. J., Waghmare, I., Verghese, S., Singh, A., Singh, A. and Kango-Singh, M. (2014). Drosophila C-terminal Src kinase regulates growth via the Hippo signaling pathway. Dev Biol. PubMed ID: 25446534

  • Nadia Singh Department of Biological Sciences, North Carolina State University, Raleigh
    Winbush, A. and Singh, N. D. (2020). Genomics of recombination rate variation in temperature-evolved Drosophila melanogaster populations. Genome Biol Evol. PubMed ID: 33247719

    Hunter, C.M., Huang, W., Mackay, T.F. and Singh, N.D. (2016). The genetic architecture of natural variation in recombination Rate in Drosophila melanogaster. PLoS Genet 12: e1005951. PubMed ID: 27035832

    Hunter, C.M., Robinson, M.C., Aylor, D.L. and Singh, N.D. (2016). Genetic background, maternal age and interaction effects mediate rates of crossing over in Drosophila melanogaster females. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 26994290

    O'Shea, K. L. and Singh, N. D. (2015). Tetracycline-exposed Drosophila melanogaster males produce fewer offspring but a relative excess of sons. Ecol Evol 5: 3130-3139. PubMed ID: 26357541

    Jackson, S., Nielsen, D. M. and Singh, N. D. (2015). Increased exposure to acute thermal stress is associated with a non-linear increase in recombination frequency and an independent linear decrease in fitness in Drosophila. BMC Evol Biol 15: 175. PubMed ID: 26310872

    Hunter, C. M. and Singh, N. D. (2014). Do Males Matter? Testing the Effects of Male Genetic Background on Female Meiotic Crossover Rates in Drosophila Melanogaster. Evolution. PubMed ID: 24889512

    Robinson, M. C., Stone, E. A. and Singh, N. D. (2014). Population genomic analysis reveals no evidence for GC-biased gene conversion in Drosophila melanogaster. Mol Biol Evol 31: 425-433. PubMed ID: 24214536

    Singh, N. D., Stone, E. A., Aquadro, C. F. and Clark, A. G. (2013). Fine-scale heterogeneity in crossover rate in the garnet-scalloped region of the Drosophila melanogaster X chromosome. Genetics 194: 375-387. PubMed ID: 23410829

    Singh, N. D., Jensen, J. D., Clark, A. G. and Aquadro, C. F. (2013). Inferences of demography and selection in an African population of Drosophila melanogaster. Genetics 193: 215-228. PubMed ID: 23105013

  • Ravinder Singh Molecular, Cellular and Developmental Biology, University of Colorado Boulder
    Sridharan, V., Heimiller, J., Robida, M. D. and Singh, R. (2016). High throughput sequencing identifies misregulated genes in the Drosophila polypyrimidine tract-binding protein (hephaestus) mutant defective in spermatogenesis. PLoS One 11: e0150768. PubMed ID: 26942929

    Heimiller, J., Sridharan, V., Huntley, J., Wesley, C. S. and Singh, R. (2014). Drosophila Polypyrimidine Tract-Binding Protein (DmPTB) Regulates Dorso-Ventral Patterning Genes in Embryos. PLoS One 9: e98585. PubMed ID: 25014769

    Hartmann, B., Castelo, R., Minana, B., Peden, E., Blanchette, M., Rio, D. C., Singh, R. and Valcarcel, J. (2011). Distinct regulatory programs establish widespread sex-specific alternative splicing in Drosophila melanogaster. RNA 17: 453-468. PubMed ID: 21233220

    Robida, M., Sridharan, V., Morgan, S., Rao, T. and Singh, R. (2010). Drosophila polypyrimidine tract-binding protein is necessary for spermatid individualization. Proc Natl Acad Sci U S A 107: 12570-12575. PubMed ID: 20616016

  • Satpal Singh Department of Pharmacology and Toxicology, University of Buffalo
    Scarpati, M., Qi, Y., Govind, S. and Singh, S. (2019). A combined computational strategy of sequence and structural analysis predicts the existence of a functional eicosanoid pathway in Drosophila melanogaster. PLoS One 14(2): e0211897. PubMed ID: 30753230

    Frolov, R. V. and Singh, S. (2013). Temperature and functional plasticity of L-type Ca channels in Drosophila. Cell Calcium. PubMed ID: 23993048

    Frolov, R. V., Bagati, A., Casino, B. and Singh, S. (2012). Potassium channels in Drosophila: historical breakthroughs, significance, and perspectives. J Neurogenet 26: 275-290. PubMed ID: 23181728

    Frolov, R. V. and Singh, S. (2012). Inhibition of ion channels and heart beat in Drosophila by selective COX-2 inhibitor SC-791. PLoS One 7: e38759. PubMed ID: 22701705

  • Pradip Sinha Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur
    Athilingam, T., Parihar, S. S., Bhattacharya, R., Rizvi, M. S., Kumar, A. and Sinha, P. (2022). Proximate Larval Epidermal Cell Layer Generates Forces for Pupal Thorax Closure in Drosophila. Genetics. PubMed ID: 35166774

    Kumari, J. and Sinha, P. (2021). Developmental expression patterns of toolkit genes in male accessory gland of Drosophila parallels those of mammalian prostate. Biol Open 10(8). PubMed ID: 34342345

    Bajpai, A., Ahmad, Q. T., Tang, H. W., Manzar, N., Singh, V., Thakur, A., Ateeq, B., Perrimon, N. and Sinha, P. (2020). A Drosophila model of oral peptide therapeutics for adult Intestinal Stem Cell tumors. Dis Model Mech 13(7). PubMed ID: 32540914

    Bajpai, A. and Sinha, P. (2019). Hh signaling from de novo organizers drive lgl neoplasia in Drosophila epithelium. Dev Biol. PubMed ID: 31557471

    Gupta, R. P., Bajpai, A. and Sinha, P. (2017). Selector genes display tumor cooperation and inhibition in Drosophila epithelium in a developmental context-dependent manner. Biol Open 6(11): 1581-1591. PubMed ID: 29141951

    Jolly, M. K., Rizvi, M. S., Kumar, A. and Sinha, P. (2014). Mathematical modeling of sub-cellular asymmetry of fat-dachsous heterodimer for generation of planar cell polarity. PLoS One 9: e97641. PubMed ID: 24841507

    Khan, S. J., Bajpai, A., Alam, M. A., Gupta, R. P., Harsh, S., Pandey, R. K., Goel-Bhattacharya, S., Nigam, A., Mishra, A., Sinha, P. (2013) Epithelial neoplasia in Drosophila entails switch to primitive cell states. Proc Natl Acad Sci U S A. PubMed ID: 23708122

    Jaiswal, M., Agrawal, N. and Sinha, P. (2006). Fat and Wingless signaling oppositely regulate epithelial cell-cell adhesion and distal wing development in Drosophila. Development 133: 925-935. PubMed ID: 16452097

  • Saurabh Sinha Dept. of Computer Science, University of Illinois, Urbana-Champaign
    Dibaeinia, P. and Sinha, S. (2021). Deciphering enhancer sequence using thermodynamics-based models and convolutional neural networks. Nucleic Acids Res 49(18): 10309-10327. PubMed ID: 34508359

    Blatti, C., Kazemian, M., Wolfe, S., Brodsky, M. and Sinha, S. (2015). Integrating motif, DNA accessibility and gene expression data to build regulatory maps in an organism. Nucleic Acids Res. PubMed ID: 25791631

    Peng, P. C., Hassan Samee, M. A. and Sinha, S. (2015). Incorporating chromatin accessibility data into sequence-to-expression modeling. Biophys J 108: 1257-1267. PubMed ID: 25762337

    Duque, T., Hassan Samee, M. A., Kazemian, M., Pham, H. N., Brodsky, M. H. and Sinha, S. (2013). Simulations of enhancer evolution provide mechanistic insights into gene regulation. Mol Biol Evol. PubMed ID: 24097306

    Kazemian, M., Pham, H., Wolfe, S. A., Brodsky, M. H. and Sinha, S. (2013). Widespread evidence of cooperative DNA binding by transcription factors in Drosophila development. Nucleic Acids Res. PubMed ID: 23847101

    Abul Hassan Samee, M., Sinha, S. (2013) Evaluating thermodynamic models of enhancer activity on cellular resolution gene expression data. Methods. PubMed ID: 23624421

    Enuameh, M. S., et al. (2013). Global analysis of Drosophila Cys2-His2 zinc finger proteins reveals a multitude of novel recognition motifs and binding determinants. Genome Res. PubMed ID: 23471540

    Bergwitz, C., Rasmussen, M. D., DeRobertis, C., Wee, M. J., Sinha, S., Chen, H. H., Huang, J. and Perrimon, N. (2012). Roles of major facilitator superfamily transporters in phosphate response in Drosophila. PLoS One 7: e31730. PubMed ID: 22359624

    Kazemian, M., Brodsky, M. H. and Sinha, S. (2011). Genome Surveyor 2.0: cis-regulatory analysis in Drosophila. Nucleic Acids Res 39: W79-85. PubMed ID: 21593125

  • Haruhiko Siomi Department of Molecular Biology, Graduate School of Medicine, Keio University, Tokyo
    Iwasaki, Y. W., Sriswasdi, S., Kinugasa, Y., Adachi, J., Horikoshi, Y., Shibuya, A., Iwasaki, W., Tashiro, S., Tomonaga, T. and Siomi, H. (2021). Piwi-piRNA complexes induce stepwise changes in nuclear architecture at target loci. Embo j: e108345. PubMed ID: 34337769

    Onishi, R., Sato, K., Murano, K., Negishi, L., Siomi, H. and Siomi, M. C. (2020). Piwi suppresses transcription of Brahma-dependent transposons via Maelstrom in ovarian somatic cells. Sci Adv 6(50). PubMed ID: 33310860
    Yamaguchi, S., Oe, A., Nishida, K. M., Yamashita, K., Kajiya, A., Hirano, S., Matsumoto, N., Dohmae, N., Ishitani, R., Saito, K., Siomi, H., Nishimasu, H., Siomi, M. C. and Nureki, O. (2020). Crystal structure of Drosophila Piwi. Nat Commun 11(1): 858. PubMed ID: 32051406 Osumi, K., Sato, K., Murano, K., Siomi, H. and Siomi, M. C. (2019). Essential roles of Windei and nuclear monoubiquitination of Eggless/SETDB1 in transposon silencing. EMBO Rep: e48296. PubMed ID: 31576653

    Murano, K., Iwasaki, Y. W., Ishizu, H., Mashiko, A., Shibuya, A., Kondo, S., Adachi, S., Suzuki, S., Saito, K., Natsume, T., Siomi, M. C. and Siomi, H. (2019). Nuclear RNA export factor variant initiates piRNA-guided co-transcriptional silencing. Embo j: e102870. PubMed ID: 31368590

    Hirakata, S., Ishizu, H., Fujita, A., Tomoe, Y. and Siomi, M. C. (2019). Requirements for multivalent Yb body assembly in transposon silencing in Drosophila. EMBO Rep 20(7): e47708. PubMed ID: 31267711

    Ishizu, H., Kinoshita, T., Hirakata, S., Komatsuzaki, C. and Siomi, M. C. (2019). Distinct and collaborative functions of Yb and Armitage in transposon-targeting piRNA biogenesis. Cell Rep 27(6): 1822-1835.e1828. PubMed ID: 31067466

    Yashiro, R., Murota, Y., Nishida, K. M., Yamashiro, H., Fujii, K., Ogai, A., Yamanaka, S., Negishi, L., Siomi, H. and Siomi, M. C. (2018). Piwi nuclear localization and its regulatory mechanism in Drosophila ovarian somatic sells. Cell Rep 23(12): 3647-3657. PubMed ID: 29925005

    Sumiyoshi, T., Sato, K., Yamamoto, H., Iwasaki, Y. W., Siomi, H. and Siomi, M. C. (2016). Loss of l(3)mbt leads to acquisition of the ping-pong cycle in Drosophila ovarian somatic cells. Genes Dev 30: 1617-1622. PubMed ID: 27474440

    Sato, K., Iwasaki, Y. W., Siomi, H. and Siomi, M. C. (2015). Tudor-domain containing proteins act to make the piRNA pathways more robust in Drosophila. Fly (Austin) [Epub ahead of print] PubMed ID: 26647059

    Sato, K., Iwasaki, Y. W., Shibuya, A., Carninci, P., Tsuchizawa, Y., Ishizu, H., Siomi, M. C. and Siomi, H. (2015). Krimper enforces an antisense bias on piRNA pools by binding AGO3 in the Drosophila germline. Mol Cell 59: 553-563. PubMed ID: 26212455

    Ishizu, H., Iwasaki, Y. W., Hirakata, S., Ozaki, H., Iwasaki, W., Siomi, H. and Siomi, M. C. (2015). Somatic Primary piRNA Biogenesis Driven by cis-Acting RNA Elements and trans-Acting Yb. Cell Rep 12: 429-440. PubMed ID: 26166564

  • Mikiko Siomi Department of Molecular Biology, Keio University School of Medicine, Tokyo
    Yamaguchi, S., Oe, A., Nishida, K. M., Yamashita, K., Kajiya, A., Hirano, S., Matsumoto, N., Dohmae, N., Ishitani, R., Saito, K., Siomi, H., Nishimasu, H., Siomi, M. C. and Nureki, O. (2020). Crystal structure of Drosophila Piwi. Nat Commun 11(1): 858. PubMed ID: 32051406

    Murano, K., Iwasaki, Y. W., Ishizu, H., Mashiko, A., Shibuya, A., Kondo, S., Adachi, S., Suzuki, S., Saito, K., Natsume, T., Siomi, M. C. and Siomi, H. (2019). Nuclear RNA export factor variant initiates piRNA-guided co-transcriptional silencing. Embo j: e102870. PubMed ID: 31368590

    Ishizu, H., Sumiyoshi, T. and Siomi, M. C. (2017). Use of the CRISPR-Cas9 system for genome editing in cultured Drosophila ovarian somatic cells. Methods [Epub ahead of print]. PubMed ID: 28552546

    Nishida, K. M., Miyoshi, K., Ogino, A., Miyoshi, T., Siomi, H., Siomi, M. C. (2013) Roles of R2D2, a Cytoplasmic D2 Body Component, in the Endogenous siRNA Pathway in Drosophila. Mol Cell. PubMed ID: 23375501

    Nagao, A., Sato, K., Nishida, K. M., Siomi, H. and Siomi, M. C. (2011). Gender-Specific Hierarchy in Nuage Localization of PIWI-Interacting RNA Factors in Drosophila. Front Genet 2: 55. PubMed ID: 22303351

    Sato, K., Nishida, K. M., Shibuya, A., Siomi, M. C. and Siomi, H. (2011). Maelstrom coordinates microtubule organization during Drosophila oogenesis through interaction with components of the MTOC. Genes Dev 25: 2361-2373. PubMed ID: 22085963

  • Helen Skaer Department of Zoology, University of Cambridge
    Saxena, A., Denholm, B., Bunt, S., Bischoff, M., VijayRaghavan, K. and Skaer, H. (2014). Epidermal growth factor signalling controls Myosin II planar polarity to orchestrate convergent extension movements during Drosophila tubulogenesis. PLoS Biol 12: e1002013. PubMed ID: 25460353

    Weavers, H. and Skaer, H. (2013). Tip cells act as dynamic cellular anchors in the morphogenesis of looped renal tubules in Drosophila. Dev Cell 27: 331-344. PubMed ID: 24229645

    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

    Bunt, S., Hooley, C., Hu, N., Scahill, C., Weavers, H. and Skaer, H. (2010). Hemocyte-secreted type IV collagen enhances BMP signaling to guide renal tubule morphogenesis in Drosophila. Dev Cell 19: 296-306. PubMed ID: 20708591

    Evans, I. R., Hu, N., Skaer, H. and Wood, W. (2010). Interdependence of macrophage migration and ventral nerve cord development in Drosophila embryos. Development 137: 1625-1633. PubMed ID: 20392742

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

    Skeath, J. B., Wilson, B. A., Romero, S. E., Snee, M. J., Zhu, Y. and Lacin, H. (2017). The extracellular metalloprotease AdamTS-A anchors neural lineages in place within and preserves the architecture of the central nervous system. Development 144(17):3102-3113. PubMed ID: 28760813

    Snee, M. J., Wilson, W. C., Zhu, Y., Chen, S. Y., Wilson, B. A., Kseib, C., O'Neal, J., Mahajan, N., Tomasson, M. H., Arur, S. and Skeath, J. B. (2016). Collaborative control of cell cycle progression by the RNA exonuclease Dis3 and Ras is conserved across species. Genetics 203: 749-762. PubMed ID: 27029730

    Yashiro, H., Loza, A. J., Skeath, J. B. and Longmore, G. D. (2014). Rho1 Regulates Adherens Junction Remodeling by Promoting Recycling Endosome Formation through Activation of Myosin II. Mol Biol Cell. PubMed ID: 25079692

    Lacin, H., Rusch, J., Yeh, R. T., Fujioka, M., Wilson, B. A., Zhu, Y., Robie, A. A., Mistry, H., Wang, T., Jaynes, J. B. and Skeath, J. B. (2014). Genome-wide identification of Drosophila Hb9 targets reveals a pivotal role in directing the transcriptome within eight neuronal lineages, including activation of Nitric Oxide Synthase and Fd59a/Fox-D. Dev Biol. PubMed ID: 24512689

    Valakh, V., Walker, L. J., Skeath, J. B. and Diantonio, A. (2013). Loss of the Spectraplakin Short Stop Activates the DLK Injury Response Pathway in Drosophila. J Neurosci 33: 17863-17873. PubMed ID: 24198375

    Guss, K. A., Benson, M., Gubitosi, N., Brondell, K., Broadie, K., Skeath, J. B. (2013) Expression and function of scalloped during drosophila development. Dev Dyn. PubMed ID: 23389965

    Park, D., Hadzic, T., Yin, P., Rusch, J., Abruzzi, K., Rosbash, M., Skeath, J. B., Panda, S., Sweedler, J. V. and Taghert, P. H. (2011). Molecular organization of Drosophila neuroendocrine cells by Dimmed. Curr Biol 21: 1515-1524. PubMed ID: 21885285

    Das Thakur, M., Feng, Y., Jagannathan, R., Seppa, M. J., Skeath, J. B. and Longmore, G. D. (2010). Ajuba LIM proteins are negative regulators of the Hippo signaling pathway. Curr Biol 20: 657-662. PubMed ID: 20303269

    Babaoglan, A. B., O'Connor-Giles, K. M., Mistry, H., Schickedanz, A., Wilson, B. A. and Skeath, J. B. (2009). Sanpodo: a context-dependent activator and inhibitor of Notch signaling during asymmetric divisions. Development 136: 4089-4098. PubMed ID: 19906847

  • Efthimios Skoulakis Institute of Molecular Biology & Genetics, Biomedical Sciences Research Center Alexander Fleming, Vari, near Athens
    Prifti, E., Tsakiri, E. N., Vourkou, E., Stamatakis, G., Samiotaki, M., Skoulakis, E. M. C. and Papanikolopoulou, K. (2022). Mical modulates Tau toxicity via cysteine oxidation in vivo. Acta Neuropathol Commun 10(1): 44. PubMed ID: 35379354

    Georganta, E. M., Moressis, A. and Skoulakis, E. M. (2021). Associative learning requires Neurofibromin to modulate GABAergic inputs to Drosophila Mushroom Bodies. J Neurosci. PubMed ID: 33972401

    Keramidis, I., Vourkou, E., Papanikolopoulou, K. and Skoulakis, E. M. C. (2020). Functional Interactions of Tau Phosphorylation Sites That Mediate Toxicity and Deficient Learning in Drosophila melanogaster. Front Mol Neurosci 13: 569520. PubMed ID: 33192295

    Roussou, I. G., Papanikolopoulou, K., Savakis, C. and Skoulakis, E. M. C. (2019). Drosophila Bruton's Tyrosine Kinase regulates habituation latency and facilitation in distinct mushroom body neurons. J Neurosci. PubMed ID: 31530645

    Papanikolopoulou, K., Roussou, I. G., Gouzi, J. Y., Samiotaki, M., Panayotou, G., Turin, L. and Skoulakis, E. M. (2019). Drosophila Tau negatively regulates translation and olfactory Long-Term Memory, but facilitates footshock habituation and cytoskeletal homeostasis. J Neurosci. PubMed ID: 31488613

    Kadas, D., Papanikolopoulou, K., Xirou, S., Consoulas, C. and Skoulakis, E. M. C. (2019). Human Tau isofSharma, A.orm-specific presynaptic deficits in a Drosophila central nervous system circuit. Neurobiol Dis 124: 311-321. PubMed ID: 30529489

    Semelidou, O., Acevedo, S. F. and Skoulakis, E. M. (2018). Temporally specific engagement of distinct neuronal circuits regulating olfactory habituation in Drosophila. Elife 7. PubMed ID: 30576281

    Kadas, D., Papanikolopoulou, K., Xirou, S., Consoulas, C. and Skoulakis, E. M. C. (2018). Human Tau isoform-specific presynaptic deficits in a Drosophila central nervous system circuit. Neurobiol Dis 124: 311-321. PubMed ID: 30529489

    Gouzi, J. Y., Bouraimi, M., Roussou, I. G., Moressis, A. and Skoulakis, E. M. C. (2018). The Drosophila Receptor Tyrosine Kinase Alk constrains long-term memory formation. J Neurosci. PubMed ID: 30030398

    Papanikolopoulou, K., Grammenoudi, S., Samiotaki, M. and Skoulakis, E. M. C. (2018). Differential effects of 14-3-3 dimers on Tau phosphorylation, stability and toxicity in vivo. Hum Mol Genet. PubMed ID: 29659825

    Maniati, K., Haralambous, K. J., Turin, L. and Skoulakis, E. M. C. (2017). Vibrational Detection of Odorant Functional Groups by Drosophila melanogaster. eNeuro 4(5). PubMed ID: 29094064

  • Kevin Slep Department of Biology, University of North Carolina, Chapel Hill
    Adikes, R. C., Hallett, R. A., Saway, B. F., Kuhlman, B. and Slep, K. C. (2017). Control of microtubule dynamics using an optogenetic microtubule plus end-F-actin cross-linker. J Cell Biol. PubMed ID: 29259096

    Byrnes, A. E. and Slep, K. C. (2017). TOG-tubulin binding specificity promotes microtubule dynamics and mitotic spindle formation. J Cell Biol [Epub ahead of print]. PubMed ID: 28512144

    Plevock, K. M., Galletta, B. J., Slep, K. C. and Rusan, N. M. (2015). Newly Characterized Region of CP190 Associates with Microtubules and Mediates Proper Spindle Morphology in Drosophila Stem Cells. PLoS One 10(12): e0144174. PubMed ID: 26649574

    Howard, A. E., Fox, J. C. and Slep, K. C. (2015). Drosophila melanogaster mini spindles TOG3 utilizes unique structural elements to promote domain stability and maintain a TOG1- and TOG2-like tubulin-binding surface. J Biol Chem 290(16): 10149-10162. PubMed ID: 25720490

    Currie, J. D., Stewman, S., Schimizzi, G., Slep, K. C., Ma, A. and Rogers, S. L. (2011). The microtubule lattice and plus-end association of Drosophila Mini spindles is spatially regulated to fine-tune microtubule dynamics. Mol Biol Cell 22(22): 4343-4361. PubMed ID: 21965297 x

  • Stephen Small Department of Biology, New York University
    Onal, P., Imaya Gunasinghe, H., Yui Umezawa, K., Zheng, M., Ling, J., Azeez, L., Dalmeus, A., Tazin, T. and Small, S. (2021). Suboptimal Intermediates Underlie Evolution of the Bicoid Homeodomain. Mol Biol Evol. PubMed ID: 33599280

    Datta, R. R., Ling, J., Kurland, J., Ren, X., Xu, Z., Yucel, G., Moore, J., Shokri, L., Baker, I., Bishop, T., Struffi, P., Levina, R., Bulyk, M. L., Johnston, R. J., Jr. and Small, S. (2018). A feed-forward relay integrates the regulatory activities of Bicoid and Orthodenticle via sequential binding to suboptimal sites. Genes Dev 32(9-10): 723-736. PubMed ID: 29764918

    Yan, J., Anderson, C., Viets, K., Tran, S., Goldberg, G., Small, S. and Johnston, R. J., (2017). Regulatory logic driving stable levels of defective proventriculus expression during terminal photoreceptor specification in flies. Development [Epub ahead of print]. PubMed ID: 28126841

    Xu, Z., Chen, H., Ling, J., Yu, D., Struffi, P. and Small, S. (2014). Impacts of the ubiquitous factor Zelda on Bicoid-dependent DNA binding and transcription in Drosophila. Genes Dev 28: 608-621. PubMed ID: 24637116

    Struffi, P., Corado, M., Kaplan, L., Yu, D., Rushlow, C. and Small, S. (2011). Combinatorial activation and concentration-dependent repression of the Drosophila even skipped stripe 3+7 enhancer. Development 138: 4291-4299. PubMed ID: 21865322

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

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

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

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

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

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

    Chen, L., Dumelie, J. G., Li, X., Cheng, M. H., Yang, Z., Laver, J. D., Siddiqui, N. U., Westwood, J. T., Morris, Q., Lipshitz, H. D. and Smibert, C. A. (2014). Global regulation of mRNA translation and stability in the early Drosophila embryo by the Smaug RNA-binding protein. Genome Biol 15: R4. PubMed ID: 24393533

    Pinder, B. D., Smibert, C. A. (2013) Smaug: An unexpected journey into the mechanisms of post-transcriptional regulation. Fly (Austin) 7. PubMed ID: 23519205

    Pinder, B. D. and Smibert, C. A. (2012). microRNA-independent recruitment of Argonaute 1 to nanos mRNA through the Smaug RNA-binding protein. EMBO Rep. PubMed ID: 23184089

    Laver, J. D., Ancevicius, K., Sollazzo, P., Westwood, J. T., Sidhu, S. S., Lipshitz, H. D. and Smibert, C. A. (2012). Synthetic antibodies as tools to probe RNA-binding protein function. Mol Biosyst 8: 1650-1657. PubMed ID: 22481296

  • Dean Smith Departments of Pharmacology and Neuroscience, University of Texas Southwestern Medical Center, Dallas
    Poudel, S., Guo, H. and Smith, D. P. (2021). PKC98E Regulates Odorant Responses in Drosophila melanogaster. J Neurosci 41(18): 3948-3957. PubMed ID: 33789918

    Scheuermann, E. A. and Smith, D. P. (2019). Odor-specific deactivation defects in a Drosophila odorant binding protein mutant. Genetics. PubMed ID: 31492805

    Guo, H., Kunwar, K. and Smith, D. (2019). Multiple channels of DEET repellency in Drosophila. Pest Manag Sci. PubMed ID: 31429190

    Guo, H., Kunwar, K. and Smith, D. (2017). Odorant receptor sensitivity modulation in Drosophila. J Neurosci 37(39): 9465-9473. PubMed ID: 28871035

    Pitts, S., Pelser, E., Meeks, J. and Smith, D. (2016). Odorant responses and courtship behaviors influenced by at4 neurons in Drosophila. PLoS One 11: e0162761. PubMed ID: 27617442

    Ha, T. S., Xia, R., Zhang, H., Jin, X. and Smith, D. P. (2014). Lipid flippase modulates olfactory receptor expression and odorant sensitivity in Drosophila. Proc Natl Acad Sci U S A. PubMed ID: 24821794

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

    Sanders, C. and Smith, D. P. (2011). LUMP is a putative double-stranded RNA binding protein required for male fertility in Drosophila melanogaster. PLoS One 6: e24151. PubMed ID: 21912621

    Kwon, Y., Kim, S. H., Ronderos, D. S., Lee, Y., Akitake, B., Woodward, O. M., Guggino, W. B., Smith, D. P. and Montell, C. (2010). Drosophila TRPA1 channel is required to avoid the naturally occurring insect repellent citronellal. Curr Biol 20: 1672-1678. PubMed ID: 20797863

    Ronderos, D. S. and Smith, D. P. (2010). Activation of the T1 neuronal circuit is necessary and sufficient to induce sexually dimorphic mating behavior in Drosophila melanogaster. J Neurosci 30: 2595-2599. PubMed ID: 20164344

  • Rachel Smith-Bolton Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign
    Tian, Y. and Smith-Bolton, R. K. (2021). Regulation of growth and cell fate during tissue regeneration by the two SWI/SNF chromatin-remodeling complexes of Drosophila. Genetics 217(1): 1-16. PubMed ID: 33683366

    Abidi, S. N. F. and Smith-Bolton, R. K. (2018). Cell fate changes induced by a Distal-less enhancer-trap transgene in the Drosophila antennal imaginal disc. Sci Rep 8(1): 4950. PubMed ID: 29563503

    Khan, S. J., Abidi, S. N. F., Skinner, A., Tian, Y. and Smith-Bolton, R. K. (2017). The Drosophila Duox maturation factor is a key component of a positive feedback loop that sustains regeneration signaling. PLoS Genet 13(7): e1006937. PubMed ID: 28753614

    Brock, A. R., Seto, M. and Smith-Bolton, R. K. (2017). Cap-n-Collar Promotes Tissue Regeneration by Regulating ROS and JNK Signaling in the Drosophila melanogaster Wing Imaginal Disc. Genetics 206(3): 1505-1520. PubMed ID: 28512185

    Khan, S. J., Abidi, S. N., Tian, Y., Skinner, A. and Smith-Bolton, R. K. (2016). A rapid, gentle and scalable method for dissociation and fluorescent sorting of imaginal disc cells for mRNA sequencing. Fly (Austin) 10(2): 73-80. PubMed ID: 27057746

  • Thomas Smithgall Department of Molecular Biology and Genetics, University of Pittsburgh School of Medicine
    O'Reilly, L. P., Zhang, X. and Smithgall, T. E. (2012). Individual Src-family tyrosine kinases direct the degradation or protection of the clock protein Timeless via differential ubiquitylation. Cell Signal. PubMed ID: 23266470

    O'Reilly, L. P., Watkins, S. C. and Smithgall, T. E. (2011). An unexpected role for the clock protein timeless in developmental apoptosis. PLoS One 6: e17157. PubMed ID: 21359199

  • Rhonda Snook Department of Animal and Plant Sciences, University of Sheffield
    Rodrigues, L. R., Zwoinska, M. K., Axel, W. W. R. and Snook, R. R. (2022). The genetic basis and adult reproductive consequences of developmental thermal plasticity. J Anim Ecol. PubMed ID: 35060127

    Rodrigues, L. R., McDermott, H. A., Villanueva, I., Djukaric, J., Ruf, L. C., Amcoff, M. and Snook, R. R. (2021). Fluctuating heat stress during development exposes reproductive costs and putative benefits. J Anim Ecol. PubMed ID: 34775602

    Garlovsky, M. D., Evans, C., Rosenow, M. A., Karr, T. L. and Snook, R. R. (2020). Seminal fluid protein divergence among populations exhibiting postmating prezygotic reproductive isolation. Mol Ecol. PubMed ID: 32939895

    Fitzpatrick, J. L., Bridge, C. D. and Snook, R. R. (2020). Repeated evidence that the accelerated evolution of sperm is associated with their fertilization function. Proc Biol Sci 287(1932): 20201286. PubMed ID: 32752988

    Garlovsky, M. D. and Snook, R. R. (2018). Persistent postmating, prezygotic reproductive isolation between populations. Ecol Evol 8(17): 9062-9073. PubMed ID: 30271566

    Porcelli, D., Gaston, K.J., Butlin, R.K. and Snook, R.R. (2016). Local adaptation of reproductive performance during thermal stress. J Evol Biol [Epub ahead of print]. PubMed ID: 27862539

    Porcelli, D., Westram, A. M., Pascual, M., Gaston, K. J., Butlin, R. K. and Snook, R. R. (2016). Gene expression clines reveal local adaptation and associated trade-offs at a continental scale. Sci Rep 6: 32975. PubMed ID: 27599812

    Immonen, E., Snook, R. R. and Ritchie, M. G. (2014). Mating system variation drives rapid evolution of the female transcriptome in Drosophila pseudoobscura. Ecol Evol 4: 2186-2201. PubMed ID: 25360260

    Jennings, J. H., Snook, R. R. and Hoikkala, A. (2014). Reproductive isolation among allopatric Drosophila montana populations. Evolution 68: 3095-3108. PubMed ID: 25302639

    Snook, R. R., Gidaszewski, N. A., Chapman, T. and Simmons, L. W. (2013). Sexual selection and the evolution of secondary sexual traits: sex comb evolution in Drosophila. J Evol Biol 26: 912-918. PubMed ID: 23496332

  • Peter Soba Center for Molecular Neurobiology (ZMNH), University of Hamburg
    Hu, C., Feng, P., Chen, M., Tang, Y. and Soba, P. (2022). Spatiotemporal changes in microtubule dynamics during dendritic morphogenesis. Fly (Austin) 16(1): 13-23. PubMed ID: 34609266

    Imambocus, B. N., Zhou, F., Formozov, A., Wittich, A., Tenedini, F. M., Hu, C., Sauter, K., Macarenhas Varela, E., Heredia, F., Casimiro, A. P., Macedo, A., Schlegel, P., Yang, C. H., Miguel-Aliaga, I., Wiegert, J. S., Pankratz, M. J., Gontijo, A. M., Cardona, A. and Soba, P. (2021). A neuropeptidergic circuit gates selective escape behavior of Drosophila larvae. Curr Biol. PubMed ID: 34798050

    Hu, C., Kanellopoulos, A. K., Richter, M., Petersen, M., Konietzny, A., Tenedini, F., Hoyer, N., Cheng, L., Poon, C. L. C., Harvey, K. F., Windhorst, S., Parrish, J. Z., Mikhaylova, M., Bagni, C., Calderon de Anda, F. and Soba, P. (2020). Conserved Tao kinase activity regulates dendritic arborization, cytoskeletal dynamics and sensory function in Drosophila. J Neurosci. PubMed ID: 31964717

    Tenedini, F. M., Saez Gonzalez, M., Hu, C., Pedersen, L. H., Petruzzi, M. M., Spitzweck, B., Wang, D., Richter, M., Petersen, M., Szpotowicz, E., Schweizer, M., Sigrist, S. J., Calderon de Anda, F. and Soba, P. (2019). Maintenance of cell type-specific connectivity and circuit function requires Tao kinase. Nat Commun 10(1): 3506. PubMed ID: 31383864

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

    Hu, C., Petersen, M., Hoyer, N., Spitzweck, B., Tenedini, F., Wang, D., Gruschka, A., Burchardt, L. S., Szpotowicz, E., Schweizer, M., Guntur, A. R., Yang, C. H. and Soba, P. (2017). Sensory integration and neuromodulatory feedback facilitate Drosophila mechanonociceptive behavior. Nat Neurosci 20(8): 1085-1095. PubMed ID: 28604684

    Meltzer, S., Yadav, S., Lee, J., Soba, P., Younger, S. H., Jin, P., Zhang, W., Parrish, J., Jan, L. Y. and Jan, Y. N. (2016). Epidermis-Derived Semaphorin Promotes Dendrite Self-Avoidance by Regulating Dendrite-Substrate Adhesion in Drosophila Sensory Neurons. Neuron 89(4): 741-755. PubMed ID: 26853303

    Han, C., Wang, D., Soba, P., Zhu, S., Lin, X., Jan, L. Y. and Jan, Y. N. (2012). Integrins regulate repulsion-mediated dendritic patterning of drosophila sensory neurons by restricting dendrites in a 2D space. Neuron 73(1): 64-78. PubMed ID: 22243747

  • Anna Marie Sokac Department of Biochemistry and Molecular Biology, Baylor
    Figard, L., Wang, M., Zheng, L., Golding, I. and Sokac, A. M. (2016). Membrane supply and demand regulates F-Actin in a cell surface reservoir. Dev Cell 37: 267-278. PubMed ID: 27165556

    Figard, L., Xu, H., Garcia, H. G., Golding, I. and Sokac, A. M. (2013). The Plasma Membrane Flattens Out to Fuel Cell-Surface Growth during Drosophila Cellularization. Dev Cell. PubMed ID: 24316147

    Zheng, L., Sepulveda, L. A., Lua, R. C., Lichtarge, O., Golding, I. and Sokac, A. M. (2013). The Maternal-to-Zygotic Transition Targets Actin to Promote Robustness during Morphogenesis. PLoS Genet 9: e1003901. PubMed ID: 24244181

    Figard, L. and Sokac, A. M. (2011). Imaging cell shape change in living Drosophila embryos. J Vis Exp. PubMed ID: 21490577

    Sokac, A. M. and Wieschaus, E. (2008). Local actin-dependent endocytosis is zygotically controlled to initiate Drosophila cellularization. Dev Cell 14: 775-786. PubMed ID: 18477459

  • Nicholas Sokol Department of Biology, Indiana University
    Ariyapala, I. S., Buddika, K., Hundley, H. A., Calvi, B. R. and Sokol, N. S. (2022). The RNA binding protein Swm is critical for Drosophila melanogaster intestinal progenitor cell maintenance. Genetics. PubMed ID: 35762963

    Buddika, K., Huang, Y. T., Ariyapala, I. S., Butrum-Griffith, A., Norrell, S. A., O'Connor, A. M., Patel, V. K., Rector, S. A., Slovan, M., Sokolowski, M., Kato, Y., Nakamura, A. and Sokol, N. S. (2022). Coordinated repression of pro-differentiation genes via P-bodies and transcription maintains Drosophila intestinal stem cell identity. Curr Biol 32(2): 386-397.e386. PubMed ID: 34875230

    Buddika, K., Xu, J., Ariyapala, I. S. and Sokol, N. S. (2021). I-KCKT allows dissection-free RNA profiling of adult Drosophila intestinal progenitor cells. Development 148(1). PubMed ID: 33246929

    Ariyapala, I. S., Holsopple, J. M., Popodi, E. M., Hartwick, D. G., Kahsai, L., Cook, K. R. and Sokol, N. S. (2020). Identification of Split-GAL4 Drivers and Enhancers That Allow Regional Cell Type Manipulations of the Drosophila melanogaster Intestine. Genetics. PubMed ID: 32988987

    Buddika, K., Ariyapala, I. S., Hazuga, M. A., Riffert, D. and Sokol, N. S. (2020). Canonical nucleators are dispensable for stress granule assembly in intestinal progenitors. J Cell Sci. PubMed ID: 32265270

    Wu, Y. C., Chawla, G. and Sokol, N. (2020). let-7-complex microRNAs regulate Broad-Z3, which together with Chinmo maintains adult lineage neurons in an immature state. G3 (Bethesda). PubMed ID: 32071070

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

    Luhur, A., Buddika, K., Ariyapala, I. S., Chen, S. and Sokol, N. S. (2017). Opposing post-transcriptional control of InR by FMRP and LIN-28 adjusts stem cell-based tissue growth. Cell Rep 21(10): 2671-2677. PubMed ID: 29212015

    Chawla, G., Deosthale, P., Childress, S., Wu, Y. C. and Sokol, N. S. (2016). A let-7-to-miR-125 MicroRNA Switch Regulates Neuronal Integrity and Lifespan in Drosophila. PLoS Genet 12: e1006247. PubMed ID: 27508495

    Chen, C. H., Luhur, A. and Sokol, N. (2015). Lin-28 promotes symmetric stem cell division and drives adaptive growth in the adult Drosophila intestine. Development 142: 3478-3487. PubMed ID: 26487778

    Chawla, G. and Sokol, N. S. (2014). ADAR mediates differential expression of polycistronic microRNAs. Nucleic Acids Res. PubMed ID: 24561617

    Luhur, A., Chawla, G., Wu, Y. C., Li, J. and Sokol, N. S. (2014). Drosha-independent DGCR8/Pasha pathway regulates neuronal morphogenesis. Proc Natl Acad Sci U S A 111: 1421-1426. PubMed ID: 24474768

  • Marla Sokolowski University of Toronto at Mississauga
    Allen, A. M. and Sokolowski, M. B. (2021). Expression of the foraging gene in adult Drosophila melanogaster. J Neurogenet: 1-21. PubMed ID: 34382904

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

    Anreiter, I., Allen, A. M., Vasquez, O. E., To, L., Douglas, S. J., Alvarez, J. V., Ewer, J. and Sokolowski, M. B. (2021). The Drosophila foraging gene plays a vital role at the start of metamorphosis for subsequent adult emergence. J Neurogenet: 1-13. PubMed ID: 33944658

    Gray, L. J., Sokolowski, M, B. and Simpson, J. S. (2021). Drosophila as a useful model for understanding the evolutionary physiology of obesity resistance and metabolic thrift. Fly (Austin) 15(1): 47-59. PubMed ID: 33704003

    Dason, J. S., Cheung, A., Anreiter, I., Montemurri, V. A., Allen, A. M. and Sokolowski, M. B. (2019). Drosophila melanogaster foraging regulates a nociceptive-like escape behavior through a developmentally plastic sensory circuit. Proc Natl Acad Sci U S A. PubMed ID: 31213548

    Struk, A. A., Mugon, J., Huston, A., Scholer, A. A., Stadler, G., Higgins, E. T., Sokolowski, M. B. and Danckert, J. (2019). Self-regulation and the foraging gene (PRKG1) in humans. Proc Natl Acad Sci U S A. PubMed ID: 30782798

    Dason, J. S., Allen, A. M., Vasquez, O. E. and Sokolowski, M. B. (2019). Distinct functions of a cGMP-dependent protein kinase in nerve terminal growth and synaptic vesicle cycling. J Cell Sci. PubMed ID: 30837290

    Allen, A. M., Anreiter, I., Vesterberg, A., Douglas, S. J. and Sokolowski, M. B. (2018). Pleiotropy of the Drosophila melanogaster foraging gene on larval feeding-related traits. J Neurogenet: 1-11. PubMed ID: 30303018

    Anreiter, I., Kramer, J. M. and Sokolowski, M. B. (2017). Epigenetic mechanisms modulate differences in Drosophila foraging behavior. Proc Natl Acad Sci U S A 114(47): 12518-12523. PubMed ID: 29078350

    Wang, S. and Sokolowski, M. B. (2017). Aggressive behaviours, food deprivation and the foraging gene. R Soc Open Sci 4(4): 170042. PubMed ID: 28484630

    Allen, A. M., Anreiter, I., Neville, M. C. and Sokolowski, M. B. (2016). Feeding-Related Traits Are Affected by Dosage of the foraging Gene in Drosophila melanogaster. Genetics [Epub ahead of print]. PubMed ID: 28007892

    Riedl, C. A., Oster, S., Busto, M., Mackay, T. F. and Sokolowski, M. B. (2016). Natural variability in Drosophila larval and pupal NaCl tolerance. J Insect Physiol 88: 15-23. PubMed ID: 26874056

  • Cedric Soler Faculty of medicine, Universite Clermont Auvergne
    Ribot, C., Soler, C., Chartier, A., Al Hayek, S., Nait-Saidi, R., Barbezier, N., Coux, O. and Simonelig, M. (2022). Activation of the ubiquitin-proteasome system contributes to oculopharyngeal muscular dystrophy through muscle atrophy. PLoS Genet 18(1): e1010015. PubMed ID: 35025870

    Laurichesse, Q., Moucaud, B., Laddada, L., Renaud, Y., Jagla, K. and Soler, C. (2021). Transcriptomic and Genetic Analyses Identify the Kruppel-Like Factor Dar1 as a New Regulator of Tube-Shaped Long Tendon Development. Front Cell Dev Biol 9: 747563. PubMed ID: 34977007

    Ghasemizadeh, A., Christin, E., Guiraud, A., Couturier, N., Abitbol, M., Risson, V., Girard, E., Jagla, C., Soler, C., Laddada, L., Sanchez, C., Jaque-Fernandez, F. I., Jacquemond, V., Thomas, J. L., Lanfranchi, M., Courchet, J., Gondin, J., Schaeffer, L. and Gache, V. (2021). MACF1 controls skeletal muscle function through the microtubule-dependent localization of extra-synaptic myonuclei and mitochondria biogenesis. Elife 10. PubMed ID: 34448452

    Laddada, L., Jagla, K. and Soler, C. (2019). Odd-skipped and Stripe act downstream of Notch to promote the morphogenesis of long appendicular tendons in Drosophila. Biol Open 8(3). PubMed ID: 30796048

  • Matthias Soller School of Biosciences, University of Birmingham
    Haussmann, I. U., Wu, Y., Nallasivan, M. P., Archer, N., Bodi, Z., Hebenstreit, D., Waddell, S., Fray, R. and Soller, M. (2022). CMTr cap-adjacent 2'-O-ribose mRNA methyltransferases are required for reward learning and mRNA localization to synapses. Nat Commun 13(1): 1209. PubMed ID: 35260552

    Bawankar, P., Lence, T., Paolantoni, C., Haussmann, I. U., Kazlauskiene, M., Jacob, D., Heidelberger, J. B., Richter, F. M., Nallasivan, M. P., Morin, V., Kreim, N., Beli, P., Helm, M., Jinek, M., Soller, M. and Roignant, J. Y. (2021). Hakai is required for stabilization of core components of the m(6)A mRNA methylation machinery. Nat Commun 12(1): 3778. PubMed ID: 34145251

    Munafo, M., Lawless, V. R., Passera, A., MacMillan, S., Bornelov, S., Haussmann, I. U., Soller, M., Hannon, G. J. and Czech, B. (2021). Channel Nuclear Pore Complex subunits are required for transposon silencing in Drosophila. Elife 10. PubMed ID: 33856346

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

    Ustaoglu, P., Haussmann, I. U., Liao, H., Torres-Mendez, A., Arnold, R., Irimia, M. and Soller, M. (2019). Srrm234, but not canonical SR and hnRNP proteins drive inclusion of Dscam exon 9 variable exons. Rna. PubMed ID: 31292260

    Haussmann, I. U., Ustaoglu, P., Brauer, U., Hemani, Y., Dix, T. C. and Soller, M. (2018). Plasmid-based gap-repair recombineered transgenes reveal a central role for introns in mutually exclusive alternative splicing in Down Syndrome Cell Adhesion Molecule exon 4. Nucleic Acids Res. PubMed ID: 30541104

    Knuckles, P., Lence, T., Haussmann, I. U., Jacob, D., Kreim, N., Carl, S. H., Masiello, I., Hares, T., Villasenor, R., Hess, D., Andrade-Navarro, M. A., Biggiogera, M., Helm, M., Soller, M., Buhler, M. and Roignant, J. Y. (2018). Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m(6)A machinery component Wtap/Fl(2)d. Genes Dev. PubMed ID: 29535189

    Haussmann, I. U., Bodi, Z., Sanchez-Moran, E., Mongan, N. P., Archer, N., Fray, R. G. and Soller, M. (2016). m6A potentiates Sxl alternative pre-mRNA splicing for robust Drosophila sex determination. Nature 540(7632): 301-304. PubMed ID: 27919081

    Zaharieva, E., Haussmann, I. U., Brauer, U. and Soller, M. (2015). Concentration and localization of co-expressed ELAV/Hu proteins control specificity of mRNA processing. Mol Cell Biol [Epub ahead of print]. PubMed ID: 26124284

    Haussmann, I. U., Hemani, Y., Wijesekera, T., Dauwalder, B. and Soller, M. (2013). Multiple pathways mediate the sex-peptide-regulated switch in female Drosophila reproductive behaviours. Proc Biol Sci 280: 20131938. PubMed ID: 24089336

    Hemani, Y. and Soller, M. (2012). Mechanisms of Drosophila Dscam mutually exclusive splicing regulation. Biochem Soc Trans 40: 804-809. PubMed ID: 22817738

  • Haiyun Song Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences
    Li, P., Ma, Z., Yu, Y., Hu, X., Zhou, Y. and Song, H. (2019). FER promotes cell migration via regulating JNK activity. Cell Prolif: e12656. PubMed ID: 31264309

    Ma, Z., Li, P., Hu, X. and Song, H. (2018). Polarity protein Canoe mediates overproliferation via modulation of JNK, Ras-MAPK and Hippo signalling. Cell Prolif: e12529. PubMed ID: 30328653

    Huang, D., Li, X., Sun, L., Huang, P., Ying, H., Wang, H., Wu, J. and Song, H. (2016). Regulation of Hippo signalling by p38 signalling. J Mol Cell Biol [Epub ahead of print]. PubMed ID: 27402810

    Feng, Y., Li, X., Ray, L., Song, H., Qu, J., Lin, S. and Lin, X. (2014). The Drosophila tankyrase regulates Wg signaling depending on the concentration of Daxin. Cell Signal 26: 1717-1724. PubMed ID: 24768997

    Wang, B., Chen, N., Wei, Y., Li, J., Sun, L., Wu, J., Huang, Q., Liu, C., Fan, C. and Song, H. (2012). Akt signaling-associated metabolic effects of dietary gold nanoparticles in Drosophila. Sci Rep 2: 563. PubMed ID: 22872808

  • Yun S. Song Integrative Biology, University of California, Berkeley
    Li, F., Lo, T. Y., Miles, L., Wang, Q., Noristani, H. N., Li, D., Niu, J., Trombley, S., Goldshteyn, J. I., Wang, C., Wang, S., Qiu, J., Pogoda, K., Mandal, K., Brewster, M., Rompolas, P., He, Y., Janmey, P. A., Thomas, G. M., Li, S. and Song, Y. (2021). The Atr-Chek1 pathway inhibits axon regeneration in response to Piezo-dependent mechanosensation. Nat Commun 12(1): 3845. PubMed ID: 34158506

    Li, F., Sami, A., Noristani, H. N., Slattery, K., Qiu, J., Groves, T., Wang, S., Veerasammy, K., Chen, Y. X., Morales, J., Haynes, P., Sehgal, A., He, Y., Li, S. and Song, Y. (2020). Glial Metabolic Rewiring Promotes Axon Regeneration and Functional Recovery in the Central Nervous System. Cell Metab. PubMed ID: 32941799

    Liu, X., Shen, J., Xie, L., Wei, Z., Wong, C., Li, Y., Zheng, X., Li, P. and Song, Y. (2019). Mitotic implantation of the transcription factor Prospero via phase separation drives terminal neuronal differentiation. Dev Cell. PubMed ID: 31866201

    Sheehan, S. and Song, Y. S. (2016). Deep learning for population genetic inference. PLoS Comput Biol 12: e1004845. PubMed ID: 27018908

    Chan, A. H., Jenkins, P. A. and Song, Y. S. (2012). Genome-wide fine-scale recombination rate variation in Drosophila melanogaster. PLoS Genet 8: e1003090. PubMed ID: 23284288

    Langley, C. H., Stevens, K., Cardeno, C., Lee, Y. C., Schrider, D. R., Pool, J. E., Langley, S. A., Suarez, C., Corbett-Detig, R. B., Kolaczkowski, B., Fang, S., Nista, P. M., Holloway, A. K., Kern, A. D., Dewey, C. N., Song, Y. S., Hahn, M. W. and Begun, D. J. (2012). Genomic variation in natural populations of Drosophila melanogaster. Genetics 192: 533-598. PubMed ID: 22673804

  • Jesper Sorensen Department of Bioscience, Aarhus University, Denmark
    Salachan, P. V. and Sorensen, J. G. (2022). Molecular mechanisms underlying plasticity in a thermally varying environment. Mol Ecol. PubMed ID: 35397190

    Salachan, P. V. and Sorensen, J. G. (2017). Critical thermal limits affected differently by developmental and adult thermal fluctuations. J Exp Biol 220(Pt 23): 4471-4478. PubMed ID: 28982965

    Manenti, T., Loeschcke, V., Moghadam, N. N. and Sorensen, J. G. (2015). Phenotypic plasticity is not affected by experimental evolution in constant, predictable or unpredictable fluctuating thermal environments. J Evol Biol. PubMed ID: 26299271

    Colinet, H., Overgaard, J., Com, E., Sorensen, J. G. (2013) Proteomic profiling of thermal acclimation in Drosophila melanogaster. Insect Biochem Mol Biol. PubMed ID: 23416132

    Sorensen, J. G., Loeschcke, V. and Kristensen, T. N. (2013). Cellular damage as induced by high temperature is dependent on rate of temperature change - investigating consequences of ramping rates on molecular and organismal phenotypes in Drosophila melanogaster. J Exp Biol 216: 809-814. PubMed ID: 23155086

    Overgaard, J., Kristensen, T. N. and Sorensen, J. G. (2012). Validity of thermal ramping assays used to assess thermal tolerance in arthropods. PLoS One 7: e32758. PubMed ID: 22427876

    Calabria, G., Dolgova, O., Rego, C., Castaneda, L. E., Rezende, E. L., Balanya, J., Pascual, M., Sorensen, J. G., Loeschcke, V. and Santos, M. (2012). Hsp70 protein levels and thermotolerance in Drosophila subobscura: a reassessment of the thermal co-adaptation hypothesis. J Evol Biol 25: 691-700. PubMed ID: 22300519

  • Sol Sotillos Centro Andaluz de Biología del Desarrollo
    Sotillos, S., Aguilar-Aragon, M. and Hombria, J. C. (2018). Functional analysis of the Drosophila RhoGAP Cv-c protein and its equivalence to the human DLC3 and DLC1 proteins. Sci Rep 8(1): 4601. PubMed ID: 29545526

    Calero-Cuenca, F. J. and Sotillos, S. (2016). Nuf and Rip11 requirement for polarity determinant recycling during Drosophila development. Small GTPases: 1-8. PubMed ID: 27687567

    Sanchez-Higueras, C., Sotillos, S. and Castelli-Gair Hombria, J. (2013). Common Origin of Insect Trachea and Endocrine Organs from a Segmentally Repeated Precursor. Curr Biol. PubMed ID: 24332544

    Hombria, J. C. and Sotillos, S. (2013). JAK-STAT pathway in morphogenesis: From organ selector to cell behavior regulator. JAKSTAT 2: e26089. PubMed ID: 24069568

    Sotillos, S., Krahn, M., Espinosa-Vazquez, J. M. and Hombria, J. C. (2013). Src kinases mediate the interaction of the apical determinant Bazooka/PAR3 with STAT92E and increase signalling efficiency in Drosophila ectodermal cells. Development 140: 1507-1516. PubMed ID: 23462467

    Letizia, A., Sotillos, S., Campuzano, S. and Llimargas, M. (2011). Regulated Crb accumulation controls apical constriction and invagination in Drosophila tracheal cells. J Cell Sci 124: 240-251. PubMed ID: 21172808

  • Rita Sousa-Nunes Centre for Developmental Neurobiology, Kings College London

    Maierbrugger, K. T., Sousa-Nunes, R. and Bateman, J. M. (2020). The mTOR pathway component Unkempt regulates neural stem cell and neural progenitor cell cycle in the Drosophila central nervous system. Dev Biol. PubMed ID: 31978396

    Shaw, R. E., Kottler, B., Ludlow, Z. N., Buhl, E., Kim, D., Morais da Silva, S., Miedzik, A., Coum, A., Hodge, J. J., Hirth, F. and Sousa-Nunes, R. (2018). In vivo expansion of functionally integrated GABAergic interneurons by targeted increase in neural progenitors. Embo j. Pubmed ID: 29728368

    Sousa-Nunes, R. and Somers, W. G. (2013). Mechanisms of asymmetric progenitor divisions in the Drosophila central nervous system. Adv Exp Med Biol 786: 79-102. PubMed ID: 23696353

    Sousa-Nunes, R., Yee, L. L. and Gould, A. P. (2011). Fat cells reactivate quiescent neuroblasts via TOR and glial insulin relays in Drosophila. Nature 471(7339): 508-512. PubMed ID: 21346761


  • Tony Southall Faculty of Natural Sciences, Department of Life Sciences, Imperial College London
    McClure, C. D., Hassan, A., Aughey, G. N., Butt, K., Estacio-Gomez, A., Duggal, A., Ying Sia, C., Barber, A. F. and Southall, T. D. (2022). An auxin-inducible, GAL4-compatible, gene expression system for Drosophila. Elife 11. PubMed ID: 35363137

    Janssens, J., Aibar, S., Taskiran, II, Ismail, J. N., Gomez, A. E., Aughey, G., Spanier, K. I., De Rop, F. V., Gonzalez-Blas, C. B., Dionne, M., Grimes, K., Quan, X. J., Papasokrati, D., Hulselmans, G., Makhzami, S., De Waegeneer, M., Christiaens, V., Southall, T. and Aerts, S. (2022). Decoding gene regulation in the fly brain. Nature 601(7894): 630-636. PubMed ID: 34987221

    Perochon, J., Yu, Y., Aughey, G. N., Medina, A. B., Southall, T. D. and Cordero, J. B. (2021). Dynamic adult tracheal plasticity drives stem cell adaptation to changes in intestinal homeostasis in Drosophila. Nat Cell Biol 23(5): 485-496. PubMed ID: 33972729

    Estacio-Gómez, A., Hassan, A., Walmsley, E., Le, L. W. and Southall, T. D. (2020). Dynamic neurotransmitter specific transcription factor expression profiles during Drosophila development. Biol Open 9(5). PubMed ID: 32493733

    Hassan, A., Araguas Rodriguez, P., Heidmann, S. K., Walmsley, E. L., Aughey, G. N. and Southall, T. D. (2020). Condensin I subunit Cap-G is essential for proper gene expression during the maturation of post-mitotic neurons. Elife 9. PubMed ID: 32255428

    Mundorf, J., Donohoe, C. D., McClure, C. D., Southall, T. D. and Uhlirova, M. (2019). Ets21c Governs Tissue Renewal, Stress Tolerance, and Aging in the Drosophila Intestine. Cell Rep 27(10): 3019-3033.e3015. PubMed ID: 31167145

    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

    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

    Aughey, G. N., Estacio Gomez, A., Thomson, J., Yin, H. and Southall, T. D. (2018). CATaDa reveals global remodelling of chromatin accessibility during stem cell differentiation in vivo. Elife 7. PubMed ID: 29481322

    Cattenoz, P. B., Popkova, A., Southall, T. D., Aiello, G., Brand, A. H. and Giangrande, A. (2016). Functional Conservation of the Glide/Gcm Regulatory Network Controlling Glia, Hemocyte, and Tendon Cell Differentiation in Drosophila. Genetics 202(1): 191-219. PubMed ID: 26567182

    Loza-Coll, M. A., Southall, T. D., Sandall, S. L., Brand, A. H. and Jones, D. L. (2014). Regulation of Drosophila intestinal stem cell maintenance and differentiation by the transcription factor Escargot. EMBO J 33(24): 2983-2996. PubMed ID: 25433031

  • John Sparrow Department of Biology, University of York
    Katzemich, A., West, R. J., Fukuzawa, A., Sweeney, S. T., Gautel, M., Sparrow, J. and Bullard, B. (2015). Binding partners of the kinase domains in Drosophila obscurin and their effect on the structure of the flight muscle. J Cell Sci. PubMed ID: 26251439

    Orfanos, Z., Leonard, K., Elliott, C., Katzemich, A., Bullard, B. and Sparrow, J. (2015). Sallimus and the Dynamics of Sarcomere Assembly in Drosophila Flight Muscles. J Mol Biol. PubMed ID: 25868382

    Molnar, I., Migh, E., Szikora, S., Kalmar, T., Vegh, A. G., Deak, F., Barko, S., Bugyi, B., Orfanos, Z., Kovacs, J., Juhasz, G., Varo, G., Nyitrai, M., Sparrow, J. and Mihaly, J. (2014). DAAM is required for thin filament formation and Sarcomerogenesis during muscle development in Drosophila. PLoS Genet 10: e1004166. PubMed ID: 24586196

    Orfanos, Z. and Sparrow, J. C. (2013). Myosin isoform switching during assembly of the Drosophila flight muscle thick filament lattice. J Cell Sci 126: 139-148. PubMed ID: 23178940

  • Maria Spletter Department Physiology Chemistry, Biomedical Center, LMU Munich
    Nikonova, E., Mukherjee, A., Kamble, K., Barz, C., Nongthomba, U. and Spletter, M. L. (2022). Rbfox1 is required for myofibril development and maintaining fiber type-specific isoform expression in Drosophila muscles. Life Sci Alliance 5(4). PubMed ID: 34996845

    Kao, S. Y., Nikonova, E., Chaabane, S., Sabani, A., Martitz, A., Wittner, A., Heemken, J., Straub, T. and Spletter, M. L. (2021). A Candidate RNAi Screen Reveals Diverse RNA-Binding Protein Phenotypes in Drosophila Flight Muscle. Cells 10(10). PubMed ID: 34685485

    Nikonova, E., Kao, S. Y. and Spletter, M. L. (2020). Contributions of alternative splicing to muscle type development and function. Semin Cell Dev Biol 104: 65-80. PubMed ID: 32070639

    Spletter, M. L., Barz, C., Yeroslaviz, A., Zhang, X., Lemke, S. B., Bonnard, A., Brunner, E., Cardone, G., Basler, K., Habermann, B. H. and Schnorrer, F. (2018). A transcriptomics resource reveals a transcriptional transition during ordered sarcomere morphogenesis in flight muscle. Elife 7. PubMed ID: 29846170

  • Alexander Spirov Computer Science Department, Stony Brook University
    Shlemov, A., Alexandrov, T., Golyandina, N., Holloway, D., Baumgartner, S. and Spirov, A. V. (2021). Quantification reveals early dynamics in Drosophila maternal gradients. PLoS One 16(8): e0244701. PubMed ID: 34411119

    Myasnikova, E. and Spirov, A. (2020). Gene regulatory networks in Drosophila early embryonic development as a model for the study of the temporal identity of neuroblasts. Biosystems: 104192. PubMed ID: 32619531

    Myasnikova, E. and Spirov, A. (2018). Robustness of expression pattern formation due to dynamic equilibrium in gap gene system of an early Drosophila embryo. Biosystems. PubMed ID: 29428618

    Cai, X., Akber, M., Spirov, A. and Baumgartner, S. (2017). Cortical movement of Bicoid in early Drosophila embryos is actin- and microtubule-dependent and disagrees with the SDD diffusion model. PLoS One 12(10): e0185443. PubMed ID: 28973031

    Holloway, D. M. and Spirov, A. V. (2017). Transcriptional bursting in Drosophila development: Stochastic dynamics of eve stripe 2 expression. PLoS One 12(4): e0176228. PubMed ID: 28437444

    Spirov, A. V., Myasnikova, E. M. and Holloway, D. M. (2016). Sequential construction of a model for modular gene expression control, applied to spatial patterning of the Drosophila gene hunchback. J Bioinform Comput Biol 14: 1641005. PubMed ID: 27122317

    Shlemov, A., Golyandina, N., Holloway, D. and Spirov, A. (2015). Shaped singular spectrum analysis for quantifying gene expression, with application to the early Drosophila embryo. Biomed Res Int 2015: 689745. PubMed ID: 25945341

    Holloway, D.M. and Spirov, A.V. (2015). Mid-embryo patterning and precision in Drosophila segmentation: Krüppel dual regulation of hunchback. PLoS One 10: e0118450. PubMed ID: 25793381

    Holloway, D. M. and Spirov, A. V. (2015). Mid-embryo patterning and precision in Drosophila segmentation: Kruppel dual regulation of hunchback. PLoS One 10: e0118450. PubMed ID: 25793381

    Zagrijchuk, E. A., Sabirov, M. A., Holloway, D. M. and Spirov, A. V. (2014). In silico evolution of the hunchback gene indicates redundancy in cis-regulatory organization and spatial gene expression. J Bioinform Comput Biol 12: 1441009. PubMed ID: 24712536

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

    Flanagan, K., Baradaran-Heravi, A., Yin, Q., Dao Duc, K., Spradling, A. C. and Greenblatt, E. J. (2022). FMRP-dependent production of large dosage-sensitive proteins is highly conserved. Genetics. PubMed ID: 35731217

    DeLuca, S. Z., Ghildiyal, M., Pang, L. Y. and Spradling, A. C. (2020). Differentiating Drosophila female germ cells initiate Polycomb silencing by regulating PRC2-interacting proteins. Elife 9. PubMed ID: 32773039

    Wang, C. and Spradling, A. C. (2020). An abundant quiescent stem cell population in Drosophila Malpighian tubules protects principal cells from kidney stones. Elife 9. PubMed ID: 32175841

    Greenblatt, E. J., Obniski, R., Mical, C. and Spradling, A. C. (2019). Prolonged ovarian storage of mature Drosophila oocytes dramatically increases meiotic spindle instability. Elife 8. PubMed ID: 31755866

    Greenblatt, E. J. and Spradling, A. C. (2018). Fragile X mental retardation 1 gene enhances the translation of large autism-related proteins. Science 361(6403): 709-712. PubMed ID: 30115809

    DeLuca, S. Z. and Spradling, A. C. (2018). Efficient Expression of Genes in the Drosophila Germline Using a UAS-Promoter Free of Interference by Hsp70 piRNAs. Genetics [Epub ahead of print]. PubMed ID: 29669732

    Sieber, M. H., Thomsen, M. B. and Spradling, A. C. (2016). Electron transport chain remodeling by GSK3 during oogenesis connects nutrient state to reproduction. Cell 164: 420-432. PubMed ID: 26824655

    Nagarkar-Jaiswal, S., DeLuca, S. Z., Lee, P. T., Lin, W. W., Pan, H., Zuo, Z., Lv, J., Spradling, A. C. and Bellen, H. J. (2015). A genetic toolkit for tagging intronic MiMIC containing genes. Elife 4. PubMed ID: 26102525

    Nagarkar-Jaiswal, S., Lee, P. T., Campbell, M. E., Chen, K., Anguiano-Zarate, S., Cantu Gutierrez, M., Busby, T., Lin, W. W., He, Y., Schulze, K. L., Booth, B. W., Evans-Holm, M., Venken, K. J., Levis, R. W., Spradling, A. C., Hoskins, R. A. and Bellen, H. J. (2015). A library of MiMICs allows tagging of genes and reversible, spatial and temporal knockdown of proteins in Drosophila. Elife 4. PubMed ID: 25824290

    Sieber, M. H. and Spradling, A. C. (2015). Steroid signaling establishes a female metabolic state and regulates SREBP to control oocyte lipid accumulation. Curr Biol [Epub ahead of print]. PubMed ID: 25802149

    Deady, L.D., Shen, W., Mosure, S.A., Spradling, A.C. and Sun, J. (2015). Matrix Metalloproteinase 2 is required for ovulation and corpus luteum formation in Drosophila. PLoS Genet. 11: e1004989. PubMed ID: 25695427

  • Simon Sprecher Department of Biology, University of Fribourg, Switzerland
    Maier, G. L., Komarov, N., Meyenhofer, F., Kwon, J. Y. and Sprecher, S. G. (2021). Taste sensing and sugar detection mechanisms in Drosophila larval primary taste center. Elife 10. PubMed ID: 34859782

    Kaldun, J. C., Lone, S. R., Humbert Camps, A. M., Fritsch, C., Widmer, Y. F., Stein, J. V., Tomchik, S. M. and Sprecher, S. G. (2021). Dopamine, sleep, and neuronal excitability modulate amyloid-beta-mediated forgetting in Drosophila. PLoS Biol 19(10): e3001412. PubMed ID: 34613972

    Mishra, A. K., Fritsch, C., Voutev, R., Mann, R. S. and Sprecher, S. G. (2021). Homothorax controls a binary Rhodopsin switch in Drosophila ocelli. PLoS Genet 17(7): e1009460. PubMed ID: 34314427

    Zhao, C., Widmer, Y. F., Diegelmann, S., Petrovici, M. A., Sprecher, S. G. and Senn, W. (2021). Predictive olfactory learning in Drosophila. Sci Rep 11(1): 6795. PubMed ID: 33762640

    Asirim, E. Z., Humberg, T. H., Maier, G. L. and Sprecher, S. G. (2020). Circadian and Genetic Modulation of Visually-Guided Navigation in Drosophila Larvae. Sci Rep 10(1): 2752. PubMed ID: 32066794

    Brunet Avalos, C., Bruggmann, R. and Sprecher, S. G. (2019). Single cell transcriptome atlas of the Drosophila larval brain. Elife 8. PubMed ID: 31746739

    Qin, B., Humberg, T. H., Kim, A., Kim, H. S., Short, J., Diao, F., White, B. H., Sprecher, S. G. and Yuan, Q. (2019). Muscarinic acetylcholine receptor signaling generates OFF selectivity in a simple visual circuit. Nat Commun 10(1): 4093. PubMed ID: 31501438

    Fritsch, C., Bernardo-Garcia, F. J., Humberg, T. H., Mishra, A. K., Miellet, S., Almeida, S., Frochaux, M. V., Deplancke, B., Huber, A. and Sprecher, S. G. (2019). Multilevel regulation of the glass locus during Drosophila eye development. PLoS Genet 15(7): e1008269. PubMed ID: 31299050

    Bernardo-Garcia, F. J., Syed, M., Jekely, G. and Sprecher, S. G. (2019). Glass confers rhabdomeric photoreceptor identity in Drosophila, but not across all metazoans. Evodevo 10: 4. PubMed ID: 30873275

    Humberg, T. H. and Sprecher, S. G. (2018). Two pairs of Drosophila central brain neurons mediate larval navigational strategies based on temporal light information processing. Front Behav Neurosci 12: 305. PubMed ID: 30568583

    de Andres-Bragado, L., Mazza, C., Senn, W. and Sprecher, S. G. (2018). Statistical modelling of navigational decisions based on intensity versus directionality in Drosophila larval phototaxis. Sci Rep 8(1): 11272. PubMed ID: 30050066

  • Saripella Srikrishna Department of Biochemistry, Banaras Hindu University, Vranasi, India
    Yadav, A. K. and Srikrishna, S. (2018). scribble (scrib) knockdown induces tumorigenesis by modulating Drp1-Parkin mediated mitochondrial dynamics in the wing imaginal tissues of Drosophila. Mitochondrion. PubMed ID: 29360576

    Singh, S. K., Srivastav, S., Yadav, A. K. and Srikrishna, S. (2017). Knockdown of APPL mimics transgenic Abeta induced neurodegenerative phenotypes in Drosophila. Neurosci Lett 648: 8-13. PubMed ID: 28336338

    Srivastav, S., Singh, S. K., Yadav, A. K. and Srikrishna, S. (2015). Folic acid supplementation rescues anomalies associated with knockdown of parkin in dopaminergic and serotonergic neurons in Drosophila model of Parkinson's disease. Biochem Biophys Res Commun 460(3): 780-785. PubMed ID: 25824034

  • Ralph Stanewsky University of Muenster
    Eick, A. K., Ogueta, M., Buhl, E., Hodge, J. J. L. and Stanewsky, R. (2022). The opposing chloride cotransporters KCC and NKCC control locomotor activity in constant light and during long days. Curr Biol 32(6): 1420-1428.e1424. PubMed ID: 35303416

    Klemz, S., Wallach, T., Korge, S., Rosing, M., Klemz, R., Maier, B., Fiorenza, N. C., Kaymak, I., Fritzsche, A. K., Herzog, E. D., Stanewsky, R. and Kramer, A. (2021). Protein phosphatase 4 controls circadian clock dynamics by modulating CLOCK/BMAL1 activity. Genes Dev 35(15-16): 1161-1174. PubMed ID: 34301769

    George, R. and Stanewsky, R. (2021). Peripheral Sensory Organs Contribute to Temperature Synchronization of the Circadian Clock in Drosophila melanogaster. Front Physiol 12: 622545. PubMed ID: 33603678

    Versteven, M., Ernst, K. M. and Stanewsky, R. (2020). A Robust and Self-Sustained Peripheral Circadian Oscillator Reveals Differences in Temperature Compensation Properties with Central Brain Clocks. iScience 23(8): 101388. PubMed ID: 32798967

    Ogueta, M., Hardie, R. C. and Stanewsky, R. (2020). Light Sampling via Throttled Visual Phototransduction Robustly Synchronizes the Drosophila Circadian Clock. Curr Biol. PubMed ID: 32502413

    Singh, S., Giesecke, A., Damulewicz, M., Fexova, S., Mazzotta, G. M., Stanewsky, R. and Dolezel, D. (2019). New Drosophila circadian clock mutants affecting temperature compensation induced by targeted mutagenesis of timeless. Front Physiol 10: 1442. PubMed ID: 31849700

    Roessingh, S., Rosing, M., Marunova, M., Ogueta, M., George, R., Lamaze, A. and Stanewsky, R. (2019). Temperature synchronization of the Drosophila circadian clock protein PERIOD is controlled by the TRPA channel PYREXIA. Commun Biol 2: 246. PubMed ID: 31286063

    Ogueta, M., Hardie, R. C. and Stanewsky, R. (2018). Non-canonical Phototransduction Mediates Synchronization of the Drosophila melanogaster Circadian Clock and Retinal Light Responses. Curr Biol. Pubmed ID: 29779871

    Chen, C., Xu, M., Anantaprakorn, Y., Rosing, M. and Stanewsky, R. (2018). nocte is required for integrating light and temperature inputs in circadian clock neurons of Drosophila. Curr Biol 28(10): 1595-1605.e1593. PubMed ID: 29754901

    Tang, X., Roessingh, S., Hayley, S. E., Chu, M. L., Tanaka, N. K., Wolfgang, W., Song, S., Stanewsky, R. and Hamada, F. N. (2017). The role of PDF neurons in setting preferred temperature before dawn in Drosophila. Elife 6. PubMed ID: 28463109

  • Alexander Stark Research Institute of Molecular Pathology, Vienna
    Haberle, V., Arnold, C. D., Pagani, M., Rath, M., Schernhuber, K. and Stark, A. (2019). Transcriptional cofactors display specificity for distinct types of core promoters. Nature. PubMed ID: 31092928

    Franz, A., Shlyueva, D., Brunner, E., Stark, A. and Basler, K. (2017). Probing the canonicity of the Wnt/Wingless signaling pathway. PLoS Genet 13(4): e1006700. PubMed ID: 28369070

    Arnold, C. D., Zabidi, M. A., Pagani, M., Rath, M., Schernhuber, K., Kazmar, T. and Stark, A. (2016). Genome-wide assessment of sequence-intrinsic enhancer responsiveness at single-base-pair resolution. Nat Biotechnol. PubMed ID: 28024147

    Stampfel, G., Kazmar, T., Frank, O., Wienerroither, S., Reiter, F. and Stark, A. (2015). Transcriptional regulators form diverse groups with context-dependent regulatory functions. Nature. PubMed ID: 26550828

    Spletter, M.L., Barz, C., Yeroslaviz, A., Schönbauer, C., Ferreira, I.R., Sarov, M., Gerlach, D., Stark, A., Habermann, B.H. and Schnorrer, F. (2014). The RNA-binding protein Arrest (Bruno) regulates alternative splicing to enable myofibril maturation in Drosophila flight muscle. EMBO Rep. [Epub ahead of print]. PubMed ID: 25532219

    Zabidi, M. A., Arnold, C. D., Schernhuber, K., Pagani, M., Rath, M., Frank, O. and Stark, A. (2014). Enhancer--core-promoter specificity separates developmental and housekeeping gene regulation. Nature. PubMed ID: 25517091

    Yanez-Cuna, J. O., Arnold, C. D., Stampfel, G., Boryn, L. M., Gerlach, D., Rath, M. and Stark, A. (2014). Dissection of thousands of cell type-specific enhancers identifies dinucleotide repeat motifs as general enhancer features. Genome Res. PubMed ID: 24714811

    Shlyueva, D., Stelzer, C., Gerlach, D., Yanez-Cuna, J. O., Rath, M., Boryn, L. M., Arnold, C. D. and Stark, A. (2014). Hormone-Responsive Enhancer-Activity Maps Reveal Predictive Motifs, Indirect Repression, and Targeting of Closed Chromatin. Mol Cell. PubMed ID: 24685159

    Meireles-Filho, A. C., Bardet, A. F., Yanez-Cuna, J. O., Stampfel, G. and Stark, A. (2013). cis-Regulatory Requirements for Tissue-Specific Programs of the Circadian Clock. Curr Biol. PubMed ID: 24332542

    Bardet, A. F., Steinmann, J., Bafna, S., Knoblich, J. A., Zeitlinger, J. and Stark, A. (2013). Identification of transcription factor binding sites from ChIP-seq data at high-resolution. Bioinformatics. PubMed ID: 23980024

    Arnold, C. D., Gerlach, D., Stelzer, C., Boryn, L. M., Rath, M., Stark, A. (2013) Genome-Wide Quantitative Enhancer Activity Maps Identified by STARR-seq. Science. PubMed ID: 23328393

    Yanez-Cuna, J. O., Dinh, H. Q., Kvon, E. Z., Shlyueva, D. and Stark, A. (2012). Uncovering cis-regulatory sequence requirements for context-specific transcription factor binding. Genome Res 22: 2018-2030. PubMed ID: 22534400

  • Michelle Starz-Gaiano UMBC, Baltimore, MD
    Berez, A., Peercy, B. E. and Starz-Gaiano, M. (2020). Development and Analysis of a Quantitative Mathematical Model of Bistability in the Cross Repression System Between APT and SLBO Within the JAK/STAT Signaling Pathway. Front Physiol 11: 803. PubMed ID: 32848815

    Saadin, A. and Starz-Gaiano, M. (2018). Cytokine exocytosis and JAK/STAT activation in the Drosophila ovary requires the vesicle trafficking regulator alpha-Snap. J Cell Sci. PubMed ID: 30404830

    Manning, L., Sheth, J., Bridges, S., Saadin, A., Odinammadu, K., Andrew, D., Spencer, S., Montell, D. and Starz-Gaiano, M. (2017). A hormonal cue promotes timely follicle cell migration by modulating transcription profiles. Mech Dev. PubMed ID: 28610887

    Saadin, A. and Starz-Gaiano, M. (2016). Identification of novel regulators of the JAK/STAT signaling pathway that control morder cell migration in the Drosophila ovary. G3 (Bethesda). PubMed ID: 27175018

    Monahan, A.J. and Starz-Gaiano, M. (2016). Apontic regulates somatic stem cell numbers in Drosophila testes. BMC Dev Biol 16: 5. PubMed ID: 26993259

    Monahan, A. J. and Starz-Gaiano, M. (2015). Socs36E limits STAT signaling via Cullin2 and a SOCS-box independent mechanism in the Drosophila egg chamber. Mech Dev. PubMed ID: 26277564

    Manning, L.A.1, Weideman, A.M., Peercy, B.E. and Starz-Gaiano, M. (2015). Tissue landscape alters adjacent cell fates during Drosophila egg development. Nat Commun 6: 7356. PubMed ID: 26082073

    Horn, L., Leips, J. and Starz-Gaiano, M. (2014). Phagocytic ability declines with age in adult Drosophila hemocytes. Aging Cell. PubMed ID: 24828474

    Monahan, A. J., Starz-Gaiano, M. (2013) Socs36E Attenuates STAT Signaling to Optimize Motile Cell Specification in the Drosophila Ovary. Dev Biol. PubMed ID: 23583584

    Montell, D. J., Yoon, W. H. and Starz-Gaiano, M. (2012). Group choreography: mechanisms orchestrating the collective movement of border cells. Nat Rev Mol Cell Biol 13: 631-645. PubMed ID: 23000794

  • Angelika Stathopoulos California Institute of Technology
    Dunipace, L., Newcomb, S. and Stathopoulos, A. (2022). brinker levels regulated by a promoter proximal element support germ cell homeostasis. Development. PubMed ID: 35037688

    Stepanik, V., Sun, J. and Stathopoulos, A. (2020). FGF Pyramus Has a Transmembrane Domain and Cell-Autonomous Function in Polarity. Curr Biol. PubMed ID: 32619487

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

    Sun, J., Macabenta, F., Akos, Z. and Stathopoulos, A. (2020). Collective Migrations of Drosophila Embryonic Trunk and Caudal Mesoderm-Derived Muscle Precursor Cells. Genetics 215(2): 297-322. PubMed ID: 32487692

    Irizarry, J., McGehee, J., Kim, G., Stein, D. and Stathopoulos, A. (2020). Twist-dependent ratchet functioning downstream from Dorsal revealed using a light-inducible degron. Genes Dev. PubMed ID: 32467225

    Dunipace, L., Akos, Z. and Stathopoulos, A. (2019). Coacting enhancers can have complementary functions within gene regulatory networks and promote canalization. PLoS Genet 15(12): e1008525. PubMed ID: 31830033

    Koromila, T. and Stathopoulos, A. (2019). Distinct roles of broadly expressed repressors support dynamic enhancer action and change in time. Cell Rep 28(4): 855-863. PubMed ID: 31340149

    Macabenta, F. and Stathopoulos, A. (2019). Migrating cells control morphogenesis of substratum serving as track to promote directional movement of the collective. Development. PubMed ID: 31239242

    Sandler, J. E., Irizarry, J., Stepanik, V., Dunipace, L., Amrhein, H. and Stathopoulos, A. (2018). A developmental program truncates long transcripts to temporally regulate cell signaling. Dev Cell 47(6): 773-784.e776. PubMed ID: 30562515

    Sun, J. and Stathopoulos, A. (2018). FGF controls epithelial-mesenchymal transitions during gastrulation by regulating cell division and apicobasal polarity. Development. PubMed ID: 30190277

    Koromila, T. and Stathopoulos, A. (2017). Broadly expressed repressors integrate patterning across orthogonal axes in embryos. Proc Natl Acad Sci U S A. PubMed ID: 28720706

    Stepanik, V., Dunipace, L., Bae, Y.K., Macabenta, F., Sun, J., Trisnadi, N. and Stathopoulos, A. (2016). The migrations of Drosophila muscle founders and primordial germ cells are interdependent. Development 143: 3206-3215. PubMed ID: 27578182

    Sandler, J. E. and Stathopoulos, A. (2016). Quantitative single-embryo profile of Drosophila genome activation and the dorsal-ventral patterning network. Genetics [Epub ahead of print]. PubMed ID: 26896327

    Irizarry, J. and Stathopoulos, A. (2015). FGF signaling supports Drosophila fertility by regulating development of ovarian muscle tissues. Dev Biol [Epub ahead of print]. PubMed ID: 259580900

  • Brian Staveley Molecular Cell Biology and Developmental Genetics Laboratory Department of Biology, Memorial University of Newfoundland, St. John's
    Hurley, E. P. and Staveley, B. E. (2021). Inhibition of Ref(2)P, the Drosophila homologue of the p62/SQSTM1 gene, increases lifespan and leads to a decline in motor function. BMC Res Notes 14(1): 53. PubMed ID: 33557921

    M'Angale, P. G. and Staveley, B. E. (2017). A loss of Pdxk model of Parkinson disease in Drosophila can be suppressed by Buffy. BMC Res Notes 10(1): 205. PubMed ID: 28606139

    M'Angale, P. G. and Staveley, B. E. (2017). Bax-inhibitor-1 knockdown phenotypes are suppressed by Buffy and exacerbate degeneration in a Drosophila model of Parkinson disease. PeerJ 5: e2974. PubMed ID: 28243526

    M'Angale, P. G. and Staveley, B. E. (2016). Overexpression of Buffy enhances the loss of parkin and suppresses the loss of Pink1 phenotypes in Drosophila. Genome: 1-7. PubMed ID: 28106473

    Merzetti, E. M., Dolomount, L. A. and Staveley, B. E. (2016). The FBXO7 homologue nutcracker and binding partner PI31 in Drosophila melanogaster models of Parkinson's disease. Genome: 1-9. PubMed ID: 27936908

    M'Angale, P. G. and Staveley, B. E. (2016). Knockdown of the putative Lifeguard homologue CG3814 in neurons of Drosophila melanogaster. Genet Mol Res 15(4). PubMed ID: 28002605

    Merzetti, E.M., Dolomount, L.A. and Staveley, B.E. (2016). The FBXO7 homologue nutcracker and binding partner PI31 in Drosophila melanogaster models of Parkinson's disease. Genome [Epub ahead of print]. PubMed ID: 27936908

    M'Angale, P. G. and Staveley, B. E. (2016). Loss of porin function in dopaminergic neurons of Drosophila is suppressed by Buffy. J Biomed Sci 23(1): 84. PubMed ID: 27881168

    M'Angale, P. G. and Staveley, B. E. (2016). The HtrA2 Drosophila model of Parkinson's disease is suppressed by the pro-survival Bcl-2 Buffy. Genome: 1-7. PubMed ID: 27848260

    M'Angale, P.G. and Staveley, B.E. (2016). Bcl-2 homologue Debcl enhances α-synuclein-induced phenotypes in Drosophila. PeerJ 4: e2461. PubMed ID: 27672511

    M'Angale, P. G. and Staveley, B. E. (2016). The Bcl-2 homologue Buffy rescues alpha-synuclein-induced Parkinson disease-like phenotypes in Drosophila. BMC Neurosci 17: 24. PubMed ID: 27192974

  • David Stein Cell and Molecular Biology University of Texas, Austin
    Amarnath, S., Stevens, L. M. and Stein, D. S. (2017). Reconstitution of Torso signaling in cultured cells suggests a role for both Trunk and Torsolike in receptor activation. Development [Epub ahead of print]. PubMed ID: 28087630

    Stein, D. S. and Stevens, L. M. (2014). Maternal control of the Drosophila dorsal-ventral body axis. Wiley Interdiscip Rev Dev Biol 3: 301-330. PubMed ID: 25124754

    Stein, D., Cho, Y. S. and Stevens, L. M. (2013). Localized serine protease activity and the establishment of Drosophila embryonic dorsoventral polarity. Fly (Austin) 7. PubMed ID: 24047959

    Cho, Y. S., Stevens, L. M., Sievermann, K. J, Nguyen, J., and Stein, D. (2012). A ventrally localized protease in the Drosophila egg controls embryo dorsoventral polarity, Curr. Biol. 22: 1013-1018. PubMed ID: 22578419

  • Mindy Steiniger Biology Department, University of Missouri, St. Louis,
    Harrington, A. W., McKain, M. R., Michalski, D., Bauer, K. M., Daugherty, J. M. and Steiniger, M. (2017). Drosophila melanogaster retrotransposon and inverted repeat-derived endogenous siRNAs are differentially processed in distinct cellular locations. BMC Genomics 18(1): 304. PubMed ID: 28415970

    Harrington, A. W. and Steiniger, M. (2016). Bioinformatic analyses of sense and antisense expression from terminal inverted repeat transposons in Drosophila somatic cells. Fly (Austin) 10(1): 1-10. PubMed ID: 26986720

    Russo, J., Harrington, A. W. and Steiniger, M. (2016). Antisense Transcription of Retrotransposons in Drosophila: An Origin of Endogenous Small Interfering RNA Precursors. Genetics 202(1): 107-121. PubMed ID: 26534950

  • Hermann Steller Strang Laboratory of Apoptosis and Cancer Biology, Rockefeller University
    Domingos, P. M., Jenny, A., Combie, K. F., Del Alamo, D., Mlodzik, M., Steller, H. and Mollereau, B. (2019). Regulation of Numb during planar cell polarity establishment in the Drosophila eye. Mech Dev: 103583. PubMed ID: 31678471

    Liu, K., Jones, S., Minis, A., Rodriguez, J., Molina, H. and Steller, H. (2019). PI31 is an adaptor protein for proteasome transport in axons and required for synaptic development. Dev Cell. PubMed ID: 31327739

    Gultekin, Y. and Steller, H. (2019). Axin proteolysis by Iduna is required for the regulation of stem cell proliferation and intestinal homeostasis in Drosophila. Development 146(6). PubMed ID: 30796047

    Gerstberger, S., Meyer, C., Benjamin-Hong, S., Rodriguez, J., Briskin, D., Bognanni, C., Bogardus, K., Steller, H. and Tuschl, T. (2017). The conserved RNA exonuclease Rexo5 is required for 3' end maturation of 28S rRNA, 5S rRNA, and snoRNAs. Cell Rep 21(3): 758-772. PubMed ID: 29045842

    Maor, G., Cabasso, O., Krivoruk, O., Rodriguez, J., Steller, H., Segal, D. and Horowitz, M. (2016). The contribution of mutant GBA to the development of parkinson disease in Drosophila. Hum Mol Genet. PubMed ID: 27162249

    Rosas-Arellano, A., Vasquez-Procopio, J., Gambis, A., Blowes, L. M., Steller, H., Mollereau, B. and Missirlis, F. (2016). Ferritin Assembly in Enterocytes of Drosophila melanogaster. Int J Mol Sci 17 [Epub ahead of print]. PubMed ID: 26861293

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

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

    Guarner, A., Manjon, C., Edwards, K., Steller, H., Suzanne, M. and Sanchez-Herrero, E. (2013). The zinc finger homeodomain-2 gene of Drosophila controls Notch targets and regulates apoptosis in the tarsal segments. Dev Biol. PubMed ID: 24144920

    Suzanne, M. and Steller, H. (2013). Shaping organisms with apoptosis. Cell Death Differ 20: 669-675. PubMed ID: 23449394

  • Per Stenberg Department of Molecular Biology, Umea University
    Lewerentz, J., Johansson, A. M., Larsson, J. and Stenberg, P. (2022). Transposon activity, local duplications and propagation of structural variants across haplotypes drive the evolution of the Drosophila S2 cell line. BMC Genomics 23(1): 276. PubMed ID: 35392795

    Zare, A., Johansson, A. M., Karlsson, E., Delhomme, N. and Stenberg, P. (2018). The gut microbiome participates in transgenerational inheritance of low-temperature responses in Drosophila melanogaster. FEBS Lett. PubMed ID: 30372516

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

    Philip, P., Boija, A., Vaid, R., Churcher, A. M., Meyers, D. J., Cole, P. A., Mannervik, M. and Stenberg, P. (2015). CBP binding outside of promoters and enhancers in Drosophila melanogaster. Epigenetics Chromatin 8: 48. PubMed ID: 26604986

    Philip, P. and Stenberg, P. (2013). Male X-linked genes in Drosophila melanogaster are compensated independently of the Male-Specific Lethal complex. Epigenetics Chromatin 6: 35. PubMed ID: 24279328

    Philip, P., Pettersson, F. and Stenberg, P. (2012). Sequence signatures involved in targeting the Male-Specific Lethal complex to X-chromosomal genes in Drosophila melanogaster. BMC Genomics 13: 97. PubMed ID: 22424303

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

    Holmqvist, P. H., Boija, A., Philip, P., Crona, F., Stenberg, P. and Mannervik, M. (2012). Preferential genome targeting of the CBP co-activator by Rel and Smad proteins in early Drosophila melanogaster embryos. PLoS Genet 8: e1002769. PubMed ID: 22737084

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

  • Harald Stenmark Dept. of Molecular Cell Biology, Oslo University Hospital
    Lie-Jensen, A., Ivanauskiene, K., Malerod, L., Jain, A., Tan, K. W., Laerdahl, J. K., Liestol, K., Stenmark, H. and Haglund, K. (2019). Centralspindlin Recruits ALIX to the Midbody during Cytokinetic Abscission in Drosophila via a Mechanism Analogous to Virus Budding. Curr Biol 29(20): 3538-3548 e3537. PubMed ID: 31607533

    Eikenes, A. H., Malerod, L., Lie-Jensen, A., Sem Wegner, C., Brech, A., Liestol, K., Stenmark, H. and Haglund, K. (2015). Src64 controls a novel actin network required for proper ring canal formation in the Drosophila male germline. Development 142(23): 4107-4118. PubMed ID: 26628094

    Morelli, E., Ginefra, P., Mastrodonato, V., Beznoussenko, G. V., Rusten, T. E., Bilder, D., Stenmark, H., Mironov, A. A. and Vaccari, T. (2014). Multiple functions of the SNARE protein Snap29 in autophagy, endocytic, and exocytic trafficking during epithelial formation in Drosophila. Autophagy 10(12): 2251-2268. PubMed ID: 25551675

  • Marcus Stensmyr Department of Biology, Lund University, Sweden
    Melo, N., Capek, M., Arenas, O. M., Afify, A., Yilmaz, A., Potter, C. J., Laminette, P. J., Para, A., Gallio, M. and Stensmyr, M. C. (2021). The irritant receptor TRPA1 mediates the mosquito repellent effect of catnip. Curr Biol. PubMed ID: 33667373

    Melo, N., Wolff, G. H., Costa-da-Silva, A. L., Arribas, R., Triana, M. F., Gugger, M., Riffell, J. A., DeGennaro, M. and Stensmyr, M. C. (2019). Geosmin attracts Aedes aegypti mosquitoes to oviposition sites. Curr Biol. PubMed ID: 31839454

    Mansourian, S., Enjin, A., Jirle, E. V., Ramesh, V., Rehermann, G., Becher, P. G., Pool, J. E. and Stensmyr, M. C. (2018). Wild african Drosophila melanogaster are seasonal specialists on marula fruit. Curr Biol. PubMed ID: 30528579

    Frank, D. D., Enjin, A., Jouandet, G. C., Zaharieva, E. E., Para, A., Stensmyr, M. C. and Gallio, M. (2017). Early integration of temperature and humidity stimuli in the Drosophila brain. Curr Biol 27(15): 2381-2388.e2384. PubMed ID: 28736172

    Mansourian, S., Corcoran, J., Enjin, A., Lofstedt, C., Dacke, M. and Stensmyr, M. C. (2016). Fecal-derived phenol induces egg-laying aversion in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 27641770

    Enjin, A., Zaharieva, E.E., Frank, D.D., Mansourian, S., Suh, G.S., Gallio, M. and Stensmyr, M.C. (2016). Humidity sensing in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 27161501

    Mansourian, S. and Stensmyr, M. C. (2015). The chemical ecology of the fly. Curr Opin Neurobiol 34C: 95-102. PubMed ID: 25747730

    Dweck, H.K., Ebrahim, S.A., Farhan, A., Hansson, B.S. and Stensmyr, M.C. (2015). Olfactory proxy detection of dietary antioxidants in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 25619769

    Dweck, H. K., Ebrahim, S. A., Kromann, S., Bown, D., Hillbur, Y., Sachse, S., Hansson, B. S. and Stensmyr, M. C. (2013). Olfactory Preference for Egg Laying on Citrus Substrates in Drosophila. Curr Biol. PubMed ID: 24316206

    Linz, J., Baschwitz, A., Strutz, A., Dweck, H. K., Sachse, S., Hansson, B. S. and Stensmyr, M. C. (2013). Host plant-driven sensory specialization in Drosophila erecta. Proc Biol Sci 280: 20130626. PubMed ID: 23595274

  • Wolfgang Stephan Biozentrum Martinsried, Munich, Germany
    Voigt, S., Erpf, A. C. and Stephan, W. (2019). Decreased temperature sensitivity of Vestigial gene expression in temperate populations of Drosophila melanogaster. Genes (Basel) 10(7). PubMed ID: 31261809

    Voigt, S., Laurent, S., Litovchenko, M. and Stephan, W. (2015). Positive Selection at the Polyhomeotic Locus Led to Decreased Thermosensitivity of Gene Expression in Temperate Drosophila melanogaster. Genetics. PubMed ID: 25855066

    Wilches, R., Voigt, S., Duchen, P., Laurent, S. and Stephan, W. (2014). Fine-mapping and selective sweep analysis of QTL for cold tolerance in Drosophila melanogaster. G3 (Bethesda) 4: 1635-1645. PubMed ID: 24970882

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

  • David Stern HHMI, Princeton University
    Duckhorn, J. C., Cande, J., Metkus, M. C., Song, H., Altamirano, S., Stern, D. L. and Shirangi, T. R. (2022). Regulation of Drosophila courtship behavior by the Tlx/tailless-like nuclear receptor, dissatisfaction. Curr Biol. PubMed ID: 35245457

    Soverna, A. F., Rodriguez, N. C., Korgaonkar, A., Hasson, E., Stern, D. L. and Frankel, N. (2020). Cis-regulatory variation in the shavenbaby gene underlies intraspecific phenotypic variation, mirroring interspecific divergence in the same trait. Evolution. PubMed ID: 33314059

    Fuqua, T., Jordan, J., van Breugel, M. E., Halavatyi, A., Tischer, C., Polidoro, P., Abe, N., Tsai, A., Mann, R. S., Stern, D. L. and Crocker, J. (2020). Dense and pleiotropic regulatory information in a developmental enhancer. Nature 587(7833): 235-239. PubMed ID: 33057197

    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

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

    Cande, J., Namiki, S., Qiu, J., Korff, W., Card, G. M., Shaevitz, J. W., Stern, D. L. and Berman, G. J. (2018). Optogenetic dissection of descending behavioral control in Drosophila. Elife 7. PubMed ID: 29943729

    Frankel, N. (2018). Comprehensive analysis of a cis-regulatory region reveals pleiotropy in enhancer function. Cell Rep 22(11): 3021-3031. PubMed ID: 29539428

    Tsai, A., Muthusamy, A. K., Alves, M. R., Lavis, L. D., Singer, R. H., Stern, D. L. and Crocker, J. (2017). Nuclear microenvironments modulate transcription from low-affinity enhancers. Elife 6. PubMed ID: 29095143

    Stern, D. L., Clemens, J., Coen, P., Calhoun, A. J., Hogenesch, J. B., Arthur, B. J. and Murthy, M. (2017). Experimental and statistical reevaluation provides no evidence for Drosophila courtship song rhythms. Proc Natl Acad Sci U S A 114(37): 9978-9983. PubMed ID: 28851830

    Crocker, J. and Stern, D. L. (2017). Functional regulatory evolution outside of the minimal even-skipped stripe 2 enhancer. Development. PubMed ID: 28760812

    Crocker, J., Tsai, A. and Stern, D. L. (2017). A fully synthetic transcriptional platform for a multicellular eukaryote. Cell Rep 18(1): 287-296. PubMed ID: 28052257

    Ding, Y., Berrocal, A., Morita, T., Longden, K. D. and Stern, D. L. (2016). Natural courtship song variation caused by an intronic retroelement in an ion channel gene. Nature 536: 329-332. PubMed ID: 27509856

    Shirangi, T. R., Wong, A. M., Truman, J. W. and Stern, D. L. (2016). Doublesex regulates the connectivity of a neural circuit controlling Drosophila male courtship song. Dev Cell 37: 533-544. PubMed ID: 27326931

  • Michael Stern Department of Biochemistry & Cell Biology, Rice University
    Aleman-Meza, B., Loeza-Cabrera, M., Pena-Ramos, O., Stern, M. and Zhong, W. (2017). High-content behavioral profiling reveals neuronal genetic network modulating Drosophila larval locomotor program. BMC Genet 18(1): 40. PubMed ID: 28499390

    Summerville, J., Faust, J., Fan, E., Pendin, D., Daga, A., Formella, J., Stern, M. and McNew, J. A. (2016). The effects of ER morphology on synaptic structure and function in Drosophila melanogaster. J Cell Sci. PubMed ID: 26906425

    Faust, J. E., Manisundaram, A., Ivanova, P. T., Milne, S. B., Summerville, J. B., Brown, H. A., Wangler, M., Stern, M. and McNew, J. A. (2014). Peroxisomes Are Required for Lipid Metabolism and Muscle Function in Drosophila melanogaster. PLoS One 9: e100213. PubMed ID: 24945818

    Johnson, C., Chun-Jen Lin, C. and Stern, M. (2012). Ras-dependent and Ras-independent effects of PI3K in Drosophila motor neurons. Genes Brain Behav 11: 848-858. PubMed ID: 22783951

    Chun-Jen Lin, C., Summerville, J. B., Howlett, E. and Stern, M. (2011). The metabotropic glutamate receptor activates the lipid kinase PI3K in Drosophila motor neurons through the calcium/calmodulin-dependent protein kinase II and the nonreceptor tyrosine protein kinase DFak. Genetics 188: 601-613. PubMed ID: 21515581

  • Ruth Steward Waksman Institute, Molecular Biology and Biochemistry, Rutgers University
    Karunendiran, A., Nguyen, C. T., Barzda, V. and Stewart, B. A. (2021). Disruption of Drosophila larval muscle structure and function by UNC45 knockdown. BMC Mol Cell Biol 22(1): 38. PubMed ID: 34256704

    Scepanovic, G. and Stewart, B. A. (2019). Analysis of Drosophila nervous system development following an early, brief exposure to ethanol. Dev Neurobiol. PubMed ID: 31472090

    Wang, F., Minakhina, S., Tran, H., Changela, N., Kramer, J. and Steward, R. (2018). Tet protein function during Drosophila development. PLoS One 13(1): e0190367. PubMed ID: 29324752

    Minakhina, S., Naryshkina, T., Changela, N., Tan, W. and Steward, R. (2016). Zfrp8/PDCD2 interacts with RpS2 connecting ribosome maturation and gene-specific translation. PLoS One 11: e0147631. PubMed ID: 26807849

    Delatte, B., et al. (2016). RNA biochemistry. Transcriptome-wide distribution and function of RNA hydroxymethylcytosine. Science 351: 282-285. PubMed ID: 26816380

    Tan, W., Schauder, C., Naryshkina, T., Minakhina, S. and Steward, R. (2016). Zfrp8 forms a complex with fragile-X mental retardation proztein and regulates its localization and function. Dev Biol. PubMed ID: 26772998

    Reilly, E., Changela, N., Naryshkina, T., Deshpande, G. and Steward, R. (2015). Discs large 5, an essential gene in Drosophila, regulates egg chamber organization. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 25795662

    Minakhina, S., Changela, N. and Steward, R. (2014). Zfrp8/PDCD2 is required in ovarian stem cells and interacts with the piRNA pathway machinery. Development 141: 259-268. PubMed ID: 24381196

    Sakaguchi, A., Joyce, E., Aoki, T., Schedl, P. and Steward, R. (2012). The histone H4 lysine 20 monomethyl mark, set by PR-Set7 and stabilized by L(3)mbt, is necessary for proper interphase chromatin organization. PLoS One 7: e45321. PubMed ID: 23024815

    Minakhina, S., Tan, W. and Steward, R. (2011). JAK/STAT and the GATA factor Pannier control hemocyte maturation and differentiation in Drosophila. Dev Biol 352: 308-316. PubMed ID: 21295568

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

    Nguyen, C. T. and Stewart, B. A. (2016). The influence of postsynaptic structure on missing quanta at the Drosophila neuromuscular junction. BMC Neurosci 17: 53. PubMed ID: 27459966

    Wang, S. J., Tsai, A., Wang, M., Yoo, S., Kim, H. Y., Yoo, B., Chui, V., Kisiel, M., Stewart, B., Parkhouse, W., Harden, N. and Krieger, C. (2014). Phospho-regulated Drosophila adducin is a determinant of synaptic plasticity in a complex with Dlg and PIP2 at the larval neuromuscular junction. Biol Open. PubMed ID: 25416060

    Kisiel, M., McKenzie, K. and Stewart, B. (2014). Localization and Mobility of Synaptic Vesicles in Myosin VI Mutants of Drosophila. PLoS One 9: e102988. PubMed ID: 25062032

    DeMill, C. M., Qiu, X., Kisiel, M., Bolotta, A. and Stewart, B. A. (2014). Investigation of the juxtamembrane region of neuronal-Synaptobrevin in synaptic transmission at the Drosophila neuromuscular junction. J Neurophysiol. PubMed ID: 24944220

    Kisiel, M., Majumdar, D., Campbell, S. and Stewart, B. A. (2011). Myosin VI contributes to synaptic transmission and development at the Drosophila neuromuscular junction. BMC Neurosci 12: 65. PubMed ID: 21745401

    Seabrooke, S. and Stewart, B. A. (2011). Synaptic transmission and plasticity are modulated by nonmuscle myosin II at the neuromuscular junction of Drosophila. J Neurophysiol 105: 1966-1976. PubMed ID: 21325687

  • Geoffrey Stilwell Biology Department, Rhode Island College
    Agudelo, A., St Amand, V., Grissom, L., Lafond, D., Achilli, T., Sahin, A., Reenan, R. and Stilwell, G. (2020). Age-dependent degeneration of an identified adult leg motor neuron in a Drosophila SOD1 model of ALS. Biol Open 9(10). PubMed ID: 32994185

    Sahin, A., Held, A., Bredvik, K., Major, P., Achilli, T. M., Kerson, A. G., Wharton, K., Stilwell, G. and Reenan, R. (2017). Human SOD1 ALS Mutations in a Drosophila Knock-In Model Cause Severe Phenotypes and Reveal Dosage-Sensitive Gain- and Loss-of-Function Components. Genetics 205(2): 707-723. PubMed ID: 27974499

    Watson, G. B., Chouinard, S. W., Cook, K. R., Geng, C., Gifford, J. M., Gustafson, G. D., Hasler, J. M., Larrinua, I. M., Letherer, T. J., Mitchell, J. C., Pak, W. L., Salgado, V. L., Sparks, T. C. and Stilwell, G. E. (2010). A spinosyn-sensitive Drosophila melanogaster nicotinic acetylcholine receptor identified through chemically induced target site resistance, resistance gene identification, and heterologous expression. Insect Biochem Mol Biol 40(5): 376-384. PubMed ID: 19944756

  • Daniel St Johnston Gurdon Institute, Cambridge
    Cheng, J., Allgeyer, E. S., Richens, J. H., Dzafic, E., Palandri, A., Lewkow, B., Sirinakis, G. and St Johnston, D. (2021). A single-molecule localization microscopy method for tissues reveals nonrandom nuclear pore distribution in Drosophila. J Cell Sci 134(24). PubMed ID: 34806753

    Doerflinger, H., Zimyanin, V. and St Johnston, D. (2022). The Drosophila anterior-posterior axis is polarized by asymmetric myosin activation. Curr Biol 32(2): 374-385.e374. PubMed ID: 34856125

    Fic, W., Bastock, R., Raimondi, F., Los, E., Inoue, Y., Gallop, J. L., Russell, R. B. and St Johnston, D. (2021). RhoGAP19D inhibits Cdc42 laterally to control epithelial cell shape and prevent invasion. J Cell Biol 220(4). PubMed ID: 33646271

    Lovegrove, H. E., Bergstralh, D. T. and St Johnston, D. (2019). The role of integrins in Drosophila egg chamber morphogenesis. Development 146(23). PubMed ID: 31784458

    Fic, W., Faria, C. and St Johnston, D. (2019). IMP regulates Kuzbanian to control the timing of Notch signalling in Drosophila follicle cells. Development 146(2). PubMed ID: 30635283

    Chen, J., Sayadian, A. C., Lowe, N., Lovegrove, H. E. and St Johnston, D. (2018). An alternative mode of epithelial polarity in the Drosophila midgut. PLoS Biol 16(10): e3000041. PubMed ID: 30339698

    Nieuwburg, R., Nashchekin, D., Jakobs, M., Carter, A. P., Khuc Trong, P., Goldstein, R. E. and St Johnston, D. (2017). Localised dynactin protects growing microtubules to deliver oskar mRNA to the posterior cortex of the Drosophila oocyte. Elife 6. PubMed ID: 29035202

    Trovisco, V., Belaya, K., Nashchekin, D., Irion, U., Sirinakis, G., Butler, R., Lee, J. J., Gavis, E. R. and St Johnston, D. (2016). bicoid mRNA localises to the Drosophila oocyte anterior by random Dynein-mediated transport and anchoring. Elife 5. PubMed ID: 27791980

    Nashchekin, D., Fernandes, A. R. and St Johnston, D. (2016). Patronin/Shot cortical foci assemble the noncentrosomal microtubule array that specifies the Drosophila anterior-posterior axis. Dev Cell 38: 61-72. PubMed ID: 27404359

    Bergstralh, D.T., Lovegrove, H.E., Kujawiak, I., Dawney, N.S., Zhu, J., Cooper, S., Zhang, R. and St Johnston, D. (2016). Pins is not required for spindle orientation in the Drosophila wing disc. Development [Epub ahead of print]. PubMed ID: 27287805

    Bergstralh, D.T., Lovegrove, H.E. and St Johnston, D. (2015). Lateral adhesion drives reintegration of misplaced cells into epithelial monolayers. Nat Cell Biol [Epub ahead of print]. PubMed ID: 26414404

  • Hugo Stocker Institute of Molecular Systems Biology, ETH Zurich
    Gupta, A. and Stocker, H. (2020). FoxO suppresses endoplasmic reticulum stress to inhibit growth of Tsc1-deficient tissues under nutrient restriction. Elife 9. PubMed ID: 32525804

    Nowak, K., Gupta, A. and Stocker, H. (2018). FoxO restricts growth and differentiation of cells with elevated TORC1 activity under nutrient restriction. PLoS Genet 14(4): e1007347. PubMed ID: 29677182

    Lujan, E., Bornemann, D. J., Rottig, C., Bayless, B. A., Stocker, H., Hafen, E., Arora, K. and Warrior, R. (2016). Analysis of novel alleles of brother of tout-velu, the Drosophila ortholog of human EXTL3 using a newly developed FRT42D ovoD chromosome. Genesis [Epub ahead of print]. PubMed ID: 27636555

    Jevtov, I., Zacharogianni, M., van Oorschot, M. M., van Zadelhoff, G., Aguilera-Gomez, A., Vuillez, I., Braakman, I., Hafen, E., Stocker, H. and Rabouille, C. (2015). TORC2 mediates the heat stress response in Drosophila by promoting the formation of stress granules. J Cell Sci. PubMed ID: 26054799

    Nowak, K., Seisenbacher, G., Hafen, E. and Stocker, H. (2013). Nutrient restriction enhances the proliferative potential of cells lacking the tumor suppressor PTEN in mitotic tissues. Elife 2: e00380. PubMed ID: 23853709

    Almudi, I., Poernbacher, I., Hafen, E., Stocker, H. (2013) The Lnk/SH2B adaptor provides a fail-safe mechanism to establish the Insulin receptor-Chico interaction. Cell Commun Signal 11: 26. PubMed ID: 23590848

    Poernbacher, I., Baumgartner, R., Marada, S. K., Edwards, K. and Stocker, H. (2012). Drosophila Pez acts in Hippo signaling to restrict intestinal stem cell proliferation. Curr Biol 22: 389-396. PubMed ID: 22305752

    Parisi, F., Riccardo, S., Daniel, M., Saqcena, M., Kundu, N., Pession, A., Grifoni, D., Stocker, H., Tabak, E. and Bellosta, P. (2011). Drosophila insulin and target of rapamycin (TOR) pathways regulate GSK3 beta activity to control Myc stability and determine Myc expression in vivo. BMC Biol 9: 65. PubMed ID: 21951762

    Seisenbacher, G., Hafen, E. and Stocker, H. (2011). MK2-dependent p38b signalling protects Drosophila hindgut enterocytes against JNK-induced apoptosis under chronic stress. PLoS Genet 7: e1002168. PubMed ID: 21829386

  • Eric Stone ANU-CSIRO Centre for Genomics, Metabolomics and Bioinformatics Australian National University
    Vensko, S. P., and Stone, E. A. (2015). Recent progress and open questions in Drosophila dosage compensation. Fly (Austin): 0. PubMed ID: 26213294

    Vensko Ii, S. P. and Stone, E. A. (2015). X-to-autosome expression and msl-2 transcript abundance correlate among Drosophila melanogaster somatic tissues. PeerJ 3: e771. PubMed ID: 25737812

    Vensko, S. P. and Stone, E. A. (2014). No evidence for a global male-specific lethal complex-mediated dosage compensation contribution to the demasculinization of the Drosophila melanogaster X chromosome. PLoS One 9: e103659. PubMed ID: 25093841

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

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

  • Mark Stopfer Laboratory of Cellular and Synaptic Neurophysiology, NIH, Bethesda, MD
    Shimizu, K. and Stopfer, M. (2017). A population of projection neurons that inhibits the lateral horn but excites the antennal lobe through chemical synapses in Drosophila. Front Neural Circuits 11: 30. PubMed ID: 28515683

    Miyazaki, T., Lin, T. Y., Ito, K., Lee, C. H. and Stopfer, M. (2015). A gustatory second-order neuron that connects sucrose-sensitive primary neurons and a distinct region of the gnathal ganglion in the Drosophila brain. J Neurogenet: 1-26. PubMed ID: 26004543

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

    Tanaka, N. K., Dye, L. and Stopfer, M. (2011). Dual-labeling method for electron microscopy to characterize synaptic connectivity using genetically encoded fluorescent reporters in Drosophila. J Neurosci Methods 194: 312-315. PubMed ID: 21074556

  • Steve Stowers Department of Cell Biology & Neuroscience, Montana State University, Bozeman
    Certel, S. J., Ruchti, E., McCabe, B. D. and Stowers, R. S. (2022). A conditional glutamatergic synaptic vesicle marker for Drosophila. G3 (Bethesda) 12(3). PubMed ID: 35100385

    McKinney, H., Sherer, L. M., Williams, J. L., Certel, S. J. and Stowers, R. S. (2020). Characterization of Drosophila Octopamine Receptor Neuronal Expression using MiMIC-converted Gal4 lines. J Comp Neurol. PubMed ID: 32060912

    Williams, J. L., Shearin, H. K. and Stowers, R. S. (2019). Conditional synaptic vesicle markers for Drosophila. G3 (Bethesda). PubMed ID: 30635441
    Shearin, H. K., Macdonald, I. S., Spector, L. P. and Stowers, R. S. (2014). Hexameric GFP and mCherry Reporters for the Drosophila GAL4, Q, and LexA Transcription Systems. Genetics. PubMed ID: 24451596

    Shearin, H. K., Dvarishkis, A. R., Kozeluh, C. D. and Stowers, R. S. (2013). Expansion of the Gateway MultiSite Recombination Cloning Toolkit. PLoS One 8: e77724. PubMed ID: 24204935

    Petersen, L. K. and Stowers, R. S. (2011). A Gateway MultiSite recombination cloning toolkit. PLoS One 6: e24531. PubMed ID: 21931740

  • Brian Stramer Kings College London
    Yolland, L., Burki, M., Marcotti, S., Luchici, A., Kenny, F. N., Davis, J. R., Serna-Morales, E., Muller, J., Sixt, M., Davidson, A., Wood, W., Schumacher, L. J., Endres, R. G., Miodownik, M. and Stramer, B. M. (2019). Persistent and polarized global actin flow is essential for directionality during cell migration. Nat Cell Biol 21(11): 1370-1381. PubMed ID: 31685997

    Matsubayashi, Y., Louani, A., Dragu, A., Sanchez-Sanchez, B. J., Serna-Morales, E., Yolland, L., Gyoergy, A., Vizcay, G., Fleck, R. A., Heddleston, J. M., Chew, T. L., Siekhaus, D. E. and Stramer, B. M. (2017). A moving source of matrix components is essential for de novo basement membrane formation. Curr Biol 27(22): 3526-3534.e3524. PubMed ID: 29129537

    Davis, J. R., Luchici, A., Mosis, F., Thackery, J., Salazar, J. A., Mao, Y., Dunn, G. A., Betz, T., Miodownik, M. and Stramer, B. M. (2015). Inter-cellular forces orchestrate contact inhibition of locomotion. Cell 161: 361-373. PubMed ID: 25799385

    Davis, J. R., Huang, C. Y., Zanet, J., Harrison, S., Rosten, E., Cox, S., Soong, D. Y., Dunn, G. A. and Stramer, B. M. (2012). Emergence of embryonic pattern through contact inhibition of locomotion. Development 139: 4555-4560. PubMed ID: 23172914

    Zanet, J., et al. (2012). Fascin promotes filopodia formation independent of its role in actin bundling. J. Cell Biol. 197(4): 477-86. PubMed Citation: 22564415

    Stramer, B., Moreira, S., Millard, T., Evans, I., Huang, C. Y., Sabet, O., Milner, M., Dunn, G., Martin, P. and Wood, W. (2010). Clasp-mediated microtubule bundling regulates persistent motility and contact repulsion in Drosophila macrophages in vivo. J Cell Biol 189: 681-689. PubMed ID: 20457764

  • Nick Strausfeld Department of Neuroscience, University of Arizona, Tucson
    Wolff, G. H. and Strausfeld, N. J. (2015). Genealogical correspondence of mushroom bodies across invertebrate phyla. Curr Biol 25: 38-44. PubMed ID: 25532890

    Ito, K., Shinomiya, K., Ito, M., Armstrong, J. D., Boyan, G., Hartenstein, V., Harzsch, S., Heisenberg, M., Homberg, U., Jenett, A., Keshishian, H., Restifo, L. L., Rossler, W., Simpson, J. H., Strausfeld, N. J., Strauss, R., Vosshall, L. B. and Insect Brain Name Working, G. (2014). A systematic nomenclature for the insect brain. Neuron 81: 755-765. PubMed ID: 24559671

    Mu, L., Ito, K., Bacon, J. P. and Strausfeld, N. J. (2012). Optic glomeruli and their inputs in Drosophila share an organizational ground pattern with the antennal lobes. J Neurosci 32: 6061-6071. PubMed ID: 22553013

    Phillips-Portillo, J. and Strausfeld, N. J. (2012). Representation of the brain's superior protocerebrum of the flesh fly, Neobellieria bullata, in the central body. J Comp Neurol 520: 3070-3087. PubMed ID: 22434505

    Sinakevitch, I., Grau, Y., Strausfeld, N. J. and Birman, S. (2010). Dynamics of glutamatergic signaling in the mushroom body of young adult Drosophila. Neural Dev 5: 10. PubMed ID: 20370889

  • Roland Strauss Neuroscience at the Johannes Gutenberg University Mainz
    Krause, T., Spindler, L., Poeck, B. and Strauss, R. (2019). Drosophila acquires a long-lasting body-size memory from visual feedback. Curr Biol. PubMed ID: 31104933

    Rieche, F., Carmine-Simmen, K., Poeck, B., Kretzschmar, D. and Strauss, R. (2018). Drosophila full-length Amyloid precursor protein is required for visual working memory and prevents age-related memory impairment. Curr Biol 28(5): 817-823.e813. PubMed ID: 29478851

    Ries, A. S., Hermanns, T., Poeck, B. and Strauss, R. (2017). Serotonin modulates a depression-like state in Drosophila responsive to lithium treatment. Nat Commun 8: 15738. PubMed ID: 28585544

    Kuntz, S., Poeck, B. and Strauss, R. (2017). Visual Working Memory Requires Permissive and Instructive NO/cGMP Signaling at Presynapses in the Drosophila Central Brain. Curr Biol [Epub ahead of print]. PubMed ID: 28216314

    Arena, P., Cali, M., Patane, L., Portera, A. and Strauss, R. (2016). A fly-inspired mushroom bodies model for sensory-motor control through sequence and subsequence learning. Int J Neural Syst: 1650035. PubMed ID: 27354193

    Triphan, T., Nern, A., Roberts, S. F., Korff, W., Naiman, D. Q. and Strauss, R. (2016). A screen for constituents of motor control and decision making in Drosophila reveals visual distance-estimation neurons. Sci Rep 6: 27000. PubMed ID: 27255169

    Ito, K., Shinomiya, K., Ito, M., Armstrong, J. D., Boyan, G., Hartenstein, V., Harzsch, S., Heisenberg, M., Homberg, U., Jenett, A., Keshishian, H., Restifo, L. L., Rossler, W., Simpson, J. H., Strausfeld, N. J., Strauss, R., Vosshall, L. B. and Insect Brain Name Working, G. (2014). A systematic nomenclature for the insect brain. Neuron 81: 755-765. PubMed ID: 24559671

    Thran, J., Poeck, B. and Strauss, R. (2013). Serum Response Factor-Mediated Gene Regulation in a Drosophila Visual Working Memory. Curr Biol. PubMed ID: 24012317

    Kuntz, S., Poeck, B., Sokolowski, M. B. and Strauss, R. (2012). The visual orientation memory of Drosophila requires Foraging (PKG) upstream of Ignorant (RSK2) in ring neurons of the central complex. Learn Mem 19: 337-340. PubMed ID: 22815538

    Poeck, B., Strauss, R. and Kretzschmar, D. (2012). Analysis of amyloid precursor protein function in Drosophila melanogaster. Exp Brain Res 217: 413-421. PubMed ID: 21912928

  • Dimitrios Stravopodis Department of Cell Biology and Biophysics, University of Athens
    Velentzas, A. D., Velentzas, P. D., Katarachia, S. A., Anagnostopoulos, A. K., Sagioglou, N. E., Thanou, E. V., Tsioka, M. M., Mpakou, V. E., Kollia, Z., Gavriil, V. E., Papassideri, I. S., Tsangaris, G. T., Cefalas, A. C., Sarantopoulou, E. and Stravopodis, D. J. (2018). The indispensable contribution of s38 protein to ovarian-eggshell morphogenesis in Drosophila melanogaster. Sci Rep 8(1): 16103. PubMed ID: 30382186

    Velentzas, A. D., Velentzas, P. D., Sagioglou, N. E., Konstantakou, E. G., Anagnostopoulos, A. K., Tsioka, M. M., Mpakou, V. E., Kollia, Z., Consoulas, C., Margaritis, L. H., Papassideri, I. S., Tsangaris, G. T., Sarantopoulou, E., Cefalas, A. C. and Stravopodis, D. J. (2016). Targeted downregulation of s36 protein unearths its cardinal role in chorion biogenesis and architecture during Drosophila melanogaster oogenesis. Sci Rep 6: 35511. PubMed ID: 27752139

    Velentzas, P. D., Velentzas, A. D., Pantazi, A. D., Mpakou, V. E., Zervas, C. G., Papassideri, I. S. and Stravopodis, D. J. (2013). Proteasome, but not autophagy, disruption results in severe eye and wing dysmorphia: a subunit- and regulator-dependent process in Drosophila. PLoS One 8: e80530. PubMed ID: 24282550

    Manta, A. K., Stravopodis, D. J., Papassideri, I. S. and Margaritis, L. H. (2013). Reactive oxygen species elevation and recovery in Drosophila bodies and ovaries following short-term and long-term exposure to DECT base EMF. Electromagn Biol Med. PubMed ID: 23781995

    Velentzas, P. D., Velentzas, A. D., Mpakou, V. E., Antonelou, M. H., Margaritis, L. H., Papassideri, I. S. and Stravopodis, D. J. (2013). Detrimental effects of proteasome inhibition activity in Drosophila melanogaster: implication of ER stress, autophagy, and apoptosis. Cell Biol Toxicol 29: 13-37. PubMed ID: 23161111

    Velentzas, P. D., Velentzas, A. D., Mpakou, V. E., Papassideri, I. S., Stravopodis, D. J. and Margaritis, L. H. (2011). Proteasome inhibition induces developmentally deregulated programs of apoptotic and autophagic cell death during Drosophila melanogaster oogenesis. Cell Biol Int 35: 15-27. PubMed ID: 20819072

  • Nicholas Stavropoulos Waksman Institute of Microbiology, Piscataway, NJ
    Li, Q., Jang, H., Lim, K. Y., Lessing, A. and Stavropoulos, N. (2021). insomniac links the development and function of a sleep-regulatory circuit. Elife 10. PubMed ID: 34908527

    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

    Alvarez-Salvado, E., Licata, A. M., Connor, E. G., McHugh, M. K., King, B. M., Stavropoulos, N., Victor, J. D., Crimaldi, J. P. and Nagel, K. I. (2018). Elementary sensory-motor transformations underlying olfactory navigation in walking fruit-flies. Elife 7. PubMed ID: 30129438

  • Andrew Straw Research Institute of Molecular Pathology, Vienna, Austria
    Panser, K., Tirian, L., Schulze, F., Villalba, S., Jefferis, G.S., Bühler, K. and Straw, A.D. (2016). Automatic segmentation of Drosophila neural compartments using GAL4 expression data reveals novel visual pathways. Curr Biol [Epub ahead of print]. PubMed ID: 27426516

    Fenk, L. M., Poehlmann, A. and Straw, A. D. (2014). Asymmetric processing of visual motion for simultaneous object and background responses. Curr Biol. PubMed ID: 25454785

    Bath, D. E., Stowers, J. R., Hormann, D., Poehlmann, A., Dickson, B. J. and Straw, A. D. (2014). FlyMAD: rapid thermogenetic control of neuronal activity in freely walking Drosophila. Nat Methods 11: 756-762. PubMed ID: 24859752

    Fuller, S. B., Straw, A. D., Peek, M. Y., Murray, R. M. and Dickinson, M. H. (2014). Flying Drosophila stabilize their vision-based velocity controller by sensing wind with their antennae. Proc Natl Acad Sci U S A 111: E1182-1191. PubMed ID: 24639532

    Censi, A., Straw, A. D., Sayaman, R. W., Murray, R. M. and Dickinson, M. H. (2013). Discriminating external and internal causes for heading changes in freely flying Drosophila. PLoS Comput Biol 9: e1002891. PubMed ID: 23468601

  • Beth Stronach Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine
    Garlena, R.A., Lennox, A.L., Baker, L.R., Parsons, T.E., Weinberg, S.M. and Stronach, B.E. (2015). Pvr receptor tyrosine kinase promotes tissue closure by coordinating corpse removal and epidermal zippering. Development [Epub ahead of print]. PubMed ID: 26293306

    Stronach, B. (2014). Extensive nonmuscle expression and epithelial apicobasal localization of the Drosophila ALP/Enigma family protein, Zasp52. Gene Expr Patterns. PubMed ID: 24846727

    Stronach, B., Lennox, A. L. and Garlena, R. A. (2014). Domain Specificity of MAP3K Family Members, MLK and Tak1, for JNK Signaling in Drosophila. Genetics. PubMed ID: 24429281

    Gonda, R. L., Garlena, R. A. and Stronach, B. (2012). Drosophila heat shock response requires the JNK pathway and phosphorylation of mixed lineage kinase at a conserved serine-proline motif. PLoS One 7: e42369. PubMed ID: 22848763

  • Gary Struhl Department of Genetics and Development, Columbia University
    Zecca, M. and Struhl, G. (2021). A unified mechanism for the control of Drosophila wing growth by the morphogens Decapentaplegic and Wingless. PLoS Biol 19(3): e3001111. PubMed ID: 33657096

    Parker, J. and Struhl, G. (2020). Control of Drosophila wing size by morphogen range and hormonal gating. Proc Natl Acad Sci U S A 117(50): 31935-31944. PubMed ID: 33257577

    Langridge, P. D. and Struhl, G. (2017). Epsin-dependent ligand endocytosis activates Notch by force. Cell 171(6): 1383-1396.e1312. PubMed ID: 29195077

    Coleman, R. T. and Struhl, G. (2017). Causal role for inheritance of H3K27me3 in maintaining the OFF state of a Drosophila HOX gene. Science [Epub ahead of print]. PubMed ID: 28302795

    Parker, J. and Struhl, G. (2015). Scaling the Drosophila wing: TOR-dependent target gene access by the Hippo pathway transducer Yorkie. PLoS Biol 13: e1002274. PubMed ID: 26474042

    Kidd, S., Struhl, G. and Lieber, T. (2015). Notch is required in adult Drosophila sensory neurons for morphological and functional plasticity of the olfactory circuit. PLoS Genet 11: e1005244. PubMed ID: 26011623

    Tomlinson, A., Mavromatakis, Y. E. and Struhl, G. (2011). Three distinct roles for notch in Drosophila R7 photoreceptor specification. PLoS Biol 9: e1001132. PubMed ID: 21886484

    Lieber, T., Kidd, S. and Struhl, G. (2011). DSL-Notch signaling in the Drosophila brain in response to olfactory stimulation. Neuron 69: 468-481. PubMed ID: 21315258

    Zecca, M. and Struhl, G. (2010). A feed-forward circuit linking wingless, fat-dachsous signaling, and the warts-hippo pathway to Drosophila wing growth. PLoS Biol 8: e1000386. PubMed ID: 20532238

  • David I. Strutt Department of Biomedical Science, The University of Sheffield
    Warrington, S. J., Strutt, H. and Strutt, D. (2022). Use of Fluorescence Recovery After Photobleaching (FRAP) to Measure In Vivo Dynamics of Cell Junction-Associated Polarity Proteins. Methods Mol Biol 2438: 1-30. PubMed ID: 35147932

    Strutt, H. and Strutt, D. (2020). DAnkrd49 and Bdbt act via Casein kinase Iε to regulate planar polarity in Drosophila. PLoS Genet 16(8): e1008820. PubMed ID: 32750048

    Strutt, H., Gamage, J. and Strutt, D. (2019). Reciprocal action of Casein Kinase Iepsilon on core planar polarity proteins regulates clustering and asymmetric localisation. Elife 8. PubMed ID: 31090542

    Strutt, H., Langton, P. F., Pearson, N., McMillan, K. J., Strutt, D. and Cullen, P. J. (2019). Retromer controls planar polarity protein levels and asymmetric localization at intercellular iunctions. Curr Biol 29(3): 484-491.e486. PubMed ID: 30661800

    Ressurreicao, M., Warrington, S. and Strutt, D. (2018). Rapid disruption of Dishevelled activity uncovers an intercellular role in maintenance of Prickle in core planar polarity protein complexes. Cell Rep 25(6): 1415-1424.e1416. PubMed ID: 30403998

    Warrington, S. J., Strutt, H., Fisher, K. H. and Strutt, D. (2017). A dual function for Prickle in regulating frizzled stability during feedback-dependent amplification of planar polarity. Curr Biol 27(18): 2784-2797.e2783. PubMed ID: 28918952

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

    Hale, R., Brittle, A. L., Fisher, K. H., Monk, N. A. and Strutt, D. (2015). Cellular interpretation of the long-range gradient of Four-jointed activity in the Drosophila wing. Elife 4. PubMed ID: 25707557

    Thomas, C. and Strutt, D. (2013). Rabaptin-5 and Rabex-5 are neoplastic tumour suppressor genes that interact to modulate Rab5 dynamics in Drosophila melanogaster. Dev Biol. PubMed ID: 24104056

    Wells, R. E., Barry, J. D., Warrington, S. J., Cuhlmann, S., Evans, P., Huber, W., Strutt, D. and Zeidler, M. P. (2013). Control of tissue morphology by Fasciclin III-mediated intercellular adhesion. Development. PubMed ID: 23946443

    Johnstone, K., Wells, R. E., Strutt, D. and Zeidler, M. P. (2013). Localised Jak/STAT pathway activation is required for Drosophila wing hinge development. PLoS One 8: e65076. PubMed ID: 23741461

  • William Stumph Biology Department, San Diego State University
    Kim, M. K., Tranvo, A., Hurlburt, A. M., Verma, N., Phan, P., Luo, J., Ranish, J. and Stumph, W. E. (2020). Assembly of SNAPc, Bdp1, and TBP on the U6 snRNA gene promoter in Drosophila melanogaster. Mol Cell Biol. PubMed ID: 32253345

    Verma, N., Hurlburt, A. M., Wolfe, A., Kim, M. K., Kang, Y. S., Kang, J. J. and Stumph, W. E. (2018). Bdp1 interacts with SNAPc bound to a U6, but not U1, snRNA gene promoter element to establish a stable protein-DNA complex. FEBS Lett. PubMed ID: 29932462

    Kang, Y. S., Kurano, M. and Stumph, W. E. (2014). The Myb domain of the largest subunit of SNAPc adopts different architectural configurations on U1 and U6 snRNA gene promoter sequences. Nucleic Acids Res. PubMed ID: 25324315

    Verma, N., Hung, K. H., Kang, J. J., Barakat, N. H. and Stumph, W. E. (2013). Differential Utilization of TATA Box-Binding Protein (TBP) and TBP-related Factor 1 (TRF1) at Different Classes of RNA Polymerase III Promoters. J Biol Chem. PubMed ID: 23955442

    Doherty, M. T., Kang, Y. S., Lee, C. and Stumph, W. E. (2012). Architectural arrangement of the small nuclear RNA (snRNA)-activating protein complex 190 subunit (SNAP190) on U1 snRNA gene promoter DNA. J Biol Chem 287: 39369-39379. PubMed ID: 23038247

    Hung, K. H. and Stumph, W. E. (2012). Localization of residues in a novel DNA-binding domain of DmSNAP43 required for DmSNAPc DNA-binding activity. FEBS Lett 586: 841-846. PubMed ID: 22449969

    Hung, K. H. and Stumph, W. E. (2011). Regulation of snRNA gene expression by the Drosophila melanogaster small nuclear RNA activating protein complex (DmSNAPc). Crit Rev Biochem Mol Biol 46: 11-26. PubMed ID: 20925482

  • Chih-ying Su School of Biological Sciences, U.C. San Diego
    Wu, S. T., Chen, J. Y., Martin, V., Ng, R., Zhang, Y., Grover, D., Greenspan, R. J., Aljadeff, J. and Su, C. Y. (2022). Valence opponency in peripheral olfactory processing. Proc Natl Acad Sci U S A 119(5). PubMed ID: 35091473

    Nava Gonzales, C., McKaughan, Q., Bushong, E. A., Cauwenberghs, K., Ng, R., Madany, M., Ellisman, M. H. and Su, C. Y. (2021). Systematic morphological and morphometric analysis of identified olfactory receptor neurons in Drosophila melanogaster. Elife 10. PubMed ID: 34423777

    Kohlmeier, P., Zhang, Y., Gorter, J. A., Su, C. Y. and Billeter, J. C. (2021). Mating increases Drosophila melanogaster females' choosiness by reducing olfactory sensitivity to a male pheromone. Nat Ecol Evol 5(8): 1165-1173. PubMed ID: 34155384

    Vulpe, A., Kim, H. S., Ballou, S., Wu, S. T., Grabe, V., Nava Gonzales, C., Liang, T., Sachse, S., Jeanne, J. M., Su, C. Y. and Menuz, K. (2021). An ammonium transporter is a non-canonical olfactory receptor for ammonia. Curr Biol 31(15): 3382-3390 e3387. PubMed ID: 34111404

  • Tin Tin Su Molecular, Cellular and Developmental Biology, University of Colorado, Boulder
    Ledru, M., Clark, C. A., Brown, J., Verghese, S., Ferrara, S., Goodspeed, A. and Su, T. T. (2022). Differential gene expression analysis identified determinants of cell fate plasticity during radiation-induced regeneration in Drosophila. PLoS Genet 18(1): e1009989. PubMed ID: 34990447

    Brown, J., Bush, I., Bozon, J. and Su, T. T. (2020). Cells with loss-of-heterozygosity after exposure to ionizing radiation in Drosophila are culled by p53-dependent and p53-independent mechanisms. PLoS Genet 16(10): e1009056. PubMed ID: 33075096

    Myat, M. M., Louis, D., Mavrommatis, A., Collins, L., Mattis, J., Ledru, M., Verghese, S. and Su, T. T. (2019). Regulators of cell movement during development and regeneration in Drosophila. Open Biol 9(5): 180245. PubMed ID: 31039676

    Verghese, S. and Su, T. T. (2018). Ionizing radiation induces stem cell-like properties in a caspase-dependent manner in Drosophila. PLoS Genet 14(11): e1007659. PubMed ID: 30462636

    Verghese, S. and Su, T. T. (2017). STAT, Wingless, and Nurf-38 determine the accuracy of regeneration after radiation damage in Drosophila. PLoS Genet 13(10): e1007055. PubMed ID: 29028797 wNotes 4: 569. PubMed ID: 22206604

    Zhou, D., Udpa, N., Gersten, M., Visk, D. W., Bashir, A., Xue, J., Frazer, K. A., Posakony, J. W., Subramaniam, S., Bafna, V. and Haddad, G. G. (2011). Experimental selection of hypoxia-tolerant Drosophila melanogaster. Proc Natl Acad Sci U S A 108: 2349-2354. PubMed ID: 21262834

  • Elio Sucena Instituto Gulbenkian de Ciencia, Oeiras Portugal
    Nunes, C., Koyama, T. and Sucena, E. (2021). Co-option of immune effectors by the hormonal signalling system triggering metamorphosis in Drosophila melanogaster. PLoS Genet 17(11): e1009916. PubMed ID: 34843450

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

    Leit&atilde;o, A.B. and Sucena, È (2015). Drosophila sessile hemocyte clusters are true hematopoietic tissues that regulate larval blood cell differentiation. Elife 4 [Epub ahead of print]. PubMed ID: 25650737

    Martins, N. E., Faria, V. G., Teixeira, L., Magalhaes, S. and Sucena, E. (2013). Host Adaptation Is Contingent upon the Infection Route Taken by Pathogens. PLoS Pathog 9: e1003601. PubMed ID: 24086131

    Arif, S., Murat, S., Almudi, I., Nunes, M. D., Bortolamiol-Becet, D., McGregor, N. S., Currie, J. M., Hughes, H., Ronshaugen, M., Sucena, E., Lai, E. C., Schlotterer, C. and McGregor, A. P. (2013). Evolution of mir-92a underlies natural morphological variation in Drosophila melanogaster. Curr Biol 23: 523-528. PubMed ID: 23453955

    Stern, D. L. and Sucena, E. (2012). Preparation and mounting of adult Drosophila structures in Canada balsam. Cold Spring Harb Protoc 2012: 373-375. PubMed ID: 22383636

    Stern, D. L. and Sucena, E. (2012). Rapid mounting of adult Drosophila structures in Hoyer's medium. Cold Spring Harb Protoc 2012: 107-109. PubMed ID: 22194256

  • Atsushi Sugie Brain Research Institute, Niigata University, Japan
    Nitta, Y. and Sugie, A. (2017). Identification of glaikit in a genome-wide expression profiling for axonal bifurcation of the mushroom body in Drosophila. Biochem Biophys Res Commun 487(4): 898-902. PubMed ID: 28465232

    Nitta, Y. and Sugie, A. (2017). DISCO interacting protein 2 determines direction of axon projection under the regulation of c-Jun N-terminal kinase in the Drosophila mushroom body. Biochem Biophys Res Commun 487(1): 116-121. PubMed ID: 28396149

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

    Nitta, Y., Yamazaki, D., Sugie, A., Hiroi, M. and Tabata, T. (2017). DISCO Interacting Protein 2 regulates axonal bifurcation and guidance of Drosophila mushroom body neurons. Dev Biol 421(2): 233-244. PubMed ID: 27908785

  • Kaoru Sugimura Institute for Integrated Cell-Material Sciences, Kyoto University
    Ikawa, K. and Sugimura, K. (2018). AIP1 and cofilin ensure a resistance to tissue tension and promote directional cell rearrangement. Nat Commun 9(1): 3295. PubMed ID: 30202062

    Arata, M., Sugimura, K. and Uemura, T. (2017). Difference in Dachsous levels between migrating cells coordinates the direction of collective cell migration. Dev Cell 42(5): 479-497.e410. PubMed ID: 28898677

    Sugimura, K. and Ishihara, S. (2013). The mechanical anisotropy in a tissue promotes ordering in hexagonal cell packing. Development 140: 4091-4101. PubMed ID: 24046322

    Sano, H., Kunwar, P. S., Renault, A. D., Barbosa, V., Clark, I. B., Ishihara, S., Sugimura, K. and Lehmann, R. (2012). The Drosophila actin regulator ENABLED regulates cell shape and orientation during gonad morphogenesis. PLoS One 7: e52649. PubMed ID: 23300733

    Ohsawa, S., Sugimura, K., Takino, K. and Igaki, T. (2012). Imaging cell competition in Drosophila imaginal discs. Methods Enzymol 506: 407-413. PubMed ID: 22341235

  • Greg Suh Skirball Institute Program of Molecular Neurobiology and Cell Biology, New York
    Min, S., Oh, Y., Verma, P., Whitehead, S. C., Yapici, N., Van Vactor, D., Suh, G. S. and Liberles, S. (2021). Control of feeding by Piezo-mediated gut mechanosensation in Drosophila. Elife 10. PubMed ID: 33599608 Lim, S. Y., You, H., Lee, J., Lee, J., Lee, Y.,

    Lee, K. A., Kim, B., Lee, J. H., Jeong, J., Jang, S., Kim, B., Choi, H., Hwang, G., Choi, M. S., Yoon, S. E., Kwon, J. Y., Lee, W. J., Kim, Y. J. and Suh, G. S. B. (2020). Identification and characterization of GAL4 drivers that mark distinct cell types and regions in the Drosophila adult gut. J Neurogenet: 1-11. PubMed ID: 33326321

    Park, J.Y., Dus, M., Kim, S., Abu, F., Kanai, M.I., Rudy, B. and Suh, G.S. (2016). SLC5A11 mediates hunger by regulating K+ channel activity. Curr Biol [Epub ahead of print]. PubMed ID: 27397890

    Dus, M., Lai, J.S., Gunapala, K.M., Min, S., Tayler, T.D., Hergarden, A.C., Geraud, E., Joseph, C.M. and Suh, G.S. (2015). Nutrient sensor in the brain directs the action of the brain-gut axis in Drosophila. Neuron [Epub ahead of print]. PubMed ID: 26074004

    Ai, M., Blais, S., Park, J. Y., Min, S., Neubert, T. A. and Suh, G. S. (2013). Ionotropic Glutamate Receptors IR64a and IR8a Form a Functional Odorant Receptor Complex In Vivo in Drosophila. J Neurosci 33: 10741-10749. PubMed ID: 23804096

    Dus, M., Ai, M., Suh, G. S. (2013) Taste-independent nutrient selection is mediated by a brain-specific Na/solute co-transporter in Drosophila. Nat Neurosci. PubMed ID: 23542692

    Min, S., Ai, M., Shin, S. A. and Suh, G. S. (2013). Dedicated olfactory neurons mediating attraction behavior to ammonia and amines in Drosophila. Proc Natl Acad Sci U S A 110: E1321-1329. PubMed ID: 23509267

    Dus, M., Min, S., Keene, A. C., Lee, G. Y. and Suh, G. S. (2011). Taste-independent detection of the caloric content of sugar in Drosophila. Proc Natl Acad Sci U S A 108: 11644-11649. PubMed ID: 21709242

    Ai, M., Min, S., Grosjean, Y., Leblanc, C., Bell, R., Benton, R. and Suh, G. S. (2010). Acid sensing by the Drosophila olfactory system. Nature 468: 691-695. PubMed ID: 21085119

  • Bill Sullivan Molecular, Cell and Developmental Biology, U. C. at Santa Cruz
    Warecki, B., Bast, I. and Sullivan, W. (2022). Visualizing the Dynamics of Cell Division by Live Imaging Drosophila Larval Brain Squashes. Methods Mol Biol 2415: 37-46. PubMed ID: 34972944

    Vicars, H., Karg, T., Warecki, B., Bast, I. and Sullivan, W. (2021). Kinetochore-independent mechanisms of sister chromosome separation. PLoS Genet 17(1): e1009304. PubMed ID: 33513180

    Warecki, B., Ling, X., Bast, I. and Sullivan, W. (2020). ESCRT-III-mediated membrane fusion drives chromosome fragments through nuclear envelope channels. J Cell Biol 219(3). PubMed ID: 32032426

    Russell, S. L., Lemseffer, N. and Sullivan, W. T. (2018). Wolbachia and host germline components compete for kinesin-mediated transport to the posterior pole of the Drosophila oocyte. PLoS Pathog 14(8): e1007216. PubMed ID: 30110391

    Warecki, B. and Sullivan, W. (2018). Micronuclei formation is prevented by Aurora B-mediated exclusion of HP1a from late-segregating chromatin in Drosophila. Genetics. PubMed ID: 29986897

    Karg, T., Elting, M. W., Vicars, H., Dumont, S. and Sullivan, W. (2017). The chromokinesin Klp3a and microtubules facilitate acentric chromosome segregation. J Cell Biol. PubMed ID: 28500183

    Brose, L., Crest, J., Tao, L. and Sullivan, W. (2017). Polo kinase mediates the phosphorylation and cellular localization of Nuf/FIP3, a Rab11 effector. Mol Biol Cell [Epub ahead of print]. PubMed ID: 28381422

    White, P. M., Serbus, L. R., Debec, A., Codina, A., Bray, W., Guichet, A., Lokey, R. S. and Sullivan, W. (2017). Reliance of Wolbachia on high rates of host proteolysis revealed by a genome-wide RNAi screen of Drosophila cells. Genetics [Epub ahead of print]. PubMed ID: 28159754

    White, P. M., Pietri, J. E., Debec, A., Russell, S., Patel, B. and Sullivan, W. (2017). Mechanisms of horizontal cell-to-cell transfer of Wolbachia spp. in Drosophila melanogaster. Appl Environ Microbiol [Epub ahead of print]. PubMed ID: 28087534

    Tao, L., Fasulo, B., Warecki, B. and Sullivan, W. (2016). Tum/RacGAP functions as a switch activating the Pav/kinesin-6 motor. Nat Commun 7: 11182. PubMed ID: 27091402

    Karg, T., Warecki, B. and Sullivan, W. (2015). Aurora B mediated localized delays in nuclear envelope formation facilitates inclusion of late segregating chromosome fragments. Mol Biol Cell [Epub ahead of print]. PubMed ID: 25877868

  • Fang-Lin Sun Tsinghua University, Beijing
    Pan, L., Xie, W., Li, K.L., Yang, Z., Xu, J., Zhang, W., Liu, L.P., Ren, X., He, Z., Wu, J., Sun, J., Wei, H.M., Wang, D., Xie, W., Li, W., Ni, J.Q. and Sun, F.L. (2015). Heterochromatin remodeling by CDK12 contributes to learning in Drosophila. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 26508632

    Lv, W. W., Wei, H. M., Wang, D. L., Ni, J. Q. and Sun, F. L. (2012). Depletion of histone deacetylase 3 antagonizes PI3K-mediated overgrowth through the acetylation of histone H4 at lysine 16. J Cell Sci. PubMed ID: 22956542

    Chen, S., Wei, H. M., Lv, W. W., Wang, D. L. and Sun, F. L. (2011). E2 ligase dRad6 regulates DMP53 turnover in Drosophila. J Biol Chem 286: 9020-9030. PubMed ID: 21205821

  • Jianjun Sun Department of Physiology and Neurobiology, University of Connecticut
    Knapp, E. M., Li, W., Singh, V. and Sun, J. (2020). Nuclear receptor Ftz-f1 promotes follicle maturation and ovulation partly via bHLH/PAS transcription factor Sim. Elife 9. PubMed ID: 32338596

    Shen, W. and Sun, J. (2020). Different modes of Notch activation and strength regulation in the spermathecal secretory lineage. Development 147(3). PubMed ID: 31988187

    Knapp, E. M., Li, W. and Sun, J. (2019). Downregulation of homeodomain protein Cut is essential for follicle maturation and ovulation. Development. PubMed ID: 31444217

    Li, W., Young, J. F. and Sun, J. (2018). NADPH oxidase-generated reactive oxygen species in mature follicles are essential for Drosophila ovulation. Proc Natl Acad Sci U S A 115(30): 7765-7770. PubMed ID: 29987037

    Damrau, C., Toshima, N., Tanimura, T., Brembs, B. and Colomb, J. (2017). Octopamine and tyramine contribute separately to the counter-regulatory response to sugar deficit in Drosophila. Front Syst Neurosci 11: 100. PubMed ID: 29379421

    Shen, W. and Sun, J. (2017). Dynamic Notch signaling specifies each cell fate in Drosophila spermathecal lineage. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 28258114

    Knapp, E. and Sun, J. (2017). Steroid signaling in mature follicles is important for Drosophila ovulation. Proc Natl Acad Sci U S A [Epub ahead of print]. PubMed ID: 28069934

    Deady, L. D. and Sun, J. (2015). A Follicle Rupture Assay Reveals an Essential Role for Follicular Adrenergic Signaling in Drosophila Ovulation. PLoS Genet 11: e1005604. PubMed ID: 26473732

    Deady, L. D., Shen, W., Mosure, S. A., Spradling, A. C. and Sun, J. (2015). Matrix metalloproteinase 2 is required for ovulation and corpus luteum formation in Drosophila. PLoS Genet 11: e1004989. PubMed ID: 25695427

    Sun, J. and Spradling, A. C. (2013). Ovulation in Drosophila is controlled by secretory cells of the female reproductive tract. Elife 2: e00415. PubMed ID: 23599892

  • Y. Henry Sun Institute of Molecular Biology, Taiwan
    Tsao, C. K., Huang, Y. F. and Sun, Y. H. (2020). Early lineage segregation of the retinal basal glia in the Drosophila eye disc. Sci Rep 10(1): 18522. PubMed ID: 33116242

    Chang, L. W., Tseng, I. C., Wang, L. H. and Sun, Y. H. (2020). Isoform-specific functions of an evolutionarily conserved 3 bp micro-exon alternatively spliced from another exon in Drosophila homothorax gene. Sci Rep 10(1): 12783. PubMed ID: 32732884

    Chang, Y. C., Tsao, C. K. and Sun, Y. H. (2018). Temporal and spatial order of photoreceptor and glia projections into optic lobe in Drosophila. Sci Rep 8(1): 12669. PubMed ID: 30140062

    Ku, H. Y. and Sun, Y. H. (2017). Notch-dependent epithelial fold determines boundary formation between developmental fields in the Drosophila antenna. PLoS Genet 13(7): e1006898. PubMed ID: 28708823

    Lee, Y. M. and Sun, Y. H. (2015). Drosophila as a model to study the role of glia in neurodegeneration. J Neurogenet: 1-11. PubMed ID: 26312528

    Lee, Y.M. and Sun, Y.H. (2015). Maintenance of glia in the optic lamina i s mediated by EGFR signaling by photoreceptors in adult Drosophila. PLoS Genet 11: e1005187. PubMed ID: 25909451

    Chu, W. C., Lee, Y. M. and Henry Sun, Y. (2013). FGF /FGFR Signal Induces Trachea Extension in the Drosophila Visual System. PLoS One 8: e73878. PubMed ID: 23991208

    Wang, C. W. and Sun, Y. H. (2012). Segregation of eye and antenna fates maintained by mutual antagonism in Drosophila. Development 139(18): 3413-21. PubMed Citation: 22912416

    Wang, L. H., Huang, Y. T., Tsai, Y. C. and Sun, Y. H. (2010). The role of eyg Pax gene in the development of the head vertex in Drosophila. Dev Biol 337: 246-258. PubMed ID: 19896935

  • Claudio Sunkel Institute for Cellular and Molecular Biology, Porto, Portugal
    Bras, R., Monteiro, A., Sunkel, C. E. and Resende, L. P. (2021). Aneuploidy facilitates dysplastic and tumorigenic phenotypes in the Drosophila gut. Biol Open. PubMed ID: 33948620

    Barbosa, J., Martins, T., Bange, T., Tao, L., Conde, C. and Sunkel, C. (2019). Polo regulates Spindly to prevent premature stabilization of kinetochore-microtubule attachments. Embo j: e100789. PubMed ID: 31849090

    Oliveira, M. S., Freitas, J., Pinto, P. A. B., de Jesus, A., Tavares, J., Pinho, M., Domingues, R. G., Henriques, T., Lopes, C., Conde, C., Sunkel, C. E. and Moreira, A. (2019). The cell cycle kinase Polo is controlled by a conserved 3'UTR regulatory sequence in Drosophila melanogaster. Mol Cell Biol. PubMed ID: 31085682

    Moreira, S., Osswald, M., Ventura, G., Goncalves, M., Sunkel, C. E. and Morais-de-Sa, E. (2019). PP1-Mediated dephosphorylation of Lgl controls apical-basal polarity. Cell Rep 26(2): 293-301.e297. PubMed ID: 30625311

    Resende, L. P., Monteiro, A., Bras, R., Lopes, T. and Sunkel, C. E. (2018). Aneuploidy in intestinal stem cells promotes gut dysplasia in Drosophila. J Cell Biol. PubMed ID: 30282810

    Moura, M., Osswald, M., Leca, N., Barbosa, J., Pereira, A. J., Maiato, H., Sunkel, C. E. and Conde, C. (2017). Protein Phosphatase 1 inactivates Mps1 to ensure efficient Spindle Assembly Checkpoint silencing. Elife 6 [Epub ahead of print]. PubMed ID: 28463114

    Carvalho, C.A., Moreira, S., Ventura, G., Sunkel, C.E. and Morais-de-Sá, E. (2015). Aurora A triggers Lgl cortical release during symmetric division to controlplanar spindle orient ation. Curr Biol. 25: 53-60. PubMed ID: 25484294

    Carvalho, C. A., Moreira, S., Ventura, G., Sunkel, C. E. and Morais-de-Sa, E. (2014). Aurora A Triggers Lgl Cortical Release during Symmetric Division to Control Planar Spindle Orientation. Curr Biol. PubMed ID: 25484294

    Conde, C., Osswald, M. and Sunkel, C. E. (2013). All together now: Polo joins the kinase network controlling the spindle assembly checkpoint in Drosophila. Fly (Austin) 7. PubMed ID: 23989224

    Mathieu, J., Cauvin, C., Moch, C., Radford, S. J., Sampaio, P., Perdigoto, C. N., Schweisguth, F., Bardin, A. J., Sunkel, C. E., McKim, K., Echard, A. and Huynh, J. R. (2013). Aurora B and cyclin B have opposite effects on the timing of cytokinesis abscission in Drosophila germ cells and in vertebrate somatic cells. Dev Cell 26: 250-265. PubMed ID: 23948252

  • Beat Suter Institute of Cell Biology, Universitat Bern
    Vazquez-Pianzola, P., Beuchle, D., Saro, G., Hernandez, G., Maldonado, G., Brunssen, D., Meister, P. and Suter, B. (2022). Female meiosis II and pronuclear fusion require the microtubule transport factor Bicaudal-D. Development. PubMed ID: 35723263

    Chippalkatti, R., Egger, B. and Suter, B. (2020). Mms19 promotes spindle microtubule assembly in Drosophila neural stem cells. PLoS Genet 16(11): e1008913. PubMed ID: 33211700

    Nag, R. N., Niggli, S., Sousa-Guimaraes, S., Vazquez-Pianzola, P. and Suter, B. (2018). Mms19 is a mitotic gene that permits Cdk7 to be fully active as a Cdk-activating kinase. Development 145(2). PubMed ID: 29361561

    Vazquez-Pianzola, P., Schaller, B., Colombo, M., Beuchle, D., Neuenschwander, S., Marcil, A., Bruggmann, R. and Suter, B. (2016). The mRNA transportome of the BicD/Egl transport machinery. RNA Biol [Epub ahead of print]. PubMed ID: 27801632

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

    Stettler, K., Li, X., Sandrock, B., Braga-Lagache, S., Heller, M., Dumbgen, L. and Suter, B. (2014). A Drosophila XPD model links cell cycle coordination with neuro-development and suggests links to cancer. Dis Model Mech. PubMed ID: 25431422

    Lu, J., Bergert, M., Walther, A. and Suter, B. (2014). Double-sieving-defective aminoacyl-tRNA synthetase causes protein mistranslation and affects cellular physiology and development. Nat Commun 5: 5650. PubMed ID: 25427601

    Vazquez-Pianzola, P., Adam, J., Haldemann, D., Hain, D., Urlaub, H. and Suter, B. (2014). Clathrin heavy chain plays multiple roles in polarizing the Drosophila oocyte downstream of Bic-D. Development. PubMed ID: 24718986

    Urwyler, O., Cortinas-Elizondo, F. and Suter, B. (2012). Drosophila sosie functions with beta(H)-Spectrin and actin organizers in cell migration, epithelial morphogenesis and cortical stability. Biol Open 1: 994-1005. PubMed ID: 23213377

    Li, X., Urwyler, O. and Suter, B. (2010). Drosophila Xpd regulates Cdk7 localization, mitotic kinase activity, spindle dynamics, and chromosome segregation. PLoS Genet 6: e1000876. PubMed ID: 20300654

  • Magali Suzanne Laboratoire de Biologie Cellulaire et Moléculaire du Controle de la Prolifération, CNRS, Toulouse
    Martin, E., Theis, S., Gay, G., Monier, B., Rouviere, C. and Suzanne, M. (2021). Arp2/3-dependent mechanical control of morphogenetic robustness in an inherently challenging environment. Dev Cell 56(5): 687-701.e687. PubMed ID: 33535069

    Gracia, M., Theis, S., Proag, A., Gay, G., Benassayag, C. and Suzanne, M. (2019). Mechanical impact of epithelial-mesenchymal transition on epithelial morphogenesis in Drosophila. Nat Commun 10(1): 2951. PubMed ID: 31273212

    Proag, A., Monier, B. and Suzanne, M. (2019). Physical and functional cell-matrix uncoupling in a developing tissue under tension. Development. PubMed ID: 31064785

    Anais Tiberghien, M., Lebreton, G., Cribbs, D., Benassayag, C. and Suzanne, M. through regulation of basal DE-Cadherin distribution. Dev Biol [Epub ahead of print]. PubMed ID: 26206615

    Monier, B., Gettings, M., Gay, G., Mangeat, T., Schott, S., Guarner, A. and Suzanne, M. (2015). Apico-basal forces exerted by apoptotic cells drive epithelium folding. Nature 518: 245-248. PubMed ID: 25607361

    Guarner, A., Manjon, C., Edwards, K., Steller, H., Suzanne, M. and Sanchez-Herrero, E. (2014). The zinc finger homeodomain-2 gene of Drosophila controls Notch targets and regulates apoptosis in the tarsal segments. Dev Biol 385: 350-365. PubMed ID: 24144920

    Coutelis, J. B., Geminard, C., Speder, P., Suzanne, M., Petzoldt, A. G. and Noselli, S. (2013). Drosophila left/right asymmetry establishment is controlled by the Hox gene abdominal-B. Dev Cell 24: 89-97. PubMed ID: 23328400

  • Takashi Suzuki Tokyo Institute of Technology
    Bai, Y. and Suzuki, T. (2022). Activity-dependent circuitry plasticity via the regulation of the histamine receptor level in the Drosophila visual system. Mol Cell Neurosci 119: 103703. PubMed ID: 35122941

    Takechi, H., Hakeda-Suzuki, S., Nitta, Y., Ishiwata, Y., Iwanaga, R., Sato, M., Sugie, A. and Suzuki, T. (2021). Glial insulin regulates cooperative or antagonistic Golden goal/Flamingo interactions during photoreceptor axon guidance. Elife 10. PubMed ID: 33666170

    Kawamura, H., Hakeda-Suzuki, S. and Suzuki, T. (2020). Activity-dependent endocytosis of Wingless regulates synaptic plasticity in the Drosophila visual system. Genes Genet Syst. PubMed ID: 33298662

    Araki, T., Osaka, J., Kato, Y., Shimozono, M., Kawamura, H., Iwanaga, R., Hakeda-Suzuki, S. and Suzuki, T. (2020). Systematic identification of genes regulating synaptic remodeling in the Drosophila visual system. Genes Genet Syst. PubMed ID: 32493879

    Shimozono, M., Osaka, J., Kato, Y., Araki, T., Kawamura, H., Takechi, H., Hakeda-Suzuki, S. and Suzuki, T. (2019). Cell surface molecule, Klingon, mediates the refinement of synaptic specificity in the Drosophila visual system. Genes Cells. PubMed ID: 31124270

    Hakeda-Suzuki, S., Takechi, H., Kawamura, H. and Suzuki, T. (2017). Two receptor tyrosine phosphatases dictate the depth of axonal stabilizing layer in the visual system. Elife 6. PubMed ID: 29116043

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

    Organisti, C., Hein, I., Grunwald Kadow, I. C. and Suzuki, T. (2014). Flamingo, a seven-pass transmembrane cadherin, cooperates with Netrin/Frazzled in Drosophila midline guidance. Genes Cells. PubMed ID: 25440577

    Hakeda-Suzuki, S. and Suzuki, T. (2014). Cell surface control of the layer specific targeting in the Drosophila visual system. Genes Genet Syst 89: 9-15. PubMed ID: 24817757

    Berger-Muller, S., Sugie, A., Takahashi, F., Tavosanis, G., Hakeda-Suzuki, S. and Suzuki, T. (2013). Assessing the role of cell-surface molecules in central synaptogenesis in the Drosophila visual system. PLoS One 8: e83732. PubMed ID: 24386266

    Hakeda, S. and Suzuki, T. (2013). Golden goal controls dendrite elongation and branching of multidendritic arborization neurons in Drosophila. Genes Cells. PubMed ID: 23919529

  • Andrew Swan University of Windsor, Ontario
    Bourouh, M., Dhaliwal, R., Rana, K., Sinha, S., Guo, Z. and Swan, A. (2016). Distinct and overlapping requirements for Cyclins A, B and B3 in Drosophila female meiosis. G3 (Bethesda) [Epub ahead of print]. PubMed ID: 27652889

    Guo, Z., Batiha, O., Bourouh, M., Fifield, E. and Swan, A. (2015). Role of Securin, Separase and Cohesins in female meiosis, and polar body formation in Drosophila. J Cell Sci. PubMed ID: 26675236

    Batiha, O. and Swan, A. (2012). Evidence that the spindle assembly checkpoint does not regulate APC(Fzy) activity in Drosophila female meiosis. Genome 55: 63-67. PubMed ID: 22196012

    Ghorbani, M., Vasavan, B., Kraja, E. and Swan, A. (2011). Cks85A and Skp2 interact to maintain diploidy and promote growth in Drosophila. Dev Biol 358: 213-223. PubMed ID: 21827746

  • Douglas Swank Rensselaer Polytechnic Institute, Troy, NY
    Loya, A. K., Van Houten, S. K., Glasheen, B. M. and Swank, D. M. (2022). Shortening deactivation: quantifying a critical component of cyclical muscle contraction. Am J Physiol Cell Physiol 322(4): C653-c665. PubMed ID: 34965153

    Bell, K. M., Kronert, W. A., Huang, A., Bernstein, S. I. and Swank, D. M. (2019). The R249Q hypertrophic cardiomyopathy myosin mutation decreases contractility in Drosophila by impeding force production. J Physiol. PubMed ID: 30950055

    Glasheen, B. M., Ramanath, S., Patel, M., Sheppard, D., Puthawala, J. T., Riley, L. A. and Swank, D. M. (2018). Five Alternative Myosin converter domains influence muscle power, stretch activation, and kinetics. Biophys J 114(5): 1142-1152. PubMed ID: 29539400

    Wang, Q., Newhard, C., Ramanath, S., Sheppard, D. and Swank, D. M. (2013). An embryonic myosin converter domain influences Drosophila indirect flight muscle stretch activation, power generation and flight. J Exp Biol. PubMed ID: 24115062

    Zhao, C. and Swank, D. M. (2013). An embryonic Myosin isoform enables stretch activation and cyclical power in Drosophila jump muscle. Biophys J 104: 2662-2670. PubMed ID: 23790374

    Eldred, C. C., Naber, N., Pate, E., Cooke, R. and Swank, D. M. (2013). Conformational changes at the nucleotide site in the presence of bound ADP do not set the velocity of fast Drosophila myosins. J Muscle Res Cell Motil 34: 35-42. PubMed ID: 23203294

    Swank, D. M. (2012). Mechanical analysis of Drosophila indirect flight and jump muscles. Methods 56: 69-77. PubMed ID: 22079350

    Wang, Q., Zhao, C. and Swank, D. M. (2011). Calcium and stretch activation modulate power generation in Drosophila flight muscle. Biophys J 101: 2207-2213. PubMed ID: 22067160

  • Sean Sweeney Department of Biology, University of York, York, UK
    Ugbode, C., Garnham, N., Fort-Aznar, L., Evans, G. J. O., Chawla, S. and Sweeney, S. T. (2020). JNK signalling regulates antioxidant responses in neurons. Redox Biol 37: 101712. PubMed ID: 32949970

    West, R. J. H., Briggs, L., Perona Fjeldstad, M., Ribchester, R. R. and Sweeney, S. T. (2018). Sphingolipids regulate neuromuscular synapse structure and function in Drosophila. J Comp Neurol. PubMed ID: 29761896

    Hebbar, S., Khandelwal, A., R, J., Hindle, S. J., Chiang, Y. N., Yew, J. Y., Sweeney, S. T. and Schwudke, D. (2017). Lipid metabolic perturbation is an early-onset phenotype in adult spin mutants: a Drosophila model for lysosomal storage disorders. Mol Biol Cell. PubMed ID: 29046397

    Hindle, S. J., Hebbar, S., Schwudke, D., Elliott, C. J. and Sweeney, S. T. (2016). A saposin deficiency model in Drosophila: Lysosomal storage, progressive neurodegeneration and sensory physiological decline. Neurobiol Dis. PubMed ID: 27913291

    Cagin, U., Duncan, O. F., Gatt, A. P., Dionne, M. S., Sweeney, S. T. and Bateman, J. M. (2015). Mitochondrial retrograde signaling regulates neuronal function. Proc Natl Acad Sci U S A 112: E6000-6009. PubMed ID: 26489648

    Oswald, M. C., West, R. J., Lloyd-Evans, E. and Sweeney, S. T. (2015). Identification of dietary alanine toxicity and trafficking dysfunction in a Drosophila model of hereditary sensory and autonomic neuropathy type 1. Hum Mol Genet [Epub ahead of print]. PubMed ID: 26395456

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

    Fogg, P. C., O'Neill, J. S., Dobrzycki, T., Calvert, S., Lord, E. C., McIntosh, R. L., Elliott, C. J., Sweeney, S. T., Hastings, M. H. and Chawla, S. (2014). Class IIa histone deacetylases are conserved regulators of circadian function. J Biol Chem 289: 34341-34348. PubMed ID: 25271152

    Lu, Y., Zhang, Z., Sun, D., Sweeney, S. T. and Gao, F. B. (2013). Syntaxin 13, a genetic modifier of mutant CHMP2B in frontotemporal dementia, is required for autophagosome maturation. Mol Cell 52: 264-271. PubMed ID: 24095276

    Hindle, S., Afsari, F., Stark, M., Middleton, C. A., Evans, G. J., Sweeney, S. T. and Elliott, C. J. (2013). Dopaminergic expression of the Parkinsonian gene LRRK2-G2019S leads to non-autonomous visual neurodegeneration, accelerated by increased neural demands for energy. Hum Mol Genet 22: 2129-2140. PubMed ID: 23396536

  • Sheyum Syed Department of Physics, University of Miami
    Lazopulo, S., Lazopulo, A., Baker, J. D. and Syed, S. (2019). Daytime colour preference in Drosophila depends on the circadian clock and TRP channels. Nature 574(7776): 108-111. PubMed ID: 31534223

    Qiao, B., Li, C., Allen, V. W., Shirasu-Hiza, M. and Syed, S. (2018). Automated analysis of long-term grooming behavior in Drosophila using a k-nearest neighbors classifier. Elife 7. PubMed ID: 29485401

    Lazopulo, A. and Syed, S. (2016). A mathematical model provides mechanistic links to temporal patterns in Drosophila daily activity. BMC Neurosci 17: 14. PubMed ID: 27090880

    Lazopulo, S., Lopez, J. A., Levy, P. and Syed, S. (2015). A Stochastic Burst Follows the Periodic Morning Peak in Individual Drosophila Locomotion. PLoS One 10(11): e0140481. PubMed ID: 26528813

  • Aron Szabo Biological Research Centre, Institute of Genetics, University of Szeged, Hungary
    Bhattacharjee, A., Urmosi, A., Jipa, A., Kovacs, L., Deak, P., Szabo, A. and Juhasz, G. (2022). Loss of ubiquitinated protein autophagy is compensated by persistent cnc/NFE2L2/Nrf2 antioxidant responses. Autophagy: 1-12. PubMed ID: 35184662

    Abraham, A., Villanyi, Z., Zsindely, N., Nagy, G., Szabo, A., Bodai, L., Henn, L. and Boros, I. M. (2022). Despite its sequence identity with canonical H4, Drosophila H4r product is enriched at specific chromatin regions. Sci Rep 12(1): 5007. PubMed ID: 35322122

    Henn, L., Szabo, A., Imre, L., Roman, A., Abraham, A., Vedelek, B., Nanasi, P. and Boros, I. M. (2020). Alternative linker histone permits fast paced nuclear divisions in early Drosophila embryo. Nucleic Acids Res 48(16): 9007-9018. PubMed ID: 32710625

    Bhattacharjee, A., Szabo, A., Csizmadia, T., Laczko-Dobos, H. and Juhasz, G. (2019). Understanding the importance of autophagy in human diseases using Drosophila. J Genet Genomics 46(4): 157-169. PubMed ID: 31080044

  • Paul Szyszka Department of Zoology, University of Otago, New Zealand
    Sehdev, A., Mohammed, Y. G., Tafrali, C. and Szyszka, P. (2019). Social foraging extends associative odor-food memory expression in an automated learning assay for Drosophila melanogaster. J Exp Biol 222(Pt 19). PubMed ID: 31527181

    Sehdev, A., Mohammed, Y. G., Triphan, T. and Szyszka, P. (2019). Olfactory Object Recognition Based on Fine-Scale Stimulus Timing in Drosophila. iScience 13: 113-124. PubMed ID: 30826726

    Egea-Weiss, A., Renner, A., Kleineidam, C. J. and Szyszka, P. (2018). High Precision of Spike Timing across Olfactory Receptor Neurons Allows Rapid Odor Coding in Drosophila. iScience 4: 76-83. PubMed ID: 30240755

  • This listing of Fly Labs and References is a work in progress. Please contact Tom Brody ([email protected]) with your suggestions or corrections.

    Return to The Interactive Fly